07.01.2015 Views

full text - Caspar Bgsu - Bowling Green State University

full text - Caspar Bgsu - Bowling Green State University

full text - Caspar Bgsu - Bowling Green State University

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Aggression<br />

Volume 75


Advances<br />

in<br />

Genetics, Volume 75<br />

Serial Editors<br />

Theodore Friedmann<br />

<strong>University</strong> of California at San Diego, School of Medicine, USA<br />

Jay C. Dunlap<br />

Dartmouth Medical School, Hanover, NH, USA<br />

Stephen F. Goodwin<br />

<strong>University</strong> of Oxford, Oxford, UK


Volume 75<br />

Aggression<br />

Edited by<br />

Robert Huber<br />

JP Scott Center for Neuroscience<br />

Mind & Behavior, Biological Sciences<br />

<strong>Bowling</strong> <strong>Green</strong> <strong>State</strong> <strong>University</strong>, <strong>Bowling</strong> <strong>Green</strong>, OH, USA<br />

Danika L. Bannasch<br />

Department of Population Health and Reproduction<br />

School of Veterinary Medicine, <strong>University</strong> of California<br />

Davis, CA, USA<br />

Patricia Brennan<br />

Department of Psychology<br />

Emory <strong>University</strong>, Atlanta, GA, USA<br />

Editorial Assistant<br />

Kate Frishman<br />

<strong>Bowling</strong> <strong>Green</strong>, OH, USA<br />

AMSTERDAM • BOSTON • HEIDELBERG • LONDON<br />

NEW YORK • OXFORD • PARIS • SAN DIEGO<br />

SAN FRANCISCO • SINGAPORE • SYDNEY • TOKYO<br />

Academic Press is an imprint of Elsevier


Academic Press is an imprint of Elsevier<br />

525 B Street, Suite 1900, San Diego, CA 92101-4495, USA<br />

225 Wyman Street, Waltham, MA 02451, USA<br />

32 Jamestown Road, London, NW1 7BY, UK<br />

Radarweg 29, POBox 211, 1000 AE Amsterdam, The Netherlands<br />

First edition 2011<br />

Copyright ß 2011 Elsevier Inc. All rights reserved.<br />

No part of this publication may be reproduced, stored in a retrieval system<br />

or transmitted in any form or by any means electronic, mechanical, photocopying,<br />

recording or otherwise without the prior written permission of the publisher<br />

Permissions may be sought directly from Elsevier’s Science & Technology Rights<br />

Department in Oxford, UK: phone (+44) (0) 1865 843830; fax (+44) (0) 1865 853333;<br />

email: permissions@elsevier.com. Alternatively you can submit your request online by<br />

visiting the Elsevier web site at http://www.elsevier.com/locate/permissions, and selecting<br />

Obtaining permission to use Elsevier material.<br />

Notice<br />

No responsibility is assumed by the publisher for any injury and/or damage to persons or<br />

property as a matter of products liability, negligence or otherwise, or from any use or operation<br />

of any methods, products, instructions or ideas contained in the material herein. Because of<br />

rapid advances in the medical sciences, in particular, independent verification of diagnoses and<br />

drug dosages should be made.<br />

ISBN: 978-0-12-380858-5<br />

ISSN: 0065-2660<br />

For information on all Academic Press publications<br />

visit our website at elsevierdirect.com<br />

Printed and bound in USA<br />

11 12 13 10 9 8 7 6 5 4 3 2 1


Contents<br />

Contributors<br />

ix<br />

1 Aggression 1<br />

Robert Huber and Patricia A. Brennan<br />

2 Evolutionary Aspects of Aggression: The Importance<br />

of Sexual Selection 7<br />

Patrik Lindenfors and Birgitta S. Tullberg<br />

I. Introduction 8<br />

II. Sexual Selection 9<br />

III. Mating Systems 12<br />

IV. When to Fight and When to Flee 13<br />

V. Case Studies: Sexual Dimorphism 16<br />

VI. Humans and the Mammalian Pattern 20<br />

Acknowledgment 20<br />

References 21<br />

3 Signaling Aggression 23<br />

Moira J. van Staaden, William A. Searcy, and<br />

Roger T. Hanlon<br />

I. Introduction 24<br />

II. Bird Song Signals Aggressive Intentions: Speak Softly<br />

and Carry a Big Stick 31<br />

III. Visual Displays Signal Aggressive Intent in Cephalopods:<br />

The Sweet Smell of Success 37<br />

Acknowledgments 44<br />

References 44<br />

v


vi<br />

Contents<br />

4 Self-Structuring Properties of Dominance Hierarchies:<br />

A New Perspective 51<br />

Ivan D. Chase and Kristine Seitz<br />

I. Introduction 52<br />

II. Definitions 53<br />

III. Animal Models 55<br />

IV. Factors Affecting Dominance Relationships in Pairs<br />

of Animals 58<br />

V. Formation of Dominance Relationships and Dominance<br />

Hierarchies in Groups 63<br />

VI. A New Approach to Explaining the Formation of Linear<br />

Hierarchies: Behavioral Processes 70<br />

VII. Conclusion 74<br />

Acknowledgments 75<br />

References 75<br />

5 Neurogenomic Mechanisms of Aggression in<br />

Songbirds 83<br />

Donna L. Maney and James L. Goodson<br />

I. Aggression in Con<strong>text</strong> 84<br />

II. Hormonal Mechanisms of Aggression 87<br />

III. Transcriptional Activity and Neural Mechanisms<br />

of Aggression in Birds 95<br />

IV. A Natural Model Uniting Social Behavior, Hormones, and<br />

Genetics 103<br />

V. Future Directions 109<br />

Acknowledgments 110<br />

References 110<br />

6 Genetics of Aggression in Voles 121<br />

Kyle L. Gobrogge and Zuoxin W. Wang<br />

I. Introduction 122<br />

II. The Prairie Vole Model 122<br />

III. Neural Correlates 125<br />

IV. Neural Circuitry 127<br />

V. Neurochemical Regulation of Selective Aggression 128<br />

VI. Molecular Genetics of Selective Aggression 136<br />

VII. Drug-induced Aggression 136<br />

VIII. Conclusions and Future Directions 138


Contents<br />

vii<br />

Acknowledgments 140<br />

References 141<br />

7 The Neurochemistry of Human Aggression 151<br />

Rachel Yanowitch and Emil F. Coccaro<br />

I. Introduction 152<br />

II. Serotonin 152<br />

III. Dopamine 157<br />

IV. Norepinephrine (Noradrenaline) 159<br />

V. GABA 160<br />

VI. Peptides 162<br />

VII. Conclusion 162<br />

References 162<br />

8 Human Aggression Across the Lifespan: Genetic<br />

Propensities and Environmental Moderators 171<br />

Catherine Tuvblad and Laura A. Baker<br />

I. Heritability of Aggression: Twin and<br />

Adoption Studies 174<br />

II. G E Interaction in Aggressive Behavior 197<br />

III. Specific Genes for Aggressive Behavior: Findings<br />

from Molecular Genetic Studies 203<br />

IV. Conclusions 205<br />

References 207<br />

9 Perinatal Risk Factors in the Development of<br />

Aggression and Violence 215<br />

Jamie L. LaPrairie, Julia C. Schechter,<br />

Brittany A. Robinson, and Patricia A. Brennan<br />

I. Introduction 216<br />

II. The Neurobiological and Psychophysiological Systems<br />

Involved in the Regulation of Aggression<br />

and Violence 217<br />

III. Perinatal Factors Related to the Development<br />

of Aggression 227<br />

IV. Genetic Contributions 238<br />

V. Conclusions 242<br />

References 243


viii<br />

Contents<br />

10 Neurocriminology 255<br />

Benjamin R. Nordstrom, Yu Gao, Andrea L. Glenn,<br />

Melissa Peskin, Anna S. Rudo-Hutt, Robert A. Schug,<br />

Yaling Yang, and Adrian Raine<br />

I. Introduction 256<br />

II. Psychodynamic Theories 257<br />

III. Neuroimaging 258<br />

IV. Neuropsychological Testing 262<br />

V. Psychophysiological Evidence 263<br />

VI. Genetics 266<br />

VII. Nongenetic Risk Factors 269<br />

VIII. The Limitations and Potential of Neurocriminology 272<br />

IX. Modifiable Risk Factor Interventions 273<br />

X. Conclusion 274<br />

References 274<br />

Index 285


Contributors<br />

Numbers in parentheses indicate the pages on which the authors’ contributions begin.<br />

Laura A. Baker (171) <strong>University</strong> of Southern California, Los Angeles,<br />

California, USA<br />

Patricia A. Brennan (1, 215) Department of Psychology, Emory <strong>University</strong>,<br />

Atlanta, Georgia, USA<br />

Ivan D. Chase (51) Department of Sociology, Stony Brook <strong>University</strong>, Stony<br />

Brook, New York, USA<br />

Emil F. Coccaro (151) Clinical Neuroscience Research Unit, Department of<br />

Psychiatry, The <strong>University</strong> of Chicago Pritzker School of Medicine,<br />

Chicago, Illinois, USA<br />

Yu Gao (255) Department of Psychology, Brooklyn College, New York, USA<br />

Andrea L. Glenn (255) Department of Child and Adolescent Psychiatry,<br />

Institute of Mental Health, Singapore, Singapore<br />

Kyle L. Gobrogge (121) Department of Psychology and Program in<br />

Neuroscience, Florida <strong>State</strong> <strong>University</strong>, Tallahassee, Florida, USA<br />

James L. Goodson (83) Department of Biology, Indiana <strong>University</strong>,<br />

Bloomington, Indiana, USA<br />

Roger T. Hanlon (23) Marine Resources Center, Marine Biological Laboratory,<br />

Woods Hole, Massachusetts, USA<br />

Robert Huber (1) JP Scott Center for Neuroscience, Mind & Behavior,<br />

Biological Sciences, <strong>Bowling</strong> <strong>Green</strong> <strong>State</strong> <strong>University</strong>, <strong>Bowling</strong> <strong>Green</strong>,<br />

Ohio, USA<br />

Jamie L. LaPrairie (215) Department of Psychology, Emory <strong>University</strong>,<br />

Atlanta, Georgia, USA<br />

Patrik Lindenfors (7) Department of Zoology, and Centre for the Study of<br />

Cultural Evolution, Stockholm <strong>University</strong>, Stockholm, Sweden<br />

Donna L. Maney (83) Department of Psychology, Emory <strong>University</strong>, Atlanta,<br />

Georgia, USA<br />

Benjamin R. Nordstrom (255) Department of Psychiatry, <strong>University</strong> of<br />

Pennsylvania, Philadelphia, USA<br />

Melissa Peskin (255) Department of Psychology, <strong>University</strong> of Pennsylvania,<br />

Philadelphia, USA<br />

Adrian Raine (255) Departments of Psychiatry, Psychology and Criminology,<br />

<strong>University</strong> of Pennsylvania, Philadelphia, USA<br />

ix


x<br />

Contributors<br />

Brittany A. Robinson (215) Department of Psychology, Emory <strong>University</strong>,<br />

Atlanta, Georgia, USA<br />

Anna S. Rudo-Hutt (255) Department of Psychology, <strong>University</strong> of<br />

Pennsylvania, Philadelphia, USA<br />

Birgitta S. Tullberg (7) Department of Zoology, Stockholm <strong>University</strong>,<br />

Stockholm, Sweden<br />

Julia C. Schechter (215) Department of Psychology, Emory <strong>University</strong>,<br />

Atlanta, Georgia, USA<br />

Robert A. Schug (255) Department of Criminal Justice, California <strong>State</strong><br />

<strong>University</strong>, Long Branch, USA<br />

William A. Searcy (23) Department of Biology, <strong>University</strong> of Miami, Coral<br />

Gables, Florida, USA<br />

Kristine Seitz (51) Department of Biology, Stony Brook <strong>University</strong>, Stony<br />

Brook, New York, USA<br />

Catherine Tuvblad (171) <strong>University</strong> of Southern California, Los Angeles,<br />

California, USA<br />

Moira J. van Staaden (23) Department of Biological Sciences and JP Scott<br />

Center for Neuroscience, Mind & Behavior, <strong>Bowling</strong> <strong>Green</strong> <strong>State</strong><br />

<strong>University</strong>, <strong>Bowling</strong> <strong>Green</strong>, Ohio, USA<br />

Zuoxin W. Wang (121) Department of Psychology and Program in Neuroscience,<br />

Florida <strong>State</strong> <strong>University</strong>, Tallahassee, Florida, USA<br />

Yaling Yang (255) Laboratory of Neuro Imaging, <strong>University</strong> of California,<br />

Los Angeles, USA<br />

Rachel Yanowitch (151) Clinical Neuroscience Research Unit, Department of<br />

Psychiatry, The <strong>University</strong> of Chicago Pritzker School of Medicine,<br />

Chicago, Illinois, USA


1<br />

Aggression<br />

Robert Huber* and Patricia A. Brennan †<br />

*JP Scott Center for Neuroscience, Mind & Behavior, Biological Sciences,<br />

<strong>Bowling</strong> <strong>Green</strong> <strong>State</strong> <strong>University</strong>, <strong>Bowling</strong> <strong>Green</strong>, Ohio, USA<br />

† Department of Psychology, Emory <strong>University</strong>, Atlanta, Georgia, USA<br />

Aggression ranks among the most misunderstood concepts in all the behavioral<br />

sciences. It is commonly viewed by the general public as an aberrant form of<br />

behavior, with situations of conflict pictured as unfavorable and stressful circumstances,<br />

brought about by amoral urges, in critical need of our cognitive control,<br />

and with negative consequences for all involved. Such a view fundamentally<br />

misunderstands the biological significance of the behaviors that occur during<br />

conflict. Deeply rooted in the demands of the natural world, an individual must<br />

fulfill its demands for self-preservation, defend its interests, or compete for<br />

limited vital resources. Basic tendencies for aggression are virtually ubiquitous<br />

throughout the animal kingdom, regardless of its bearer’s neural or cognitive<br />

faculties, phylogenetic origins, or sociobiological circumstances. Just as widespread,<br />

however, are fundamental rules that govern physical conflict, such that<br />

cases of unbridled hostility are surprisingly rare. In most species, visual<br />

and elaborately ritualized displays effectively channel aggression, structure how<br />

individuals interact, and govern the conflict’s resolution.<br />

As we witness animals engaged in situations of conflict, we cannot help<br />

but be drawn in by the behavior’s inherent relevance to our own biological roots.<br />

The knowledge that human aggression arises from our genetic heritage makes<br />

it all the more likely that it is of an adaptive nature. As we study the<br />

individuals and environments where aggression is most commonly displayed,<br />

we gain a better understanding of when aggression and violence may serve<br />

an adaptive function and when it may not. Current research points to the<br />

importance of delineating subtypes of aggression, focusing on such concepts as<br />

proactive and reactive, direct and indirect, and adolescent limited versus life<br />

course persistent. Each of these types of aggression has a distinct etiology and<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00016-2


2 Huber and Brennan<br />

utility, depending upon the social environment in which the individual must<br />

function. For example, reactive aggression is aggression that occurs in response to<br />

a threat in the external environment. It is easy to see how this type of aggression<br />

may have its basis in our inherent survival instincts. Nevertheless, a propensity<br />

for reactive aggression may be truly dysfunctional in environments that pose low<br />

levels of threat, like many of those in which children in Western societies are<br />

now raised. A more comprehensive understanding of how biology, behavior, and<br />

environment intersect is paramount in the study of human violence and<br />

aggression.<br />

Studies of aggression, its motives, and its causes are of central interest to a<br />

wide range of academic disciplines—behavioral genetics, evolution, neuroscience,<br />

psychology, sociology, and criminology, to name just a few. Despite the<br />

wealth of empirical and theoretical attention, it is remarkable that a comprehensive<br />

synthesis of aggression has stubbornly remained elusive. This partly stems<br />

from the fact that the term “aggression” neither maps cleanly onto a monolithic<br />

behavioral phenomenon nor lends itself to representation by a simple explanatory<br />

concept. Another explanation for this paradox must reside in the absence of a<br />

unified, operational definition of aggression across disciplines, or even of a general<br />

agreement on what the term actually includes. For instance, most psychologists<br />

define aggression as “all behavior that is intended to cause bodily harm.” Other<br />

widely adopted classifications of aggression recognize subtypes, ranging from<br />

competition between males, a mother’s efforts to protect her offspring, or fighting<br />

as a learned response to cope with a particular situation. Biologists regard a<br />

definition focused solely on injury as insufficient as this excludes a wide range of<br />

threat behaviors directed at rivals, for example, birds that challenge their adversaries<br />

with song, an impala’s exaggerated strutting as a signal of strength, or a wolf’s<br />

territorial claims via scent markings. Moreover, there is little agreement on<br />

whether a predator’s hunting behavior should be included. A lion chasing and<br />

killing a gazelle undoubtedly inflicts injury, but is this more akin to a cow cropping<br />

the top off a clump of grass, or to an elephant bull inflicting serious injury to a rival<br />

in battle Moreover, behavior in aggressive encounters always balances contrasting<br />

impulses for approach and attack with a tendency to flee—rarely is either<br />

present entirely alone. To acknowledge the difficulty of disentangling these<br />

components, the term “agonistic behavior” has been introduced. The term specifically<br />

addresses the balance of forces for both attacking and fleeing and it accommodates<br />

all instances of attack and threat (i.e., offensive agonistic behavior) as<br />

well as escape and submission (i.e., defensive agonistic behavior).<br />

As with any other characteristic, natural selection is assumed to<br />

enhance aggression’s overall effectiveness. High ranking individuals are likely<br />

to display a favorable combination of strength, along with an ability to titer<br />

their levels of aggression, to pick fights that are winnable, and to only compete<br />

in those that are worth it. Hyperaggressive phenotypes exist in most systems.


1. Aggression 3<br />

These exhibit fighting that greatly exceeds the most effective norm, they<br />

readily launch the initial attack even in situations where they ought not to,<br />

are overly eager to escalate or retaliate, show a willingness to follow an<br />

excessively physical trajectory even when an opponent has already withdrawn,<br />

or fail to back down in situations where there is little prospect of winning.<br />

Such behaviors rarely make for an effective strategy, as they coincide with<br />

greater risk of injury or death, or in the best-case scenario, attaining a<br />

low rank.<br />

A thorough analysis of aggression minimally demands that we (1)<br />

capture the essence of an inherently multifaceted phenotype, (2) address underlying<br />

elements of motivation that are not always readily observed or elicited,<br />

(3) understand the various scenarios and con<strong>text</strong>s that influence its expression,<br />

(4) decipher the neural, hormonal, and genetic causes that are at work, and (5)<br />

explore how its components are shaped by evolution. The chapters in this<br />

volume aim to provide a comprehensive overview of these topics as their authors<br />

unravel the individual behavioral and neural strands constituting situations of<br />

conflict.<br />

Initial chapters of the volume characterize the elemental building<br />

blocks of aggression; they assess, precisely delineate, and account for aggression’s<br />

different and unique components, and explain how intricate behavioral constructs<br />

often emerge from much simpler roots. The initial chapters review<br />

aggression from a predominantly evolutionary perspective. Conflicts are energetically<br />

costly and carry inherent risks. Natural selection offers a powerful conceptual<br />

tool as it focuses on an individual’s behavioral strategies and decision<br />

making in ways that maximize its fitness. Evolution can only exert its influence<br />

on characters that depend, at least partially, on genetic underpinnings. Lindenfors<br />

and Tullberg (Chapter 2) discuss the significance of sexual selection as a key<br />

evolutionary structuring force in aggression. In most scenarios, ritualized displays<br />

take the place of unchecked, aggressive interactions. Game theory offers a<br />

powerful framework for why animals only tend to fight with great ferocity<br />

when a resource of exceptional value is at stake. Resources are rarely worth the<br />

risk of sustaining injury, and competing individuals will do best by resolving<br />

conflicts with ritualized displays only. Skill in assessing the relative strength of an<br />

opponent is key for navigating the demands, risks, and opportunities of social<br />

living. The review by van Staaden and colleagues (Chapter 3) discusses a<br />

prominent role for signaling aggressive behaviors, which permit individuals to<br />

obtain valid estimates of an opponent’s true strength. Once an animal is bested<br />

by an opponent, it is always better to adopt submissive behavior and accept<br />

subordinate status, rather than risk something far worse. A wide range of attributes<br />

decides between victory and defeat. With prominent asymmetries in the<br />

size of weapons, strength, or agility, fights are often quickly resolved. In many<br />

instances, though, social success will depend also on an ability to form successful


4 Huber and Brennan<br />

alliances, to harness cognitive skills, or to inherit status from high-ranking kin. A<br />

paired dominance relationship is established when prior encounters produce a<br />

lasting polarity in the outcome of future bouts. In its most common form, the past<br />

loser will be less likely to initiate further bouts against the winner or will retreat<br />

quickly if confronted. As individuals repeatedly meet and interact with others,<br />

higher order social organization emerges through a series of sequential dyadic<br />

interactions. Individuals of many species, including humans, tend to arrange<br />

themselves in largely linear social hierarchies. Although individual characteristics<br />

such as size, strength, or agility are relatively fixed and may indeed<br />

influence rank, Chase and Seitz (Chapter 4) illustrate that these qualities are<br />

more often overshadowed by con<strong>text</strong>ual factors and chance events.<br />

The search for proximate mechanisms underlying aggression requires us<br />

to view aggression’s natural building blocks, to recognize the various factors that<br />

control them, and to effectively label their behavioral expression in the form of<br />

consistent and reliable phenotypes. Our understanding of the biological basis of<br />

aggression in all vertebrates, including humans, has been built largely upon<br />

discoveries first made in birds. An extensive literature indicates that hormonal<br />

mechanisms are shared between humans and many avian species. This recent<br />

development of hormonal, neuroendocrine, and genetic tools has established<br />

songbirds as powerful models for understanding the neural basis and evolution of<br />

vertebrate aggression. Maney and Goodson (Chapter 5) discuss the contributions<br />

of field endocrinology toward a theoretical framework linking aggression with sex<br />

steroids, explore evidence that the neural substrates of aggression are conserved<br />

across vertebrate species, and describe a promising new songbird model for<br />

studying the molecular genetic mechanisms underlying aggression. Voles have<br />

recently emerged as a key model for a genetic dissection of social behavior and its<br />

underlying neural mechanisms. Gobrogge and Wang (Chapter 6) discuss its<br />

utility for the study of aggression and review recent findings that illustrate the<br />

neurochemical mechanisms underlying pair bonding-induced aggression. Endogenous<br />

brain chemicals play a key role in the control of aggression in many taxa<br />

including humans. Neurotransmitters effectively pattern and modulate the expression<br />

of basic behavioral components. Genetic abnormalities in a number of<br />

neurotransmitter pathways have been implicated in aggression-related disorders.<br />

Yanowitch and Coccaro (Chapter 7) review work that demonstrates that neurotransmitter<br />

function is intricately linked to aggressive state.<br />

Subsequent chapters focus on human aggression, with an emphasis on<br />

genetic and other biological factors. Human ingenuity for inflicting intentional<br />

harm is without equal, although warring tendencies may already be rooted in a<br />

deep, prehuman past. Instances of violence have been documented for a range of<br />

nonhuman apes and may have arguably wired into our genes when our more<br />

aggressive ancestors won against our less aggressive ancestors in terms of survival<br />

and reproduction. Aside from an unprecedented potential for carnage and


1. Aggression 5<br />

destruction, humans are at the same time also capable of the most remarkable<br />

instances of compassion, understanding, and peaceful negotiation. The direction<br />

depends on each individual’s ethical codes and moral norms driven by the<br />

societal expectations, good parenting, or social con<strong>text</strong>s. A clear vision has<br />

emerged where “natural” tendencies for aggression appear to be ubiquitous, but<br />

so too are a plethora of sophisticated mechanisms that keep conflicts in check,<br />

channel aggression, negotiate fighting signals, resolve conflicts, and ultimately<br />

govern social group structure.<br />

Tuvblad and Baker (Chapter 8) demonstrate that genetic influences<br />

serve as powerful predictors of human aggression and violence. The relative<br />

influence of genetics depends upon developmental age, type of aggression, and<br />

the environmental con<strong>text</strong> faced by the individual. LaPrairie and colleagues<br />

(Chapter 9) review research linking perinatal factors and aggression and conclude<br />

in a similar fashion that perinatal and neurodevelopmental factors<br />

influence the expression of aggressive behaviors. Nordstrom and colleagues<br />

(Chapter 10) further detail the importance of recognizing the role of brain<br />

functioning deficits in the risk for aggressive outcomes. Recent advances in<br />

imaging technology have enabled a far greater understanding of these influences<br />

on human behavior and the risks of criminal outcomes. Importantly, the chapters<br />

on human aggression also emphasize the fact that biology is not destiny and that,<br />

in the case of human aggression and violence, there is much that can and should<br />

be done in terms of early intervention and prevention.<br />

The list of significant challenges in aggression research remains daunting<br />

and the need to harness the <strong>full</strong> power of interdisciplinary approaches now<br />

appears more urgent than ever. Aside from our need to reconcile simple questions<br />

over terminology, a number of more serious impediments remain to be<br />

acknowledged. Concepts seem so intimately connected that we are tempted to<br />

view them as essentially overlapping, or to even use them synonymously (e.g.,<br />

measures of an inherent tendency to fight, effectiveness in a contest, or the<br />

ability to socially dominate others). A common fallacy views these simply as<br />

separate perspectives onto the same, unitary phenomenon of aggression. For a<br />

synthesis to emerge we must accept aggression’s multidimensional nature and<br />

recognize that the term “aggression” simply serves as an overarching label for an<br />

entangled complex of multiple, distinct components, causes, and functions. This<br />

volume comes at a critical juncture for defining a broader view of aggression and<br />

with it we hope to help define the structural elements that comprise the behavior<br />

in its <strong>full</strong> complexity.<br />

The time is now right to bridge theoretical frameworks, combine<br />

experimental approaches, and relate significant findings across the many individual<br />

disciplines that are instrumental in the analysis of aggression. Center<br />

initiatives can serve as intellectual hubs for the comprehensive study of social<br />

conflict, violence, and related phenomena. Bringing together individual


6 Huber and Brennan<br />

researchers from a broad range of disciplines to foster integrative and overarching<br />

themes, such centers provide a forum for a rich exchange of ideas, the<br />

development of human resources, a clearinghouse for notable discoveries, and<br />

to publicize their societal relevance through public outreach. The development<br />

of viewpoints spanning formerly separate disciplines, such as the one aimed for<br />

in this book, is cause for optimism that the future is not quite as far off as we<br />

had feared.


2<br />

Evolutionary Aspects of<br />

Aggression: The Importance of<br />

Sexual Selection<br />

Patrik Lindenfors* ,† and Birgitta S. Tullberg*<br />

*Department of Zoology, Stockholm <strong>University</strong>, Stockholm, Sweden<br />

† Centre for the Study of Cultural Evolution, Stockholm <strong>University</strong>, Stockholm,<br />

Sweden<br />

I. Introduction<br />

II. Sexual Selection<br />

III. Mating Systems<br />

IV. When to Fight and When to Flee<br />

V. Case Studies: Sexual Dimorphism<br />

VI. Humans and the Mammalian Pattern<br />

Acknowledgment<br />

References<br />

ABSTRACT<br />

Aggressive behaviors in animals, for example, threat, attack, and defense, are<br />

commonly related to competition over resources, competition over mating<br />

opportunities, or fights for survival. In this chapter, we focus on aggressive<br />

competition over mating opportunities, since this competition explains much<br />

of the distribution of weaponry and large body size, but also because this type of<br />

competition sheds light on the sex skew in the use of violence in mammals,<br />

including humans. Darwin (1871) termed this type of natural selection, where<br />

differences in reproductive success are caused by competition over mates, sexual<br />

selection. Not all species have a pronounced competition over mates, however.<br />

Instead, this aspect of sociality is ultimately determined by ecological factors.<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00009-5


8 Lindenfors and Tullberg<br />

In species where competition over mates is rampant, this has evolutionary effects<br />

on weaponry and body size such that males commonly bear more vicious weapons<br />

and are larger than females. A review of sexual selection in mammals reveals how<br />

common aggressive competition over mating opportunities is in this group.<br />

Nearly half of all mammal species exhibit male-biased sexual size dimorphism,<br />

a pattern that is clearly linked to sexual selection. Sexual selection is also<br />

common in primates, where it has left clear historical imprints in body mass<br />

differences, in weaponry differences (canines), and also in brain structure differences.<br />

However, when comparing humans to our closest living primate relatives,<br />

it is clear that the degree of male sexual competition has decreased in the<br />

hominid lineage. Nevertheless, our species displays dimorphism, polygyny, and<br />

sex-specific use of violence typical of a sexually selected mammal. Understanding<br />

the biological background of aggressive behaviors is fundamental to understanding<br />

human aggression. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

Why does aggression exist in nature Darwin (1859, 1871) pointed out that the<br />

ultimate explanation for any trait has to be found in the effect that it has on<br />

survival and reproduction. From an evolutionary standpoint, individuals should<br />

thus mainly be expected to fight over resources, for survival and for mating<br />

opportunities, because these are what mainly affect how many genes that individual<br />

contributes to the gene pool of the coming generation. Another prediction<br />

is that the amount of aggression displayed in the encounters should increase<br />

with increasing value of the fought-over resource. Aggressive behaviors are<br />

associated with costs, and individuals are simply expected to take higher risks,<br />

that is, pay potentially higher costs, with increasing potential gains. In this<br />

chapter, we focus on aggression over mating opportunities—sexual selection—<br />

in mammals in general and in primates in particular. We focus on sexual selection<br />

because evidence suggests that it is the primary reason why animals fight with<br />

conspecifics and because it is the most likely explanation of some aspects of<br />

human aggression, such as why males tend to be more aggressive than females.<br />

An important point about evolutionary explanations is the philosophical<br />

distinction between proximate and ultimate explanations. Take sex, for<br />

example. Do humans have sex because it feels good or in order to have children<br />

Most sexual intercourse in current society probably has very little to do with<br />

actual procreation; on the contrary, there are many birth control methods<br />

available to make it possible to have sex without this resulting in a pregnancy.<br />

Despite the fact that protected sex happens “because” it feels good, the evolutionary<br />

explanation of sexual intercourse is “because” of procreation. This is<br />

where the crucial distinction between proximate and ultimate explanations


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 9<br />

comes into play. A proximate explanation is the explanation that is closest to the<br />

event that is to be explained. The higher, ultimate explanation is instead the<br />

deeper reason for why something happened.<br />

In biology, the division in ultimate and proximate explanations has<br />

been extended to what is usually termed “Tinbergen’s four questions”<br />

(Tinbergen, 1963); the four potential explanations of any behavior: (1) survival<br />

value or adaptive function, (2) phylogenetic history, (3) individual development,<br />

and (4) causal mechanisms such as hormonal mediation of behavior. The<br />

first two of Tinbergen’s questions are ultimate whereas the latter two are proximate.<br />

To <strong>full</strong>y shed light on a biological phenomenon all four types of questions<br />

are needed and the answers complement each other. However, in this chapter,<br />

we focus entirely on ultimate, evolutionary answers to the question of why<br />

aggression exists and takes the form it does.<br />

II. SEXUAL SELECTION<br />

Natural selection is all about who gets to reproduce and who does not (Darwin,<br />

1859). A central aspect of getting to reproduce is to survive until the opportunity to<br />

reproduce arises and to gain access to resources enabling you to do so, but another<br />

important aspect concerns direct competition in connection with the reproductive<br />

act itself. Darwin termed this second aspect “sexual selection”: differences in<br />

reproductive success caused by competition over mates (Darwin, 1871).<br />

Why did Darwin give a specific name to one part of natural selection:<br />

why not just stick to the umbrella term “natural selection” Darwin had noted<br />

that there often seems to be a conflict of interest between traits that increase<br />

survival and traits that increase reproduction; many traits that give an advantage<br />

in reproduction have negative consequences for survival. A male peacock’s large<br />

tail feathers are a prime example of such a trait. How can such a long colorful tail<br />

evolve when it makes the bearer simultaneously more visible and less adept at<br />

escaping predators When thinking about this problem before having formulated<br />

the theory of sexual selection, Darwin wrote in a letter to his friend, the botanist<br />

Asa Gray, the famous line: “The sight of a feather in a peacock’s tail, whenever<br />

I gaze at it, makes me sick!” (Darwin, 1860).<br />

To clarify this second aspect of natural selection—selection that has to<br />

do with competition over mates—Darwin wrote a follow-up to “On the Origin of<br />

Species” (1859), “The Descent of Man and Selection in Relation to Sex” (1871).<br />

In this book, Darwin points out that there are two potential kinds of competition<br />

over mates, two forms of sexual selection. Either individuals of one sex (usually<br />

males) can fight with each other over mating opportunities (intrasexual selection)<br />

or, alternatively, individuals of one sex (usually females) can choose<br />

individuals of the other sex on the basis of some trait (intersexual selection).


10 Lindenfors and Tullberg<br />

This second form of sexual selection is the explanation of the peacock’s tail:<br />

peahens simply find it attractive and prefer to mate with the peacocks with the<br />

most elaborate tail. Further, the tail provides information on the genetic quality<br />

of the male—it is an honest signal (Petrie, 1994).<br />

It is noteworthy that it was the idea of intersexual selection that caused<br />

the most furious debate in Darwin’s time, foremost because it was judged utterly<br />

questionable that female aesthetical judgment could be the ultimate explanation<br />

for so many conspicuous characters in nature. However, partner choice is a more<br />

peaceful process than direct competition within a sex. Thus, because it induces<br />

so much aggression in nature, we focus in this chapter mainly on intrasexual<br />

selection; physical competition over mating opportunities. It should be pointed<br />

out that the two forms of sexual selection sometimes occur simultaneously, for<br />

instance, in lekking species where females choose as mating partners the winning<br />

males from physical competition (Andersson, 1994).<br />

Sexual selection arises when one sex limits the reproductive success of<br />

the other. Most often it is females who are the limiting resource for the reproductive<br />

success of males due to a fundamental asymmetry between males and<br />

females in their defining characteristic, their gametes. Males are designated by<br />

their smaller, mobile gametes, called sperm cells. Females are designated by their<br />

larger, nutrition-carrying gametes, called eggs. Males can make more gametes<br />

than females, simply because sperm are energetically cheaper to make than eggs;<br />

thus, there is a fundamental reproductive difference between males and females.<br />

This initial asymmetry has consequences. Making sperm is cheap and easy, so<br />

this is not what limits the reproductive possibilities of males. Making eggs, on the<br />

other hand, is much costlier. Thus, sexual selection commonly—but not exclusively—affects<br />

males, because given an equal sex ratio, male reproductive success<br />

is limited by access to matings with females. Conversely, female reproductive<br />

success is limited by the number of eggs she can produce (Andersson, 1994). This<br />

sex specificity is so common that the reverse pattern, termed sex role reversal, is<br />

subject to intense interest from evolutionary biologists when it occurs (e.g., Ralls,<br />

1976; Vincent et al., 1992).<br />

An important experimental verification of this theoretical insight was<br />

made by the geneticist Bateman (1948), who experimented on fruit flies. Bateman<br />

noted a pattern demonstrating that the number of offspring a male fruit fly<br />

can have is directly correlated with his number of matings. The same does not<br />

hold true for females, who have roughly the same number of offspring no matter<br />

how many times they mate (as long as it is at least once). This pattern is termed<br />

Bateman’s principle. Later studies, however, have documented that a number of<br />

exceptions to Bateman’s principle exist in nature (Birkhead, 2001). Individuals<br />

are not only sperm and eggs; there are a number of additional factors that need to<br />

be incorporated to understand what is going on in different species. In mammals,<br />

especially, one needs to incorporate two unique adaptations. While the energy


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 11<br />

investments in the mammal zygotes differ only marginally in relation to the body<br />

mass of most mammals, the cost to mammal females greatly exceeds that to males<br />

due to effects of pregnancy and lactation. This energy investment inequality has<br />

existed since the origin of the class Mammalia, 125 million years ago.<br />

There are some exceptions to the general mammalian pattern, however.<br />

For instance, some mammal babies are so expensive to bring up to maturation that<br />

both sexes have to partake in the upbringing for it to be possible. In these species,<br />

where males and females work together to guard and rear the young, intrasexual<br />

competition occurs just prior to pair formation. In these species, the two sexes are<br />

usually morphologically alike. In other mammal species, however, competition<br />

between males over mating opportunities is fierce. In some species this affects the<br />

entire social life of the species, in that males physically exclude other males from<br />

the group. The result is a social system akin to a harem structure, with immature<br />

males roaming outside the social gathering or forming bachelor groups.<br />

The importance of sexual selection in understanding aggression in<br />

mammals is most clearly illustrated by the presence and absence of weaponry.<br />

For example, male ungulates are commonly equipped with horns while females<br />

are not. Horns would be a good weapon to fend off predators, especially when you<br />

need to defend your young, or to fight off conspecific competitors. But most<br />

young are cared for by single mothers; the fathers—who have the weapons—are<br />

absent. Ungulate horns are commonly ready just in time for rutting season and<br />

are then shed (e.g., deer). Instead of predator defense, male ungulates mainly use<br />

their horns to fight each other (Caro et al., 2003; Stankowich and Caro, 2009).<br />

A similar case can be made for the large, sharp canines of primates (Thorén et al.,<br />

2006), and large body size in male mammals in general (Lindenfors et al., 2007a).<br />

Such sex-skewed distribution of size and weaponry, in combination with observations<br />

of fierce aggression, is what enables us to assert that most serious conflict and<br />

aggression in mammals is over mating opportunities.<br />

Sexual selection acting primarily on one sex may have indirect but<br />

pronounced consequences for the relationship between the sexes. Thus, direct<br />

conflicts between males sometimes result in conflicts of interest between males<br />

and females. Early thoughts on this issue (Parker, 1979, Trivers, 1972; Williams,<br />

1966) have received much empirical support (Arnqvist and Rowe, 2005), and it<br />

now almost seems the norm rather than an exception that there exists such a<br />

conflict and that this becomes more severe under strong intrasexual competition.<br />

This can lead to the interesting phenomena of sexually antagonistic coevolution<br />

where males and females become involved in an arms race, as traits in one sex<br />

entice the evolution of resistance in the other (Holland and Rice, 1998;<br />

Gavrilets et al., 2001; and others). On the other hand, intrasexual competition<br />

can lead to one sex dominating the other. With regard to aggression and physical<br />

prowess, the common situation in mammals, including primates (Hrdy, 1981), is<br />

that males are physically dominant over females.


12 Lindenfors and Tullberg<br />

III. MATING SYSTEMS<br />

Not all animals have clear sexual differences (Fairbairn et al., 2007). In birds, for<br />

example, many species of gulls and penguins are so alike that it is impossible to<br />

determine the sex except by closer inspections of the genitals. At the other<br />

extreme are mallards, where the sexes are so different that Linnaeus classified<br />

them as two different species (Andersson, 1994). In mammals, we find the same<br />

variation even within a given mammalian order. Thus, within Pinnipedia we<br />

have, on the one hand, elephant seals where males may weigh up to five times as<br />

much as females and on the other hand, species such as Baikal seals where<br />

females are of similar weight as males (Lindenfors et al., 2002). These differences<br />

in dimorphism are due to differences in the degree of sexual selection. But why<br />

are there differences in the degree of sexual selection between species to start<br />

with<br />

Fundamentally, this question is about factors affecting male and female<br />

social group size. These issues are commonly addressed by focusing on the<br />

ecological variables that determine the spatiotemporal distribution of females,<br />

based on the expectation that resources and predation account for variation in<br />

female reproductive success. By comparison, access to females is generally assumed<br />

to be the major factor influencing male reproductive success (Emlen and<br />

Oring, 1977; Trivers, 1972; Wilson, 1975). After risks and resources have<br />

determined the spatiotemporal distribution of females, the distribution of<br />

females is in turn expected to influence the degree of male intrasexual competition<br />

(Emlen and Oring, 1977). For instance, a group of concurrently fertile<br />

females opens the field for male competition and monopolization. The general<br />

framework is therefore that social evolution is driven by females.<br />

The theoretical expectation that social evolution is ultimately driven by<br />

female distribution is empirically supported by comparative studies on primates, a<br />

group for which there is a significant correlation between evolution of male and<br />

female sociality (e.g., Altmann, 1990; Mitani et al., 1996; Nunn, 1999). Further, a<br />

phylogenetic investigation has shown that the evolution of female group size<br />

precedes the evolution of male group size, that is, that evolutionary changes in<br />

male group size lag changes in female group size (Lindenfors et al., 2004).<br />

Ecological factors determine whether it is possible for a male to monopolize<br />

several mating opportunities. For example, elephant seal females give birth<br />

on beaches. With a limited number of suitable beaches available in the elephant<br />

seal range, females tend to crowd together when giving birth. Elephant seals<br />

mate soon after they have given birth, so at the time of mating females are<br />

gathered tightly on limited stretches of beach. Males can exclude other males<br />

from a stretch of beach and thereby secure matings with a large number of<br />

females. Successful males in this competition gain all matings, while the losers<br />

get none. Fighting among elephant seals over mating opportunities is thus a


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 13<br />

fierce and bloody affair, a scenario which has resulted in extreme size dimorphism.<br />

In other pinniped species, females give birth in isolated caves on the polar<br />

ice pack; thus, no opportunity exists to monopolize matings. Without evolutionary<br />

pressure for male fighting ability, the sexes are more equal in size (Lindenfors<br />

et al., 2002).<br />

In conclusion, the ultimate cause for differences in mating systems can<br />

be traced back to ecological circumstances. The differences in mating systems in<br />

turn trigger differences in aggressive competition for mating opportunities which<br />

is what drives the evolution of sex differences in size and weaponry. These<br />

morphological sex differences are clear indicators of the severity of male–male<br />

aggression.<br />

IV. WHEN TO FIGHT AND WHEN TO FLEE<br />

Given that fighting is most often about mating opportunities, how are they<br />

predicted to pan out in terms of ferocity, number of behaviors involved, length<br />

of time, and so on There are two things that determine the ferocity of fighting:<br />

the value of the object being fought over and the risks involved. The problem<br />

can be reduced to a cost-benefit analysis. A male can not give up at first instance<br />

to maximize his chances of survival, because that would result in total nonreproduction.<br />

Neither can he go “all-in” in just any aggressive encounter if there<br />

exists only a minute chance of success. Instead, males in competitive situations<br />

have to weigh the probabilities of success, injury, and survival against each other,<br />

while considering other factors such as energy expenditures and probabilities of<br />

success in future interactions with other competitors. It is important to note that<br />

animals make calculations and decisions about how to act, but such processes do<br />

not necessarily require the consciousness about the process usually ascribed to<br />

human decision-making. Rather, animals are believed to use cues with regard<br />

to the environment, as well as their own and the opponent’s current status, and<br />

to use this information in an unconscious way when making decisions.<br />

One consequence this accounting has had over evolutionary history is<br />

that competitive interactions often take the form of a “sequential assessment<br />

game”. Simply put, this prescribes that each competitor should attempt to assess<br />

his opponent’s strength using as little energy as possible. Escalation should only<br />

be initiated by the competitor that feels he has the upper hand, or by either<br />

opponent if they cannot determine who is superior (Enquist and Leimar, 1983).<br />

Thus, a meeting between two deer males often starts out with a stage of roaring,<br />

which acts as a forcedly honest signal of body size. If this does not settle who is<br />

the larger/stronger, it is followed by “parallel walking,” where each competitor<br />

tries to judge the size and strength of the other by walking back and forth in<br />

parallel. Only if it is still unclear who is the larger or stronger will the


14 Lindenfors and Tullberg<br />

Fight (8)<br />

Approach<br />

(50)<br />

Roar<br />

contest<br />

(33)<br />

No roar<br />

contest<br />

(17)<br />

Parallel<br />

walk<br />

(17)<br />

One stag<br />

withdraws<br />

(16)<br />

Parallel<br />

walk<br />

(7)<br />

One stag<br />

withdraws<br />

(9)<br />

Fight (5)<br />

One stag<br />

withdraws<br />

(2)<br />

No<br />

parallel<br />

walk<br />

(10)<br />

Fight (1)<br />

One stag<br />

withdraws<br />

(9)<br />

Figure 2.1. Sequential assessment in red deer (from Clutton-Brock and Albon, 1979).<br />

competition escalate to actual fighting (Clutton-Brock and Albon, 1979;<br />

Fig. 2.1). Thanks to this “game,” really fierce fights only happen between<br />

opponents of equal size—inferior competitors flee quickly to fight another day<br />

(and another opponent).<br />

There is a common interest among fighters in trying to expend as little<br />

energy as possible while simultaneously minimizing the possibility of injury. For<br />

example, wolves and other canids ritualistically greet each other several times<br />

each day and display dominance or submission on a regular basis—they do not<br />

determine their relationship at every encounter. Some cichlid fish have a system<br />

akin to that of red deer, with different stages of escalation (Brick, 1999). Male<br />

lions fight savagely only if they stand a good chance of winning a pride of females.<br />

Research shows that they determine the quality of their rivals on cues from each<br />

other’s manes (West and Packer, 2002). Lekking birds such as black grouse have<br />

distinctive courtship rituals where they make calls and visual displays, an odd<br />

mix of strength comparison and showing off, where females can pick winners<br />

according to some criterion, sometimes just by copying other females’ choices<br />

(Andersson, 1994; Dugatkin and Godin, 1993; Wade and Pruett-Jones, 1990).<br />

Seldom do fights turn into vicious fighting, and when they do it is usually because<br />

either the contestants are judged by each other to be of equal strength, or because


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 15<br />

the benefit of winning—the value of the contested item—is much larger than the<br />

cost of losing. If the choice is reproduction or death, fights become deadly. This is<br />

why fights among elephant seals are so fierce and bloody. The chance to mate<br />

occurs only once per year and most males never even get close. For the successful<br />

males it is another story—in a study of Southern elephant seals, harem holders<br />

accounted for 89.6% of the recorded paternities (Fabiani et al., 2004; Fig. 2.2).<br />

The sequential assessment game is a variant of a game theoretical setup<br />

termed the “hawk-dove game” (see also Chapter 3). In this game, there are two<br />

possible strategies: always fight (“hawk”) and always yield (“dove”), where it is<br />

assumed that the two competitors have equal fighting ability. An Evolutionarily<br />

Number of males Number of males<br />

12<br />

8<br />

4<br />

0<br />

12<br />

8<br />

4<br />

RUB96<br />

SF96<br />

SF97<br />

0 1 2 7 0 1 12 1 24<br />

SI196 SI296 SI297<br />

0<br />

0 1 24 0 1 2 8 21 0<br />

12<br />

1 2 8 32<br />

Number of males<br />

8<br />

4<br />

SM96<br />

HH<br />

NHH<br />

0<br />

0 2 12<br />

Number of paternities<br />

Figure 2.2. Number of paternities achieved by the harem holder (HH) and the other males (NHM)<br />

associated with each harem in seven different populations of Southern elephant seals<br />

(from Fabiani et al., 2004).


16 Lindenfors and Tullberg<br />

Stable Strategy is a strategy which, if adopted by a population of players, cannot<br />

be invaded by any alternative strategy. It has been shown that the ESS is a mix of<br />

hawks and doves with proportions determined by the cost of fighting in relation<br />

to the benefit of winning (Maynard Smith, 1982). This game provides theoretical<br />

information that in a population of nonfighters it is profitable to be a fighter,<br />

and vice versa. If one extends the game to include a strategy called “assessor” that<br />

determines whether it will act as a “hawk” or a “dove” based on some criterion—<br />

for example, depending on priority at the resource—one can arrive at the<br />

sequential assessment game. The assessment strategy is also an ESS (Maynard<br />

Smith, 1982).<br />

The prediction from these game theoretical models is that populations<br />

where individuals compete over resources or matings should consist of individuals<br />

utilizing different strategies depending on situation, where important<br />

factors are the current size and physical state of self and opponents and the<br />

value of resources (for instance, the number of females in the group being fought<br />

over). In this con<strong>text</strong>, it should be noted that some animal populations have<br />

evolved alternatives to fighting strategies, usually known as sneaker strategies.<br />

Such males are usually much smaller than fighting males and can covertly sneak<br />

matings from females while the fighters are occupied with physical combat<br />

(Gross, 1996).<br />

V. CASE STUDIES: SEXUAL DIMORPHISM<br />

As mentioned above, animal groups differ in both the way and the degree to<br />

which they are exposed to sexual selection, and this will have great effects on the<br />

evolution of sex differences (Fairbairn et al., 2007). Although mammals as a<br />

group are characterized by a high degree of intrasexual selection (as compared<br />

with, for instance, birds, where intersexual selection seems to be more common),<br />

there is variation in the strength of sexual selection both within and among<br />

mammalian orders. An example of this variation is the pinnipeds (seals, sea lions,<br />

and walruses) where there exists a clear relationship between harem size and<br />

sexual size dimorphism (Lindenfors et al., 2002; Fig. 2.3).<br />

In this section, we bring up some studies that have compared different<br />

mammalian groups with respect to the consequences of sexual selection on<br />

behavioral and morphological evolution. There are 4629 extant or recently<br />

extinct mammalian species, as listed by Wilson and Reeder (1993). In a survey<br />

of 1370 of these, Lindenfors et al. (2007a) showed that sexual selection is a<br />

prevalent selective force in mammals. With a cutoff point at a 10% size difference<br />

in either direction to “count” as sexual dimorphism, mammals were, on<br />

average, male-biased size dimorphic (average male/female mass ratio ¼ 1.184;<br />

paired t-test p0.001; Table 2.1) with males being larger than females in 45% of


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 17<br />

0.8<br />

log(male/female weight) contrasts<br />

-1.5<br />

0.4<br />

0.0<br />

-1.0 -0.5 0.0 0.5 1.0 1.5<br />

-0.4<br />

-0.8<br />

log(harem size) contrasts<br />

Figure 2.3. Regression line through the origin on harem size and body weight dimorphism. The data<br />

points are from a phylogenetic independent contrasts analysis. There is a significant<br />

relationship between harem size and sexual size dimorphism (b¼0.376, p¼0.000,<br />

R 2 ¼0.577, n¼36) (from Lindenfors et al., 2002).<br />

extant species (Table 2.1). Systematists recognize 26 monophyletic mammalian<br />

orders (Wilson and Reeder (1993)). When investigating each order separately,<br />

the majority of orders also turned out to be significantly male-biased dimorphic<br />

(average male/female mass ratio >1.0 and p


18 Lindenfors and Tullberg<br />

Table 2.1. Summary of the Patterns of Dimorphism Found in Mammals<br />

Order<br />

Number of<br />

recognized<br />

species<br />

Number of<br />

species with<br />

body mass data<br />

Average<br />

dimorphism<br />

Sexual size<br />

dimorphism<br />

Mammalia<br />

All mammals 4629 1370 1.184 p 0.001<br />

Subclass Prototheria<br />

Monotremata (Monotremes) 3 2 1.273 –<br />

Subclass Metatheria<br />

Didelphimorphia (American<br />

63 13 1.323 p ¼ 0.002<br />

marsupials)<br />

Paucituberculata (Shrew oppossums) 5 2 1.840 –<br />

Microbiotheria (Monito del monte) 1 1 1.044 –<br />

Dasyuromorphia (Dasyuroids) 63 24 1.465 p 0.001<br />

Peramelemorphia (Bandicoots<br />

21 9 1.496 p ¼ 0.015<br />

and bilbies)<br />

Notoryctemorphia (Marsupial moles) 2 0 – –<br />

Diprotodontia (Kangaroos, etc.) 117 63 1.306 p 0.001<br />

Subclass Eutheria<br />

Insectivora (Insectivores) 428 59 1.048 p ¼ 0.081<br />

Macroscelidea (Elephant shrews) 15 5 0.964 p ¼ 0.142<br />

Scandentia (Tree shrews) 19 1 – –<br />

Dermoptera (Colugos) 2 0 – –<br />

Chiroptera (Bats) 925 354 0.999 p ¼ 0.091<br />

Primates (Primates) 233 198 1.247 p 0.001<br />

Xenarthra (sloths, armadillos,<br />

29 4 0.914 p ¼ 0.216<br />

and anteaters)<br />

Pholidota (Pangolins) 7 3 1.767 p ¼ 0.001<br />

Lagomorpha (Rabbits and pikas) 80 21 0.930 p ¼ 0.012<br />

Rodentia (Rodents) 2015 295 1.092 p 0.001<br />

Cetacea (Whales, dolphins, and 78 10 1.414 p ¼ 0.082<br />

porpoises)<br />

Carnivora (Carnivores) 271 180 1.476 p 0.001<br />

Tubulidentata (Aardwark) 1 0 – –<br />

Proboscidea (Elephants) 2 2 1.900 –<br />

Hyracoidea (Hyraxes) 6 1 1.111 –<br />

Sirenia (Dugongs and manatees) 5 0 – –<br />

Perissodactyla (Horses, rhinos,<br />

18 8 1.164 p ¼ 0.156<br />

and tapirs)<br />

Artiodactyla (Antelopes,<br />

camels, pigs, etc.)<br />

220 115 1.340 p 0.001<br />

Dimorphism is given as male mass/female mass. Mammals and the majority of mammalian orders<br />

are on average male-biased dimorphic (average dimorphism>1.0 and p0.05) or female-biased dimorphism (Lagomorpha:<br />

average dimorphism


2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 19<br />

“more” and “less” sexually selected (polygynous) sister taxa. These tests<br />

revealed that a higher degree of sexual selection was associated with a higher<br />

degree of male-biased dimorphism. More polygynous taxa not only had larger<br />

males but also larger females than their less polygynous sister taxa. These results<br />

indicate that sexual selection is a significant explanatory factor of both sexual<br />

dimorphism as such, and of the general size increase in many mammalian<br />

lineages.<br />

In primates, the mammal order humans belong to, the pattern is similar.<br />

Again using mating system as a three-state unordered categorical variable,<br />

testing for differences in dimorphism between “more” and “less” sexually selected<br />

sister taxa, a higher degree of sexual selection was associated with a higher degree<br />

of male-biased dimorphism. Again, more polygynous taxa also had larger males<br />

and females than their less polygynous sister taxa (Lindenfors, 2002; Lindenfors<br />

and Tullberg, 1998). Here, however, a novel method investigating temporal<br />

order of events revealed not only a correlation but also a causal link between<br />

sexual selection and sexual size dimorphism where changes in mating systems<br />

occurred before changes in the degree of sexual selection (Lindenfors and<br />

Tullberg, 1998). Using similar methods, sexual selection has also been shown<br />

to be an important determinant of sexual dimorphism in canine size in primates<br />

(Thorén et al., 2006), although primate canines are also of importance in<br />

predator defense (Harvey et al., 1978). Thus, both body size and canine size<br />

bear witness to an evolutionary history of male–male aggression in primates.<br />

This selection history has its grounds in a sexual difference in behavior.<br />

While males compete more over matings than females, female reproduction is<br />

instead limited by resource allocation (Emlen and Oring, 1977). These differing<br />

demands should be expected to produce variation in the relative sizes of various<br />

brain structures, just as they are expected to produce differences in other morphological<br />

structures. However, data on brain structures in primates are not<br />

available for males and females separately. Instead, investigating species differences<br />

in brain structures and comparing them on basis of differences in the<br />

species-typical degree of sexual selection, research has shown that the degree<br />

of male intrasexual selection is positively correlated with several structures<br />

involved in autonomic functions and sensory-motor skills, and in pathways<br />

relating to aggression and aggression control (Lindenfors et al., 2007b).<br />

The sizes of the mesencephalon, diencephalon (containing the hypothalamus),<br />

and amygdala, all involved in governing aggressive behaviors, are<br />

positively correlated with the degree of sexual selection, whereas the size of the<br />

septum, which has a role in facilitating aggression control, is negatively correlated<br />

with the degree of sexual selection. These correlations indicate that sexual<br />

selection affects physical combat skills. Moreover, male group size was positively<br />

correlated with the relative volume of the diencephalon and negatively<br />

correlated with relative septum size, further strengthening the conclusion that


20 Lindenfors and Tullberg<br />

aggression is an evolutionarily important component of male–male interactions<br />

(Lindenfors et al., 2007b). Thus, primate brain organization also reflects a history<br />

of male–male aggression.<br />

VI. HUMANS AND THE MAMMALIAN PATTERN<br />

So where do humans fit into this picture Humans are one of the sexually sizedimorphic<br />

species in the primate order in Table 2.1. But compared to our closest<br />

relatives (chimpanzees, bonobos, gorillas, and orangutans) humans have the<br />

lowest degree of size dimorphism (Lindenfors, 2002), indicating a decreasing<br />

degree of sexual selection over human evolution. Nevertheless, in all measurements<br />

of length that have ever been carried out in human populations, males<br />

have been taller than females. The average dimorphism in humans from these<br />

surveys is 1.07 (Gustafsson and Lindenfors, 2004). Does this mean that we<br />

exhibit a tendency toward the polygyny that accompanies such size dimorphism<br />

According to the Ethnographic Atlas Codebook (Gray, 1999), a database<br />

of cultural characteristics for 1231 comparable cultures from around the<br />

world, polygyny is common in 48% of human societies. In another 37% polygyny<br />

is allowed, and in only 15% is monogamy the norm. Only four reported societies<br />

are considered polyandrous. From these data and the degree of human size<br />

dimorphism, one may draw the conclusion that humans are at least more<br />

polygynous than monogamous (see also Low, 2000), and also that Western<br />

cultures fall within the monogamous 15% of the world. Interestingly, intergroup<br />

differences in size dimorphism are not correlated with differences in the degree of<br />

polygyny (Gustafsson and Lindenfors, 2004), a clear indication that cultural<br />

evolution proceeds faster than biological evolution.<br />

There are more indications that humans have an evolutionary history of<br />

sex differences as an explanatory factor in human aggression. For example, men<br />

commit most of the world’s violent acts that are reported to the police. Men are<br />

consequently overrepresented in the world’s prisons. Women typically make up<br />

only 10–15% of the prison population (Harrendorf et al., 2010). Further,<br />

soldiering is most often an all-male vocation (personal observation). Humans<br />

are, however, the products of both biological and cultural inheritance (Boyd and<br />

Richerson, 2005). Here, we have presented only the biological side of the story,<br />

but we agree with Archer (2009) that sexual selection probably is the best<br />

explanation for the magnitude and nature of human sex differences in aggression.<br />

Humans fit into the mammalian scheme of things very well.<br />

Acknowledgment<br />

This research was supported through a generous research grant from the Swedish Research Council<br />

(P. L.).


References<br />

2. Evolutionary Aspects of Aggression: The Importance of Sexual Selection 21<br />

Altmann, J. (1990). Primate males go where the females are. Anim. Behav. 39, 193–195.<br />

Andersson, M. (1994). Sexual Selection. Princeton <strong>University</strong> Press, New Jersey.<br />

Archer, J. (2009). Does sexual selection explain human sex differences in aggression Behav. Brain<br />

Sci. 32, 249–311.<br />

Arnqvist, G., and Rowe, L. (2005). Sexual Conflict. Princeton <strong>University</strong> Press, Princeton.<br />

Bateman, A. J. (1948). Intra-sexual selection in Drosophila. Heredity 2, 349–368.<br />

Bininda-Emonds, O. R. P., et al. (2007). The delayed rise of present-day mammals. Nature 446,<br />

507–512.<br />

Birkhead, T. (2001). Promiscuity: An Evolutionary History of Sperm Competition. Harvard<br />

<strong>University</strong> Press, Cambridge.<br />

Boyd, R., and Richerson, P. J. (2005). The Origin and Evolution of Cultures. Oxford <strong>University</strong><br />

Press, Oxford.<br />

Brick, O. (1999). A test of the sequential assessment game: The effect of increased cost of sampling.<br />

Behav. Ecol. 10, 726–732.<br />

Caro, T. M., Graham, C. M., Stoner, C. J., and Flores, M. M. (2003). Correlates of horn and antler<br />

shape in bovids and cervids. Behav. Ecol. Sociobiol. 55, 32–41.<br />

Clutton-Brock, T. H., and Albon, S. D. (1979). The roaring of red deer and the evolution of honest<br />

advertisement. Behaviour 69, 146–170.<br />

Darwin, C. (1859). The Origin of Species by Means of Natural Selection. Penguin, London.<br />

Darwin, C. (1860). Darwin Correspondence Project. Letter 2743—Darwin, C. R. to Gray, Asa,<br />

3 Apr (1860).<br />

Darwin, C. (1871). The Descent of Man and Selection in Relation to Sex. Murray, London.<br />

Dugatkin, L. A., and Godin, J.-G. J. (1993). Female mate copying in the guppy (Poecilia reticulata):<br />

Age-dependent effects. Behav. Ecol. 4, 289–292.<br />

Emlen, S. T., and Oring, L. W. (1977). Ecology, sexual selection, and the evolution of mating<br />

systems. Science 197, 215–223.<br />

Enquist, M., and Leimar, O. (1983). Evolution of fighting behavior: Decision rules and assessment of<br />

relative strength. J. Theor. Biol. 102, 387–410.<br />

Fabiani, A., Galimberti, F., Sanvito, S., and Hoelzel, A. R. (2004). Extreme polygyny among<br />

southern elephant seals on Sea Lion Island, Falkland Islands. Behav. Ecol. 15, 961–969.<br />

Fairbairn, D. J., Blanckenhorn, W. U., and Székely, T. (eds.) (2007). In “Sex, Size and Gender Roles:<br />

Evolutionary Studies of Sexual Size Dimorphism”. Oxford <strong>University</strong> Press, Oxford.<br />

Gavrilets, S., Arnqvist, G., and Friberg, U. (2001). The evolution of female mate choice by sexual<br />

conflict. Proc. R. Soc. Lond. B 268, 531–539.<br />

Gray, J. P. (1999). A corrected ethnographic atlas. World Cultures 10, 24–85.<br />

Gross, M. R. (1996). Alternative reproductive strategies and tactics: Diversity within sexes. Trends<br />

Ecol. Evol. 11, 92–97.<br />

Gustafsson, A., and Lindenfors, P. (2004). Human size evolution: No allometric relationship between<br />

male and female stature. J. Hum. Evol. 47, 253–266.<br />

Harrendorf, S., Heiskanen, M., and Malby, S. (2010). International Statistics on Crime and Justice.<br />

United Nations Office on Drugs and Crime (UNODC), Helsinki, Finland.<br />

Harvey, P. H., Kavanagh, M., and Clutton-Brock, T. H. (1978). Sexual dimorphism in primate teeth.<br />

J. Zool. 186, 475–485.<br />

Holland, B., and Rice, W. R. (1998). Chase-away sexual selection: Antagonistic seduction versus<br />

resistance. Evolution 52, 1–7.<br />

Hrdy, S. B. (1981). The Woman that Never Evolved. Harvard <strong>University</strong> Press, Cambridge.<br />

Lindenfors, P. (2002). Sexually antagonistic selection on primate size. J. Evol. Biol. 15, 595–607.


22 Lindenfors and Tullberg<br />

Lindenfors, P., and Tullberg, B. S. (1998). Phylogenetic analyses of primate size evolution:<br />

The consequences of sexual selection. Biol. J. Linn. Soc. 64, 413–447.<br />

Lindenfors, P., Tullberg, B., and Biuw, M. (2002). Phylogenetic analyses of sexual selection and<br />

sexual size dimorphism in pinnipeds. Behav. Ecol. Sociobiol. 52, 188–193.<br />

Lindenfors, P., Fröberg, L., and Nunn, C. L. (2004). Females drive primate social evolution.<br />

Proc. R. Soc. Lond. B 271, S101–S103.<br />

Lindenfors, P., Gittleman, J. L., and Jones, K. E. (2007a). Sexual size dimorphism in mammals.<br />

In “Sex, Size and Gender Roles: Evolutionary Studies of Sexual Size Dimorphism” (D. J. Fairbairn,<br />

W. U. Blanckenhorn, and T. Szekely, eds.), pp. 19–26. Oxford <strong>University</strong> Press, Oxford.<br />

Lindenfors, P., Nunn, C. L., and Barton, R. A. (2007b). Primate brain architecture and selection in<br />

relation to sex. BMC Biol. 5, 20.<br />

Low, B. S. (2000). Why Sex Matters. Princeton <strong>University</strong> Press, Princeton.<br />

Maynard Smith, J. (1982). Evolution and the Theory of Games. Cambridge <strong>University</strong> Press,<br />

Cambridge.<br />

Mitani, J. C., Gros-Louis, J., and Manson, J. H. (1996). Number of males in primate groups:<br />

Comparative tests of competing hypotheses. Am. J. Primatol. 38, 315–332.<br />

Nunn, C. L. (1999). The number of males in primate social groups: A comparative test of the<br />

socioecological model. Behav. Ecol. Sociobiol. 46, 1–13.<br />

Parker, G. A. (1979). Sexual selection and sexual conflict. In “Sexual Selection and Reproductive<br />

Competition in Insects” (M. S. Blum and N. A. Blum, eds.), pp. 123–166. Academic Press,<br />

New York.<br />

Petrie, M. (1994). Improved growth and survival of offspring of peacocks with more elaborate trains.<br />

Nature 371, 598–599.<br />

Ralls, K. (1976). Mammals in which females are larger than males. Q. Rev. Biol. 51, 245–276.<br />

Stankowich, T., and Caro, T. (2009). Evolution of weaponry in female bovids. Proc. R. Soc. Lond. B<br />

276, 4329–4334.<br />

Thorén, S., Lindenfors, P., and Kappeler, P. M. (2006). Phylogenetic analyses of dimorphism in<br />

primates: Evidence for stronger selection on canine size than on body size. Am. J. Phys. Anthropol.<br />

130, 50–59.<br />

Tinbergen, N. (1963). On aims and methods in ethology. Z. Tierpsychol. 20, 410–433.<br />

Trivers, R. L. (1972). Parental investment and sexual selection. In “Sexual Selection and the Descent<br />

of Man 1871–1971” (B. Campbell, ed.), pp. 136–179. Aldine, Chicago, IL.<br />

Vincent, A., Ahnesjö, I., Berglund, A., and Rosenqvist, G. (1992). Pipefishes and seahorses: Are they<br />

all sex role reversed Trends Ecol. Evol. 7, 237–241.<br />

Wade, M. J., and Pruett-Jones, S. G. (1990). Female copying increases the variance in male mating<br />

success. Proc. Natl. Acad. Sci. USA 87, 5749–5753.<br />

West, P. M., and Packer, C. (2002). Sexual selection, temperature, and the lion’s mane. Science 297,<br />

1339–1343.<br />

Williams, G. C. (1966). Adaptation and Natural Selection. Princeton <strong>University</strong> Press, Princeton.<br />

Wilson, E. O. (1975). Sociobiology, the New Synthesis. Belknap, Cambridge, MA.<br />

Wilson, D. E., and Reeder, D. M. (eds.) (1993). In “Mammal Species of the World: A Taxonomic and<br />

Geographic Reference”. 2 nd edn. Smithsonian Institution Press, Washington and London.


3<br />

Signaling Aggression<br />

Moira J. van Staaden,* William A. Searcy, †<br />

and Roger T. Hanlon ‡<br />

*Department of Biological Sciences and JP Scott Center for Neuroscience,<br />

Mind & Behavior, <strong>Bowling</strong> <strong>Green</strong> <strong>State</strong> <strong>University</strong>, <strong>Bowling</strong> <strong>Green</strong>, Ohio,<br />

USA<br />

† Department of Biology, <strong>University</strong> of Miami, Coral Gables, Florida, USA<br />

‡ Marine Resources Center, Marine Biological Laboratory, Woods Hole,<br />

Massachusetts, USA<br />

I. Introduction<br />

A. An ethological approach to aggression<br />

B. The classic game theory model<br />

C. Signaling games<br />

D. Threat displays and why they are part of aggression<br />

E. Evolutionary issues<br />

F. The challenge of “incomplete honesty”<br />

G. Case studies in aggressive signaling<br />

II. Bird Song Signals Aggressive Intentions: Speak Softly and<br />

Carry a Big Stick<br />

III. Visual Displays Signal Aggressive Intent in Cephalopods:<br />

The Sweet Smell of Success<br />

A. Cuttlefish agonistic bouts<br />

B. Squid agonistic bouts<br />

C. From molecules to aggression: Contact pheromone triggers<br />

strong aggression in squid<br />

D. Signaling aggression in humans<br />

Acknowledgments<br />

References<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00008-3


24 van Staaden et al.<br />

ABSTRACT<br />

From psychological and sociological standpoints, aggression is regarded as intentional<br />

behavior aimed at inflicting pain and manifested by hostility and attacking<br />

behaviors. In contrast, biologists define aggression as behavior associated with<br />

attack or escalation toward attack, omitting any stipulation about intentions and<br />

goals. Certain animal signals are strongly associated with escalation toward<br />

attack and have the same function as physical attack in intimidating opponents<br />

and winning contests, and ethologists therefore consider them an integral part of<br />

aggressive behavior. Aggressive signals have been molded by evolution to make<br />

them ever more effective in mediating interactions between the contestants.<br />

Early theoretical analyses of aggressive signaling suggested that signals could<br />

never be honest about fighting ability or aggressive intentions because weak<br />

individuals would exaggerate such signals whenever they were effective in<br />

influencing the behavior of opponents. More recent game theory models, however,<br />

demonstrate that given the right costs and constraints, aggressive signals<br />

are both reliable about strength and intentions and effective in influencing<br />

contest outcomes. Here, we review the role of signaling in lieu of physical<br />

violence, considering threat displays from an ethological perspective as an<br />

adaptive outcome of evolutionary selection pressures. Fighting prowess is conveyed<br />

by performance signals whose production is constrained by physical ability<br />

and thus limited to just some individuals, whereas aggressive intent is encoded in<br />

strategic signals that all signalers are able to produce. We illustrate recent<br />

advances in the study of aggressive signaling with case studies of charismatic<br />

taxa that employ a range of sensory modalities, viz. visual and chemical signaling<br />

in cephalopod behavior, and indicators of aggressive intent in the territorial calls<br />

of songbirds. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

Although physical fighting, including the killing of conspecifics, is widespread in<br />

nonhuman animals just as it is in humans, the majority of contests and disputes<br />

in nonhuman animals are settled without physical fighting. Rather than resorting<br />

to immediate physical combat, nonhuman animals often engage instead in<br />

extended bouts of signaling, making prominent display of their weapons (e.g.,<br />

antlers, claws, and teeth), or running through a repertoire of highly stereotyped<br />

agonistic signals. With their high cognitive capacity, primates (humans<br />

included) are particularly good at reducing social tensions and resolving conflicts<br />

using agonistic signaling as opposed to sheer physical force (Cheney et al., 1986).


3. Signaling Aggression 25<br />

Such aggressive signaling is found in virtually all of the multicellular<br />

taxa and can involve all communication modalities. Orthoptera (Alexander,<br />

1961; Simmonds and Bailey, 1993) and many other insects (Clark and Moore,<br />

1995; Jonsson et al., 2011) use aggressive song to defend resources, and the use of<br />

territorial song in birds is well known (Searcy and Yasukawa, 1990; Stoddard<br />

et al., 1988). Calls are employed to similar effect in the dramatic displays of large<br />

mammals or frog choruses (Bee et al., 1999; Reby et al., 2005; Wagner, 1992),<br />

and more subtly by other vertebrate taxa such as fish (Raffinger and Ladich,<br />

2009). In these scenarios, signaling can be just as effective as physical attack in<br />

intimidating opponents and winning contested resources.<br />

Chemical signals are widely used to signal resource defense and fighting<br />

ability, deposited either as scent marks in fixed locales by terrestrial species (Page<br />

and Jaeger, 2004) or contained in urine released during aggressive interactions in<br />

some aquatic organisms (Breithaupt and Eger, 2002). Visual signals are perhaps<br />

the most familiar and easily appreciated of aggressive displays, beginning with<br />

Darwin’s (1871) graphic illustration of aggression and fear in the facial expression<br />

of the domestic dog. Visual signs of aggression include variable pigment<br />

patterns of many fish and cephalopods (DiMarco and Hanlon, 1997; Moretz and<br />

Morris, 2003), and the ritualized display of weapons (Huber and Kravitz, 1995;<br />

Lundrigan, 1996) or inedible objects as “props” (Murphy, 2008).<br />

Phylogenetic comparative analyses demonstrate that many of these<br />

aggressive signals allowing opponents to resolve contests without physical<br />

harm evolved from nonsignaling behaviors through the process of ritualization<br />

(Scott et al., 2010; Turner et al., 2007). Whereas agonistic behavior runs the<br />

gamut from passivity, defense, and escape to <strong>full</strong> conflict, here, we reserve the<br />

terms aggressive/threatening behavior for that subset of agonistic behavior associated<br />

with the escalation toward physical fighting (Searcy and Beecher, 2009).<br />

A. An ethological approach to aggression<br />

The ethological approach to aggression derives historically from the traditional<br />

instincts and drives articulated by Lorenz (1978). Although the simple psychohydraulic<br />

model of motivation underlying this view proved inadequate in the<br />

long term, the idea that aggression is based on both internal state and external<br />

stimuli, and the proposed value of a comparative evolutionary approach, were<br />

both far-sighted and enduring. The classic On Aggression (Lorenz, 1963) which<br />

was written for a popular audience, highlighted aggression as a natural, evolved<br />

function, with a founding basis in other instincts, and a central role in animal<br />

communication. A more nuanced view is found in his work known as the Russian<br />

manuscript (Lorenz, 1995). In this, Lorenz discussed animals and humans separately,<br />

not because of any fundamental difference in their biology, but because<br />

he believed it necessary for the reader to have an adequate frame of reference.


26 van Staaden et al.<br />

Much current research on the biology of aggression focuses on identifying<br />

the physiological substrate to violence (i.e., on proximate cause and nonadaptive<br />

features). The ethological or sociobiological approach, in contrast,<br />

focuses attention on the ultimate causes and adaptive forms of aggressive behavior<br />

(e.g., Chen et al., 2002; Huber and Kravitz, 1995; Miczek, et al., 2007; Natarajan<br />

et al., 2009): how and why has evolution molded complex agonistic interactions<br />

built on reciprocal displays of threat or submission, affect or intent<br />

B. The classic game theory model<br />

Evolutionary fitness is measured in terms of the number of offspring an individual<br />

produces over the course of its lifetime. In the evolutionary race to transmit their<br />

genes to the following generations at a higher frequency than that of their<br />

conspecifics, these individuals must compete for access to all the resources<br />

necessary to create and raise their progeny, including mates, dominance rights,<br />

and desirable territory. Winners in this intraspecific competition thus stand to<br />

gain both immediate personal advantages such as food, space, and safety, as well<br />

as long-term evolutionary fitness, that is, more offspring and therefore copies of<br />

their genes in subsequent generations. Simulation approaches from game theory<br />

have long provided a theoretical framework for analyzing and predicting the<br />

outcomes of competitive interactions. The classic “Hawks” and “Doves” game<br />

(Maynard Smith and Price, 1973) considers symmetrical contests between pairs<br />

of individuals who are equivalent in every respect (equal size, strength, fighting<br />

ability, etc.), differing only in behavioral/fighting strategy in intraspecific<br />

encounters. Hawk strategists are those who will always choose to fight when<br />

they encounter a conspecific at a contested resource. Dove strategists, in contrast,<br />

always retreat from an individual behaving as a Hawk, rather than engage them<br />

in combat. Hawks always best Doves, but they incur costs when they compete<br />

against other Hawks. The outcome between two Dove strategists is randomly<br />

determined. Each conflict consists of a series of agonistic moves (incorporating<br />

provocation, escalation, retaliation, etc.) with rewards or costs assigned to each<br />

contestant according to a particular payoff matrix (Table 3.1).<br />

Populations are expected to converge on an evolutionarily stable strategy<br />

(ESS), strategies that once they are predominant cannot be invaded by any<br />

other strategy. The ESS depends critically on the ratio of what an individual<br />

stands to gain over what it stands to lose in a fight. Thus, in the common<br />

situation where the cost of injury exceeds the benefits of winning, populations<br />

are expected to adjust to balanced proportions of the two strategies with the<br />

majority of individuals behaving as Doves, while a smaller number of Hawk<br />

strategists persists. Only in extreme situations where the value of a resource<br />

greatly exceeds the cost of injury, will a Hawk strategy be superior and can<br />

become so widespread as to completely replace the Dove strategy. For instance,


3. Signaling Aggression 27<br />

Table 3.1. The Payoff Matrix for the Hawk–Dove Game Shows the Consequences that Result When<br />

a Player of a Given Strategy (Left Column) Encounters Another Player’s Strategy<br />

Hawk<br />

Dove<br />

Hawk Tie [(V C)/2] Win [V]<br />

Dove Lose [0] Tie [V/2]<br />

Choices are assumed to be rational where each individual would prefer to win, prefer to tie rather<br />

than lose, and prefer to lose over receiving injury. In this payoff matrix, V (value of the contested<br />

resource) and C (cost of an escalated fight) determines the net outcome when different strategies<br />

meet. In encounters between Hawks, the winner gains control over the value of the resource while<br />

the losing Hawk sustains an injury. In the common scenario, where the value of the resource is less<br />

than the cost of injury (i.e., C>V), average payoff in a Hawk meeting a Hawk is negative and<br />

less than that of a Dove meeting a Hawk. Only in rare situations, when the value of the resource<br />

exceeds the cost of injury, will Hawk be unequivocally the superior strategy.<br />

intense fighting among male elephant seals results in the victorious male both<br />

monopolizing a section of the beach and gaining sole reproductive access to the<br />

harem of females which resides there. In the vast majority of cases, however,<br />

resources are rarely worth the risk of injury, and competing individuals would do<br />

best to resolve conflicts via ritualized displays.<br />

C. Signaling games<br />

The earliest game theoretical analyses of aggressive signaling were pessimistic<br />

about the evolutionary stability of such systems (Caryl, 1979; Maynard Smith,<br />

1974, 1979). Their reasoning was that if we assume that signals can help in<br />

winning contests by conveying high levels of aggression or fighting ability, then<br />

it becomes advantageous for all individuals to give the highest levels of these<br />

signals. If all individuals signal maximally, then there is no information in the<br />

signal about either aggressive intentions or fighting ability. The first rigorous<br />

game theoretical model to demonstrate that reliable aggressive signaling could be<br />

evolutionarily stable was a mutual signaling game in which two interactants<br />

chose between two cost-free signals to create a stable global strategy (Enquist,<br />

1985). This model demonstrated how threat displays reveal information about<br />

the strength or condition of the contestants via their choice of action in<br />

aggressive encounters. The players in this game each have a hidden state<br />

(strength or weakness) which determines their ability to win physical fights.<br />

An honest weak individual gives a signal conveying weakness, and abandons the<br />

contest if the other individual gives a signal conveying strength. A dishonest<br />

weak individual can success<strong>full</strong>y bluff other weak individuals by giving the signal<br />

of strength, but at a cost of sometimes being attacked by a better fighter if the


28 van Staaden et al.<br />

opponent turns out to be strong. If the cost of being attacked by a stronger<br />

individual is high relative to the benefit of winning contests, then bluffing may<br />

not be advantageous, and honest signaling can be evolutionarily stable.<br />

There followed a slew of variant Hawk/Dove models which attempted to<br />

accommodate the diversity of interactions between senders and receivers (e.g.,<br />

Enquist and Leimar, 1983; Leimar and Enquist, 1984; Maynard Smith and<br />

Harper, 1988; Skyrms, 2009). These models of communication may be classified<br />

into five structures based on the relative timing of the (signal and/or response)<br />

choices made by the two players during the game (reviewed in Hurd and Enquist,<br />

2005). Mutual signaling games, which most closely resemble agonistic interactions<br />

between animals, are increasingly being used as models (e.g., Kim, 1995;<br />

Számadó, 2000). In this structure, both players signal, and react to their opponent’s<br />

signal, in biologically realistic ways. Genetic algorithms are also being<br />

used to examine non-ESS solutions to these games (Hamblin and Hurd, 2007).<br />

Alternative approaches employ simulation methods and neural networks<br />

(Noble, 2000; Wheeler and de Bourcier, 1995) to explore communication in<br />

animal contests.<br />

D. Threat displays and why they are part of aggression<br />

Aggression is costly to participants not only in terms of energy expenditure and<br />

the potential for injury but also because of opportunity costs. Time spent in<br />

physical conflict is time that is not available for other vital activities such as<br />

exploring, feeding, or mating. Thus, there are selective advantages to reducing<br />

aggression. Threat displays are a critical component of aggression because they<br />

modulate competitive social interactions among conspecifics. If signaling is<br />

effectively delivered by a sender and appropriately interpreted by the intended<br />

receiver it might be so subtle that the interaction is rendered virtually invisible<br />

to an outside observer. Alternatively, if sender and receiver perceive the competitive<br />

difference between them to be slight, the social interaction is prolonged,<br />

escalates in intensity, and may ultimately culminate in levels of overt conflict<br />

that result in physical damage or death of one or both interactants.<br />

In such aggressive signaling contests, two kinds of information are<br />

important to receivers: information on the signaler’s willingness to escalate<br />

(aggressiveness motivation) and on its fighting ability (resource-holding potential)<br />

(Searcy and Beecher, 2009). Classification schemes based on the type of<br />

interaction in which communication takes place and the nature of the signals<br />

used converge on the following signal categories (Hurd and Enquist, 2005;<br />

Maynard Smith and Harper, 2003; Vehrencamp, 2000).<br />

Performance signals are signals constrained to a subset of signalers either<br />

by differences in the ability to perform them (Maynard Smith, 1982), or by<br />

possessing the information needed to produce them (Hurd and Enquist, 2005).


3. Signaling Aggression 29<br />

Performance displays (“index signals” of Maynard Smith and Harper, 2003) have<br />

excellent empirical support, as do models of their use (e.g., Enquist and Leimar,<br />

1983; Leimar and Enquist, 1984). Examples include the lateral displays of many<br />

fish (Enquist and Jakobsson, 1986) or the pitch of calls in many frog and mammal<br />

species (e.g., Bee et al., 1999; Reby et al., 2005), both “unfakeable” signals as they<br />

are determined by the sender’s size and fighting ability.<br />

Strategic signals are available to all signalers, and may be either classic<br />

handicaps or conventional signals (Hurd and Enquist, 2005). Classic handicaps<br />

have some inherent cost, independent of receiver response, and variation in the<br />

level of cost experienced by different individuals produces different optimum<br />

signaling levels (Grafen, 1990). Evidence for handicapped displays is theoretical<br />

(Zahavi, 1987) rather than empirical, though threat displays have been shown to<br />

advertise endurance in lizards (Brandt, 2002) and grasshoppers (<strong>Green</strong>field and<br />

Minckley, 1993). Conventional signals are arbitrary with respect to signal design<br />

and therefore dependent for meaning on an agreement between the signaler and<br />

receiver. Honesty of conventional signals in agonistic interactions is maintained<br />

by two forms of receiver-dependent stabilizing costs (Enquist, 1985; Guilford and<br />

Dawkins, 1995); receiver retaliation (Enquist, 1985) has empirical support<br />

(Molles and Vehrencamp, 2001) and vulnerability handicap (Zahavi, 1987) for<br />

which empirical support is contradictory (Laidre and Vehrencamp, 2008; Searcy<br />

et al., 2006). Most threat displays appear to be conventional signaling systems.<br />

Examples include color patches and song-type sharing in birds (Molles and<br />

Vehrencamp, 2001; Vehrencamp, 2000). Aggressiveness motivation (or willingness<br />

to escalate) is most likely to be encoded this way (Hurd and Enquist, 2005).<br />

E. Evolutionary issues<br />

Empirical analysis of aggressive signaling is more complex than the classic ESS<br />

modeling approach would suggest. This is in large part attributable to the fact<br />

that evolution is not necessarily equilibrial (Houston and McNamara, 1999).<br />

An individual’s success or failure in using signals depends upon how other<br />

individuals use and interpret those signals, that is, it is a trait under frequencydependent<br />

selection (Maynard Smith, 1982). In addition to frequency<br />

dependence of the signal phenotype itself, selection pressures acting on signaler<br />

and receiver in a communicating dyad may be distinct if their genetic interests or<br />

risk profiles (Searcy and Nowicki, 2006) are not identical, or if signals have dual<br />

functions, affecting both aggression and mate choice (Wong and Candolin,<br />

2005). Selection may also modify the responsiveness of other individuals to the<br />

signals (Arak and Enquist, 1995). Thus, like other significant evolutionary<br />

problems such as sexual selection and conflict, signaling strategies may lack<br />

stable equilibria and remain in constant evolutionary flux. Understanding the


30 van Staaden et al.<br />

evolution of behavioral phenotypes under such nonequilibrial conditions<br />

requires dynamic approaches which have yet to be adequately deployed in the<br />

game-theoretical modeling of biological signaling (Hurd and Enquist, 2005).<br />

F. The challenge of “incomplete honesty”<br />

In animal contests, selection should favor displays providing reliable information<br />

about the fighting ability or aggressive intent of competitors. However, considerable<br />

theoretical work predicts that low levels of deception may occur within<br />

otherwise honest signaling systems (Adams and Mesterton-Gibbons, 1995;<br />

Számadó 2000). Strategic signals (i.e., ones of intent) are particularly prone to<br />

such corruption because they typically involve low production costs (Maynard<br />

Smith, 1974, 1979, 1982). Testing for such incomplete honesty is challenging<br />

because it is difficult to distinguish dishonest signals from natural variation in<br />

signal size (Moore et al., 2009), and between a successful bluff and an honest<br />

signal, especially when signaled information is continuous rather than discrete.<br />

Hughes (2000) suggested that dishonesty could be detected by analysis of signal<br />

residuals, the residuals from a measure of the regression of signal structure on<br />

competitive ability. Whereas receivers take advantage of the strong relationship<br />

between signal and fighting ability, for example, signalers take advantage of the<br />

variation around this relationship. If individuals who exaggerate signals benefit<br />

from doing so, they should perform more repetitions of the signaling activity than<br />

those who do not exaggerate (Hughes, 2000). Empirical examples of incomplete<br />

honesty, though still comparatively rare, suggest this is not a fixed behavioral<br />

trait, and depends on con<strong>text</strong> as well as signal residuals (Arnott and Elwood,<br />

2010; Hughes, 2000; Lailvaux et al., 2009).<br />

G. Case studies in aggressive signaling<br />

Using animal models and invasive techniques (e.g., drugs, hormones, brain<br />

lesions, and gene knockouts), we have made great strides in unraveling the<br />

mechanisms and internal states underlying aggression in controlled lab situations.<br />

This is true also with respect to aggressive signaling (see Chapter 5).<br />

Studies of nonmodel organisms are a necessary complement to this approach as<br />

these can provide the telling exceptions in field situations where more complex<br />

social/physical environments permit <strong>full</strong> expression of behaviors and analysis of<br />

adaptive function (see Logue et al., 2010). Below, we present two case studies of<br />

taxa employing multimodal signaling systems to art<strong>full</strong>y modulate aggressive<br />

interactions in complex social systems.


3. Signaling Aggression 31<br />

II. BIRD SONG SIGNALS AGGRESSIVE INTENTIONS: SPEAK SOFTLY<br />

AND CARRY A BIG STICK<br />

The use of song by songbirds provides an excellent illustration of how signals<br />

function in aggression in nonhuman animals. The songbirds (suborder Passeres)<br />

consist of over 4000 species of birds, which are distinguished in part by their<br />

intricate vocal musculature. This musculature functions most importantly in the<br />

production of the complex vocalizations from which the songbirds derive their<br />

name. Most species in the group are territorial and monogamous, and their songs<br />

are used in both territory defense and mate attraction (Catchpole and Slater,<br />

2008; Searcy and Andersson, 1986). At least in temperate zone species, songs are<br />

given mainly by males and mainly during the breeding season. Some attributes of<br />

song and singing behavior have evolved to function in attracting females and<br />

persuading them to mate, but others have evolved to function in aggressive<br />

communication between males in the con<strong>text</strong> of claiming and defending<br />

a territory.<br />

Many of the signals employed by songbirds in aggressive communication<br />

can be illustrated using the signaling behavior of song sparrows (Melospiza melodia).<br />

Song sparrow songs (Fig. 3.1) are multiparted—that is, they contain multiple<br />

phrases differing in structure (Mulligan, 1963). Individual males sing several<br />

versions of the species’ song, each consisting of a distinct and largely nonoverlapping<br />

set of phrases. These distinct versions are called song types (Fig. 3.1), and the<br />

collection of song types sung by one male is his song repertoire. Repertoire sizes vary<br />

geographically in song sparrows, with averages in the range of 8–12 song types per<br />

male (Peters et al., 2000). Male song sparrows produce their repertoires with<br />

“eventual variety,” meaning that they sing several to many repetitions of<br />

one song type before switching to another. The successive repetitions of a song<br />

type are themselves typically not identical, but instead show differences that are<br />

audible (Borror, 1965; Saunders, 1924) but of lower magnitude than differences<br />

between song types (Nowicki et al., 1994). The minor variations of a song type are<br />

termed song variants (Fig. 3.1). Song sparrows respond to differences between song<br />

variants (Stoddard et al., 1988) but less strongly than to differences between song<br />

types (Searcy et al., 1995).<br />

In some species of songbirds, different song types have different functions;<br />

for example, in wood warblers (Parulidae) some song types may be<br />

specialized for male–female communication and others for male–male signaling<br />

(Byers, 1996; Spector, 1992; Weary et al., 1994; but see Beebee, 2004). In song<br />

sparrows, however, all song types are thought to be functionally equivalent, and<br />

in that sense “redundant.” Even with redundant song types, however, certain<br />

signals can be produced with a repertoire of song types that are not possible with<br />

a single type. Some of these signals have been suggested to be aggressive.


32 van Staaden et al.<br />

Figure 3.1. Spectrograms of two variants of each of three song types from a male song sparrow<br />

recorded in northwestern Pennsylvania. Each row shows two variants of one song type.<br />

Note that virtually every note differs between the different song types, whereas the two<br />

variants of any one song type tend to differ only in their endings.<br />

Singing behaviors associated with aggressive con<strong>text</strong>s in song sparrows<br />

include:<br />

1. Song-type switching. If a bird sings more than one song type, it can vary the<br />

frequency with which it switches between song types, and switching frequency<br />

becomes a possible signal. Song-type switching frequency has been suggested<br />

to be a conventional signal of aggression (Vehrencamp, 2000)—conventional<br />

in the sense that the meaning of the signal is arbitrary with respect to its form.<br />

In song sparrows, type-switching frequency increases in aggressive con<strong>text</strong>s,<br />

for example, during counter singing between territorial males or when an<br />

outside male intrudes on a territory (Kramer and Lemon, 1983; Kramer<br />

et al., 1985; Searcy et al., 2000). In other species, the opposite pattern<br />

holds—type-switching frequency decreases in aggressive con<strong>text</strong>s (Molles<br />

and Vehrencamp, 1999; Searcy and Yasukawa, 1990). The fact that either<br />

pattern can occur supports the arbitrariness of the signal (Vehrencamp, 2000).<br />

2. Variant switching. In song sparrows, variant-switching frequency also<br />

increases in aggressive con<strong>text</strong>s, and the increase is if anything more<br />

consistent than the increase in type switching (Searcy et al., 2000). Given


3. Signaling Aggression 33<br />

the evidence that male song sparrows attend to variant switching (Searcy<br />

et al., 1995; Stoddard et al., 1988), variant-switching frequency is another<br />

potential aggressive signal.<br />

3. Song-type matching. Matching is a behavior in which one male replies to a<br />

rival with the same song type that the rival has just sung. Matching can occur<br />

by chance, but in song sparrows it has been shown that when wholly or<br />

partially shared songs are played to males on or near their territories, those<br />

males match the playback songs at levels significantly higher than chance<br />

(Anderson et al., 2005; Burt et al., 2002; Stoddard et al., 1992). Song sparrows<br />

match strangers more than neighbors (Stoddard et al., 1992), and are more<br />

aggressive in general toward strangers (Stoddard et al., 1990), providing<br />

further support for matching as an aggressive signal.<br />

4. Song rate. The number of songs produced per unit time is a parameter that<br />

birds can vary even if they sing only a single song type. In some species of<br />

songbirds, territory owners consistently increase song rates in aggressive<br />

con<strong>text</strong>s (Vehrencamp, 2000). Song sparrows have shown this pattern in<br />

some experiments (Kramer et al., 1985) but not in others (Peters et al., 1980;<br />

Searcy et al., 2000).<br />

5. Soft song. In her classic monograph on song sparrow behavior, Nice (1943)<br />

noted that during intense aggressive encounters, male song sparrows produce<br />

songs of especially low amplitude. In some other songbirds, such soft songs are<br />

produced during courtship as well as during aggression (Dabelsteen et al.,1998),<br />

but in song sparrows they apparently are given only in aggressive con<strong>text</strong>s.<br />

Anderson et al. (2008) found that the amplitude of soft songs was as much as<br />

36 dB lower than the amplitude of the loudest normal or “broadcast” songs.<br />

The five singing behaviors listed above are all associated with aggressive<br />

con<strong>text</strong>s in song sparrows, but signals used in aggressive con<strong>text</strong>s can convey<br />

submission or escape as well as attack, in which case they would be considered<br />

“agonistic” but not “aggressive.” These alternative interpretations seem particularly<br />

likely a priori in the case of soft songs. To test whether a signal is aggressive<br />

rather than submissive, it is necessary to determine whether the signal predicts<br />

aggressive escalation (Searcy and Beecher, 2009). Aggressive escalation includes<br />

outright physical attack of course, but also includes other behaviors that lead up<br />

to attack, such as approach to a rival or giving signals that are higher in a<br />

hierarchy of aggressive signaling.<br />

A test of the predictive power of singing behaviors was carried out for<br />

song sparrows by Searcy et al. (2006). In this study, a brief playback of song<br />

sparrow song was used to elicit aggressive signaling from a territory owner. After a<br />

5-min period during which displays were recorded, a taxidermic mount of a song<br />

sparrow was revealed on the subject’s territory, posed above the loudspeaker, in<br />

conjunction with another brief playback. The subject was then given a set period


34 van Staaden et al.<br />

of time (14 min) to attack or not attack the mount. Of 95 males that were tested,<br />

20 attacked and 75 did not. The display behavior of attackers and nonattackers<br />

was then compared, focusing on the five singing behaviors discussed above, plus<br />

wing-waving, a display in which a male fans one or both wings while remaining<br />

perched; this is the most prominent visual display given by song sparrows during<br />

aggressive contests. For the initial recording period, none of the display measures<br />

differed significantly between attackers and nonattackers, though the number of<br />

soft songs approached significance. A second analysis focused on the 1-min period<br />

directly before attack in the attacking subjects, using a matching time period in<br />

nonattackers as the control. Here, number of soft songs was significantly higher in<br />

attackers than nonattackers, whereas none of the other five measures differed<br />

(Fig. 3.2). In single-variable discriminant function analyses, the number of soft<br />

songs was the only display that discriminated between attackers and nonattackers;<br />

this display correctly predicted presence/absence of attack in 74% of the tested<br />

males. Soft song is thus a reliable signal of aggressive intentions in song sparrows.<br />

A B C<br />

Soft songs<br />

5<br />

4<br />

3<br />

P = 0.00015<br />

2<br />

1<br />

0<br />

No Yes<br />

Type switching frequency<br />

0.2<br />

NS<br />

0.15<br />

0.1<br />

0.05<br />

0<br />

No Yes<br />

Variant switching frequency<br />

1<br />

0.75<br />

0.5<br />

0.25<br />

0<br />

NS<br />

No<br />

Yes<br />

D<br />

8<br />

E<br />

1<br />

F<br />

1.5<br />

Total songs<br />

6<br />

4<br />

2<br />

NS<br />

Matching songs<br />

0.75<br />

0.5<br />

0.25<br />

NS<br />

Wing waves<br />

1<br />

0.5<br />

NS<br />

0<br />

No<br />

Attack<br />

Yes<br />

0<br />

No<br />

Attack<br />

Yes<br />

0<br />

No<br />

Attack<br />

Yes<br />

Figure 3.2. Display measures (means.e.) for the 1 min just prior to attack for male song sparrows<br />

that attacked compared to a matching 1-min period for nonattackers. The display<br />

measures are (A) number of soft songs, (B) type-switching frequency, (C) variantswitching<br />

frequency, (D) total songs, (E) number of matching songs, and (F) number<br />

of bouts of wing-waving. Only soft songs showed a significant difference between<br />

attackers and nonattackers. Redrawn from data in Searcy et al. (2006).


3. Signaling Aggression 35<br />

The use of soft, low-amplitude vocalizations as the most threatening of<br />

signals is somewhat counterintuitive, but this result has since been replicated in<br />

additional species. Ballentine et al. (2008) did a parallel study of aggressive<br />

signaling in swamp sparrows (Melospiza georgiana), a close relative of song<br />

sparrows, using methods similar to those of Searcy et al. (2006). Swamp sparrows<br />

have simpler songs than song sparrows, but again have repertoires of apparently<br />

redundant song types. In addition to songs, males give two types of calls in<br />

aggressive con<strong>text</strong>s, buzzes and wheezes (Ballentine et al., 2008; Mowbray,<br />

1997). In swamp sparrows as in song sparrows, wing-waving is the most prominent<br />

visual display given during aggressive encounters.<br />

In 40 trials with swamp sparrows, 9 males attacked a taxidermic mount<br />

of a conspecific male and 31 did not. For the initial recording period, five of seven<br />

display measures did not differ between attackers and nonattackers; these were<br />

switching frequency, number of matching songs, number of broadcast songs,<br />

number of rasps, and number of wheezes. Two measures were significantly higher<br />

in attackers: number of soft songs and number of wing waves. In a forward,<br />

stepwise discriminant function analysis, soft songs entered first, followed by rasps,<br />

and these together correctly classified 83% of males as attackers or nonattackers.<br />

For the 1 min prior to attack, soft songs and wing waves were again the only two<br />

display measures that differed between attackers and nonattackers. For this time<br />

period, a discriminant function including soft songs and wing waves was the best<br />

predictor of attack, classifying 85% of males correctly.<br />

Hof and Hazlett (2010) have recently performed a similar experiment<br />

with black-throated blue warblers (Dendroica caerulescens), which are also in the<br />

songbird suborder but in another family (Parulidae). In 54 trials with blackthroated<br />

blue warblers, 19 males attacked the mount and 35 did not. Hof and<br />

Hazlett (2010) compared attackers and nonattackers for four display measures:<br />

type-switching frequency, total number of songs, number of soft songs, and<br />

number of ctuk calls. For both an initial recording period and the 1 min prior<br />

to attack, only the number of soft songs differed significantly between attackers<br />

and nonattackers, with attackers giving substantially more. In logistic regressions<br />

based on either time period, soft song was the only significant predictor of attack.<br />

In a logistic regression that incorporated displays for the entire trial, soft song<br />

correctly predicted attack behavior in a very impressive 93% of subjects.<br />

In all three of the songbird species reviewed above, most of the displays<br />

given in aggressive con<strong>text</strong>s are not predictive of attack. One theory about such<br />

displays is that they were at one time predictors of attack, but that over evolutionary<br />

time their reliability was undermined by the spread of bluffing<br />

(Andersson, 1980). If an aggressive display is beneficial in intimidating opponents,<br />

such that the benefit of giving it is greater than any costs, then selection<br />

will favor its use in individuals that do not intend to attack as well as in those<br />

that do. Use of the display will then increase in frequency among individuals not


36 van Staaden et al.<br />

intending attack, until at some point the signal ceases to be informative about<br />

attack likelihood. Another hypothesis is that these agonistic displays have<br />

evolved to convey messages other than imminent attack. Possible alternative<br />

messages include at one extreme retreat or submission, but another possibility is<br />

for a display to threaten a degree of aggressive escalation that falls short of attack.<br />

Song-type matching in song sparrows, for example, has been suggested to be part<br />

of a hierarchy of progressively more aggressive signals, which starts with singing a<br />

shared song, precedes to type matching, then to staying on the match, soft song,<br />

and finally attack (Beecher and Campbell, 2005; Searcy and Beecher, 2009).<br />

Because matching is low in this hierarchy of escalation, with several steps<br />

intervening between it and attack, matching would not be expected to be very<br />

informative about attack likelihood; nevertheless, it might still be predictive of<br />

the next level of escalation. Whether matching is predictive in this manner<br />

requires further testing.<br />

Among the small number of songbird species that have been studied in<br />

this regard, soft song has emerged as an unusually reliable predictor of attack.<br />

Why a display whose distinguishing characteristic is low amplitude should be<br />

consistently favored for the highest level of aggressive signaling is not well<br />

understood. One hypothesis is that by using soft song during an encounter with<br />

an intruder, a territory owner lowers the chance of interference from other rival<br />

males by preventing them from eavesdropping on the interaction (McGregor and<br />

Dabelsteen, 1996), thereby concealing from them that an intrusion is taking<br />

place. In contradiction to this idea, Searcy and Nowicki (2006) found that, in<br />

song sparrows, more intrusions by third party males occurred during simulated<br />

interactions between an owner giving soft songs and an intruder giving loud<br />

songs than during interactions in which both owner and intruder gave loud<br />

songs. In other words, use of soft songs if anything increased interference by<br />

other rivals. A second hypothesis is that soft song is favored as an aggressive<br />

signal because its low amplitude makes its target unambiguous: only the male<br />

that is being confronted can discern the signal, so only he can be the target.<br />

Another way of stating this is that soft song is a performance signal subject to an<br />

informational constraint (Hurd and Enquist, 2005) that forces it to be honest at<br />

least with respect to the identity of its target.<br />

If a display is a reliable signal of aggressive intentions, as is soft song,<br />

then theory predicts that it should be effective in changing the behavior of at<br />

least some opponents to the signaler’s advantage (Enquist, 1985). In other words,<br />

a believable threat should intimidate some opponents, presumably the weaker<br />

ones, causing them to concede whatever resource is being contested. Effectiveness<br />

in this sense has not yet been demonstrated for soft song, in part because<br />

arranging tests of the effectiveness of displays in territorial defense is quite<br />

difficult (Searcy and Nowicki, 2000). Recent work with corn crakes (Crex<br />

crex), which are not songbirds and do not sing, shows that low amplitude calls


3. Signaling Aggression 37<br />

predict attack, and suggests that these soft calls cause some receivers to retreat<br />

(Rek and Osiejuk, 2011). Effectiveness in intimidating opponents has been<br />

demonstrated in some other aggressive signaling systems (Dingle, 1969; Fugle<br />

et al., 1984; Wagner, 1992).<br />

III. VISUAL DISPLAYS SIGNAL AGGRESSIVE INTENT IN<br />

CEPHALOPODS: THE SWEET SMELL OF SUCCESS<br />

Cephalopods—squid, octopus, and cuttlefish—are marine molluscs with large<br />

complex brains and highly diverse behavior (Hanlon and Messenger, 1996).<br />

They are highly visual animals, exemplified partly by their huge optic lobes<br />

that represent more than half of their central nervous system. These soft-bodied<br />

cephalopods are renowned for their rapid adaptive coloration: individuals of each<br />

species can instantly (


38 van Staaden et al.<br />

Early game theory models of agonistic behavior predicted that animals<br />

should not signal their probability of attack to their opponents. As Maynard<br />

Smith (1982) argued, if animals signaled their aggressive motivation during a<br />

fight, there would be strong selective pressure for animals to “bluff” and to signal<br />

the highest motivational state possible; such a system would likely be invaded by<br />

cheaters and become unreliable. However, some animals do signal intent<br />

(Hauser and Nelson, 1991), and below we provide an unusual example of this<br />

in cuttlefish.<br />

As in birds, cephalopods signal aggressive intent but they do so with<br />

visual signals (chromatic skin patterns) as well as body postures (parallel positioning<br />

and arm postures). Two examples are given: one from cuttlefish (Order<br />

Sepioidea) and one from squid (Order Teuthoidea). In addition, a new finding is<br />

described in which a molecular trigger of aggression has been found in squid.<br />

A. Cuttlefish agonistic bouts<br />

In the Intense Zebra Display of the European cuttlefish, Sepia officinalis, the males<br />

turn on high-contrast stripes and dark eye ring and extend their large 4th arm<br />

toward the opponent (Fig. 3.3C). Such agonistic encounters between males can<br />

lead to aggressive grappling and biting. The experiments of Adamo and Hanlon<br />

(1996) showed that one visual component of the display—the facial darkness—<br />

was by far the most highly variable in expression, and was a good predictor of<br />

outcome in encounters in which one male withdrew. In non-escalated encounters,<br />

the male that ultimately withdrew always maintained a less dark face than<br />

its opponent (Fig. 3.3A). When the face of a displaying cuttlefish became lighter,<br />

the other male either remained in the Intense Zebra Display but did not<br />

approach closely or lightened the intensity of its own display within 15 s.<br />

When both males maintained a dark face, the agonistic encounters usually<br />

escalated to physical pushing, and sometimes to grappling and biting (Fig. 3.3B).<br />

Why would males show an agonistic display to a rival male but simultaneously<br />

signal their intent not to be aggressive Adamo and Hanlon (1996)<br />

pointed out that sexual recognition in cephalopods is poorly developed, and that<br />

the Intense Zebra Display (with 4th arm extended) identifies the signaler as a<br />

male. The authors suggest that male cuttlefish that are not prepared to attack an<br />

opponent still give the modified (i.e., light-faced) Intense Zebra Display to<br />

convey two messages: (1) that it is male, but (2) it is not prepared to escalate<br />

to aggressive physical contact. As the authors point out, when agonistic displays<br />

perform more than one function, signaling intent (i.e., signaling its likely<br />

subsequent behavior) can be an ESS. Unless the fight escalated to grappling<br />

and biting, there would be little cost to cheaters in this system since males that


3. Signaling Aggression 39<br />

A<br />

Darkness of face (pixel darkness)<br />

220<br />

210<br />

200<br />

190<br />

180<br />

170<br />

160<br />

4<br />

Non-escalated encounter<br />

6 8 10 12 14 16<br />

C<br />

B<br />

Darkness of face (pixel darkness)<br />

220<br />

210<br />

200<br />

190<br />

180<br />

170<br />

160<br />

4<br />

Escalated encounter<br />

6 8 10 12 14 16<br />

Time (s)<br />

D<br />

Percentage of losing males<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Intense<br />

Zebra<br />

Extended<br />

4th arm<br />

Winners<br />

Losers<br />

First encounter<br />

Second encounter (no copulation)<br />

Second encounter (after copulation)<br />

Figure 3.3. Cuttlefish signal intent to escalate a fight with a dark face component to their Intense<br />

Zebra Display. (A, B) Differences in facial darkness during a non-escalated versus<br />

escalated encounter. (C) Two males in Intense Zebra Display with different degrees of<br />

facial darkness. (D) When males that lost a fight copulated with a female, they became<br />

more aggressive in the successive fight. From Adamo and Hanlon (1996).<br />

bluffed (i.e., gave a dark-faced Intense Zebra Display but had little fighting<br />

motivation and/or ability) could withdraw at the next stage of agonistic behavior<br />

with little penalty.<br />

In the same study, the authors allowed losing males to copulate with a<br />

female after a bout, and retested them with the male each had lost to. The former<br />

losers increased facial darkness dramatically in those encounters, showed a longlasting<br />

Intense Zebra Display, and did not withdraw from an opponent<br />

(Fig. 3.3D), thus supporting the contention that facial darkness signals the<br />

animal’s motivational state (i.e., tendency to attack).


40 van Staaden et al.<br />

B. Squid agonistic bouts<br />

Male–male fights in Loligo plei are complex visual displays that include up to 21<br />

behaviors. There is a hierarchy of agonistic signals that sometimes culminates in<br />

an aggressive physical lateral display and fin beating (Fig. 3.4A and B), which are<br />

then followed by chase or flee. DiMarco and Hanlon (1997) tested whether<br />

dominance was based upon the duration or frequency of these behaviors, but it<br />

was not. Instead, they found that certain visual features such as the lateral flame<br />

markings (Fig. 3.4B, top squid) could be expressed with high contrast and that<br />

this was a visual factor in escalation of the agonistic bout.<br />

Two distinct tactics were exhibited by fighting males in this set of<br />

laboratory experiments: (1) long bouts with slow escalation from visual signaling<br />

to chasing and fleeing, or (2) short bouts with very rapid escalation from visual<br />

signaling to lateral displaying, aggressive physical fin beating, followed by chasing<br />

and fleeing (Fig. 3.4C). It is noteworthy that the second tactic occurred when<br />

a female was present (i.e., when a potential resource value was present). As shown<br />

in Fig. 3.4D, the presence of a female in various combinations had a dramatic<br />

effect on the nature and duration of the agonistic interactions. Longest bouts<br />

(mean 14 min) occurred when only two males were present. Bouts became<br />

progressively shorter when either two males and one female were assembled<br />

simultaneously, or two males were interacting and a female was then added<br />

(mean 9 and 3 min, respectively). But when a male and female were put in a<br />

tank and allowed to pair, and then a nonpaired male was added, tactic 2 was used<br />

and the highly aggressive interaction lasted only 30 s (a 28 difference over the<br />

simple two male scenario). As a control, when females were added to male/<br />

female pairs, there were no agonistic interactions (Fig. 3.4D).<br />

In this squid species, the lateral display represents an escalation of<br />

aggression because it involves parallel posturing and the simultaneous expression<br />

of many high-contrast visual signals, which collectively give the impression of<br />

making the squid look larger (e.g., the mid-ventral ridge of the mantle protrudes<br />

vertically as in the dewlap extension of geckos). Fin beating is a physical, robust<br />

contest of pushing that can transmit information about strength and size of the<br />

competing individuals.<br />

C. From molecules to aggression: Contact pheromone triggers strong<br />

aggression in squid<br />

In the squid Loligo pealei, which conducts visual agonistic bouts similar to L. plei<br />

(above), it was found recently that females deposit a contact pheromone in the<br />

outer tunic of egg capsules that they lay on the sea floor. When males see the egg<br />

capsules (even in the absence of females), they are visually attracted to them and<br />

then physically contact the eggs, which leads to extremely aggressive fighting


A<br />

B<br />

Escalation<br />

Miscellaneous visual signaling<br />

Visual signaling and<br />

lateral display<br />

First<br />

parallel positioning<br />

First<br />

fin beating<br />

Lateral display and<br />

fin beating<br />

Last<br />

fin beating<br />

Chase and flee<br />

Contest duration<br />

C<br />

Mean contest duration (min)<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Misc. visual signaling<br />

Chase and flee<br />

Misc. visual signaling<br />

Lateral display<br />

Fin beating<br />

Chase and flee<br />

Alternative fighting tactics<br />

D<br />

Average contest duration (min)<br />

20<br />

18<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

/ +/ /+ /+/ /<br />

Combination<br />

Figure 3.4. Male squid (Loligo plei) use complex lateral displays to conduct shorter fights with higher aggression when a resource value (i.e., a female mate) is<br />

present. (A) Schema depicting the escalation pathway to a typical <strong>full</strong>y escalated agonistic contest. (B) Photograph of a <strong>full</strong>y deployed lateral<br />

display and fin beating. The display comprises six visual components: arm spots, dorsal arm iridophores, stitchwork fins, mid-ventral ridge,<br />

tentacular stripe, and lateral flame. (C) Short fights are more complex and involve lateral displays and physical fin beating. (D) The shortest, most<br />

dramatic fights occur when a mating pair is already formed, and a rival male is introduced to the tank (♂♀þ♂).


42 van Staaden et al.<br />

2. Physical<br />

contact<br />

3. Extreme<br />

aggression<br />

1. Visual<br />

attraction<br />

Aggressin<br />

Human b-microseminoprotein<br />

N<br />

N<br />

s s sss<br />

ss s s s<br />

s s sss<br />

ss s s s<br />

Figure 3.5. When male squids see egg capsules on the sea floor, then approach and touch them, this<br />

leads to immediate and dramatic change from calm swimming to extreme fighting. The<br />

contact pheromone (“aggressin” or Loligo b-MSP) is in the tunic of the egg capsules and<br />

is similar in structure to that found in humans, mice, and other vertebrates.<br />

C<br />

C<br />

within a minute or two (Fig. 3.5) (Cummins et al., 2011). Thus, there is a twostep<br />

sensory process: visual attraction to eggs followed by contact chemoreception<br />

that induces onset of aggression.<br />

In controlled experiments, the 10 kDa protein pheromone (termed<br />

Loligo b-microseminoprotein, b-MSP) was isolated and coated onto a clear<br />

glass flask containing egg capsules, and males that touched the glass (but not<br />

the eggs) began to signal, fight, and bite each other violently within seconds.<br />

Glass flasks without the pheromone coating failed to elicit those aggressive<br />

behaviors. Thus, direct contact with the protein molecules immediately led to<br />

the <strong>full</strong> cascade of complex aggressive fighting in the absence of females. Given<br />

that aggression is often considered to be a result of multiple interactions of<br />

physiology, hormones, sensory stimuli, etc., this finding reminds us that perhaps<br />

in some cases there are straightforward pathways to aggression. In fact, the<br />

proximate mechanisms that trigger or strengthen aggression are not well<br />

known for many taxa (Wingfield et al., 2005).<br />

There is a noteworthy vertebrate/mammalian connection to this<br />

finding. As shown in Fig. 3.6, the b-MSPs are highly conserved throughout the<br />

animal kingdom. The greatest known concentration of b-MSPs is in human and<br />

rodent seminal fluid, yet regrettably the functions of b-MSPs are unknown in any<br />

taxa except cephalopods, as explained above (Cummins et al., 2011). As those<br />

authors suggest, it would be worthwhile to look for an aggression function for


3. Signaling Aggression 43<br />

0.1<br />

Human (Homo sapien)<br />

Other primate (consensus)<br />

Pig (Sus scrofa)<br />

Rat (Rattus norvegicus)<br />

Mouse (Mus musculus)<br />

Red jungle fowl (Gallus gallus)<br />

Zebra finch (Taeniopygia guttata)<br />

Ostrich (Struthio camelus)<br />

Northern pike (Esox lucius)<br />

Atlantic salmon (Salmo salar)<br />

Zebrafish (Danio rerio)<br />

Lancet (Branchiostoma belcheri)<br />

California mussel (Mytilus californianus)<br />

Pacific oyster (Crassostrea gigas)<br />

Bay scallop (Argopecten irradians)<br />

Limpet (Lottia gigantea)<br />

Longfin squid (Loligo pealeii)<br />

Abalone (Haliotis diversicolor)<br />

Bobtail squid (Euprymna scolopes)<br />

Rotifer (Adineta vaga)<br />

Human (Homo sapien) CRISP<br />

Mouse (Mus musculus) CRISP<br />

Snake (Rhinoplocephalus nigrescens) CRISP<br />

Frog (Xenopus laevis) CRISP<br />

Phylum Chordata Phylum Mollusca Phylum Rotifera<br />

Figure 3.6. Evolutionary origins and conservation of b-microseminoproteins. The tree shows<br />

phylogenetic relationships among the protein sequences. The b-MSPs identified in<br />

the Cummins et al. (2011) study are within the dotted lines.<br />

b-MSPs in mammals and other vertebrates, given the molecular similarity and<br />

unique structure of these proteins, all of which seem to be most concentrated in<br />

exocrine glands in many taxa. Such findings remind us that multisensory cues are<br />

often involved in stimulating behaviors and that a good deal more research is<br />

needed before we understand subjects such as aggression.<br />

D. Signaling aggression in humans<br />

In humans, as in other species, signaler and receiver have both evolved to use<br />

variation in aggressive signal structure to their own advantage. In the case of<br />

human speech, fundamental vocal frequency is perceived to be associated with<br />

social cues for dominance and submissiveness (Bolinger, 1978; Huron et al.,2009;<br />

Ohala, 1994), with vocal pitch height used to signal aggression (low pitch), or


44 van Staaden et al.<br />

appeasement (high pitch). Moreover, a strong correlation with eyebrow position<br />

suggests an intermodal linkage between vocal and facial expressions (Huron et al.,<br />

2009). Evidence implicates male dominance competition (Puts et al.,2006), rather<br />

than intersexual selection (see Chapter 2), as the selective origin of this performance<br />

signal. Similarly, handgrip strength is correlated with level of aggression<br />

and appears to be an honest signal for quality in males (Gallup et al., 2007).<br />

Mathematical models show, however, that the tradeoff of deceptive efficacy and<br />

dishonest signals of intent often favors signalers who produce imperfectly deceptive<br />

signals over perfectly honest or perfectly deceptive ones (Andrews, 2002).<br />

Competition among coalition groups (a characteristic shared with chimpanzees)<br />

initiated a social arms race, culminating in extraordinary human cognitive abilities<br />

(Flinn et al.,2005), capable of parsing aggressive signals (Paul and Thelen, 1983),<br />

and competitive displays (Hawkes and Bird, 2002). This great capacity for signaling<br />

is outstripped only by the uniquely human ability to extend our phenotype<br />

with weaponry—with the unfortunate consequence that our potential to inflict<br />

damage frequently exceeds our ability to control aggression.<br />

Rather than maximizing its absolute amount, natural selection enhances<br />

the overall effectiveness of aggression. In invertebrates, where individuals generally<br />

pursue a solitary existence, physical superiority primarily determines the eventual<br />

outcome of contests, and most fights are quickly resolved on the basis of prominent<br />

asymmetries in body or weapon size. In vertebrates, which must navigate the<br />

demands and opportunities of social living, aggressive success is largely contingent<br />

on the development of social competence. In this case, natural selection favors<br />

those with an ability to effectively anticipate their chances well in advance of a<br />

contest, and to signal strength while hiding any intentions to eventually withdraw.<br />

Generating and interpreting aggressive signals to form successful alliances and to<br />

inherit status from high-ranking kin, is thus key to winning both short-term<br />

contests and long-term evolutionary success.<br />

Acknowledgments<br />

Production of this chapter was partially supported by funding from NSF grant DUE-0757001 (to M. v. S.).<br />

W. A. S. thanks his collaborators on the sparrow signaling research including Steve Nowicki, Rindy<br />

Anderson, Barb Ballentine, Mike Beecher, and Susan Peters. R. T. H. is grateful for partial funding from<br />

NSF grant IBN-0415519 and many wonderful colleagues who participated in these experiments and field<br />

observations.<br />

References<br />

Adamo, S. A., and Hanlon, R. T. (1996). Do cuttlefish (Cephalopoda) signal their intentions to<br />

conspecifics during agonistic encounters Anim. Behav. 52, 73–81.<br />

Adams, E. S., and Mesterton-Gibbons, M. (1995). The cost of threat displays and the stability of<br />

deceptive communication. J. Theor. Biol. 175, 405–421.


3. Signaling Aggression 45<br />

Alexander, R. D. (1961). Aggressiveness, territoriality, and sexual behavior in field crickets<br />

(Orthoptera: Gryllidae). Behaviour 17, 130–223.<br />

Anderson, R. C., Searcy, W. A., and Nowicki, S. (2005). Partial song matching in an eastern<br />

population of song sparrows, Melospiza melodia. Anim. Behav. 69, 189–196.<br />

Anderson, R. C., Searcy, W. A., Peters, S., and Nowicki, S. (2008). Soft song in song sparrows:<br />

Acoustic structure and implications for signal function. Ethology 114, 662–676.<br />

Andersson, M. (1980). Why are there so many threat displays J. Theor. Biol. 86, 773–781.<br />

Andrews, P. W. (2002). The influence of postreliance detection on the deceptive efficacy of<br />

dishonest signals of intent: Understanding facial clues to deceit as the outcome of signaling<br />

tradeoffs. Evol. Hum. Behav. 23, 103–121.<br />

Arak, A., and Enquist, M. (1995). Conflict, receiver bias and the evolution of signal form.<br />

Phil. Trans. R. Soc. Lond. B 349, 337–344.<br />

Arnott, G., and Elwood, R. W. (2010). Signal residuals and hermit crab displays: Flaunt it if you have<br />

it! Anim. Behav. 79, 137–143.<br />

Ballentine, B., Searcy, W. A., and Nowicki, S. (2008). Reliable aggressive signalling in swamp<br />

sparrows. Anim. Behav. 75, 693–703.<br />

Bee, M. A., Perrill, S. A., and Owen, P. C. (1999). Size assessment in simulated territorial encounters<br />

between male green frogs (Rana clamitans). Behav. Ecol. Sociobiol. 45, 177–184.<br />

Beebee, M. D. (2004). The function of multiple singing modes: Experimental tests in yellow warblers,<br />

Dendroica petechia. Anim. Behav. 67, 1089–1097.<br />

Beecher, M. D., and Campbell, S. E. (2005). The role of unshared songs in singing interactions<br />

between neighbouring song sparrows. Anim. Behav. 70, 1297–1304.<br />

Bolinger, D. L. (1978). Intonation across languages. In “Universals of Human Language”, Vol. 2:<br />

Phonology (J. H. <strong>Green</strong>berg, C. A. Ferguson, and E. A. Moravcsik, eds.), pp. 471–524. Stanford<br />

<strong>University</strong> Press, Palo Alto, CA.<br />

Borror, D. J. (1965). Song variation in Maine song sparrows. Wilson Bull. 77, 5–37.<br />

Brandt, Y. (2002). Lizard threat display handicaps endurance. Proc. R. Soc. Lond. B 270, 1061–1068.<br />

Breithaupt, T., and Eger, P. (2002). Urine makes the difference: Chemical communication in<br />

fighting crayfish made visible. J. Exp. Biol. 205, 1221–1231.<br />

Burt, J. M., Bard, S. C., Campbell, S. E., and Beecher, M. D. (2002). Alternative forms of song<br />

matching in song sparrows. Anim. Behav. 63, 1143–1151.<br />

Byers, B. E. (1996). Messages encoded in the songs of chestnut-sided warblers. Anim. Behav. 52,<br />

691–705.<br />

Caryl, P. G. (1979). Communication by agonistic displays: What can games theory contribute to<br />

ethology Behaviour 68, 136–169.<br />

Catchpole, C. K., and Slater, P. J. B. (2008). Bird Song: Biological Themes and Variations.<br />

Cambridge <strong>University</strong> Press, Cambridge.<br />

Chen, S., Lee, A. Y., Bowens, N. M., Huber, R., and Kravitz, E. A. (2002). Fighting fruit flies:<br />

A model system for the study of aggression. Proc. Natl. Acad. Sci. USA 99, 5664–5668.<br />

Cheney, D., Seyfarth, R., and Smuts, B. (1986). Social relationships and social cognition in<br />

nonhuman primates. Science 234, 1361–1366.<br />

Clark, D. C., and Moore, A. J. (1995). Genetic aspects of communication during male-male<br />

competition in the Madagascar hissing cockroach: Honest signalling of size. Heredity 75, 198–205.<br />

Cummins, S. F., Boal, J. G., Buresch, K. C., Kuanpradit, C., Sobhon, P., Holm, J. B., Degnan, B. M.,<br />

Nagle, G. T., and Hanlon, R. T. (2011). Extreme aggression in male squid induced by a<br />

b-MSP-like pheromone. Curr. Biol. 21, 322–327.<br />

Dabelsteen, T., McGregor, P. K., Lampe, H. M., Langmore, N. E., and Holland, J. (1998). Quiet song<br />

in song birds: An overlooked phenomenon. Bioacoustics 9, 89–105.<br />

Darwin, C. (1871). The Descent of Man and Selection in Relation to Sex. Murray, London.


46 van Staaden et al.<br />

DiMarco, F. P., and Hanlon, R. T. (1997). Agonistic behavior in the squid Loligo plei (Loliginidae,<br />

Teuthoidea): Fighting tactics and the effects of size and resource value. Ethology 103, 89–108.<br />

Dingle, H. (1969). A statistical and information analysis of aggressive communication in the mantis<br />

shrimp Gonodactylus bredini Manning. Anim. Behav. 17, 561–575.<br />

Enquist, M. (1985). Communication during aggressive interactions with particular reference to<br />

variation in choice of behaviour. Anim. Behav. 33, 1152–1161.<br />

Enquist, M., and Jakobsson, S. (1986). Decision making and assessment in the fighting behaviour of<br />

Nannacara anomala (Cichlidae, Pisces). Ethology 72, 143–153.<br />

Enquist, M., and Leimar, O. (1983). Evolution of fighting behaviour: Decision rules and assessment of<br />

relative strength. J. Theor. Biol. 102, 387–410.<br />

Flinn, M. V., Geary, D. C., and Ward, C. V. (2005). Ecological dominance, social competition, and<br />

coalitionary arms races: Why humans evolved extraordinary intelligence. Evol. Hum. Behav. 26,<br />

10–46.<br />

Fugle, G. N., Rothstein, S. I., Osenberg, C. W., and McGinley, M. A. (1984). Signals of status in<br />

wintering white-crowned sparrows, Zonotrichia leucophrys gambelii. Anim. Behav. 32, 86–93.<br />

Gallup, A. G., White, D. D., and Gallup, G. G. (2007). Handgrip strength predicts sexual behavior,<br />

body morphology, and aggression in male college students. Evol. Hum. Behav. 28, 423–429.<br />

Grafen, A. (1990). Biological signals as handicaps. J. Theor. Biol. 144, 517–546.<br />

<strong>Green</strong>field, M. D., and Minckley, R. L. (1993). Acoustic dueling in tarbush grasshoppers: Settlement<br />

of territorial contests via alternation of reliable signals. Ethology 95, 309–326.<br />

Gross, M. R. (1996). Alternative reproductive strategies and tactics: Diversity within sexes. Trends<br />

Ecol. Evol. 11, 92–98.<br />

Guilford, T., and Dawkins, M. S. (1995). What are conventional signals Anim. Behav. 49,<br />

1689–1695.<br />

Hall, K. C., and Hanlon, R. T. (2002). Principal features of the mating system of a large spawning<br />

aggregation of the giant Australian cuttlefish Sepia apama (Mollusca: Cephalopoda). Mar. Biol.<br />

140, 533–545.<br />

Hamblin, S., and Hurd, P. L. (2007). Genetic algorithms and non-ESS solutions to game theory<br />

models. Anim. Behav. 74, 1005–1018.<br />

Hanlon, R. T., and Messenger, J. B. (1996). Cephalopod Behaviour. Cambridge <strong>University</strong> Press,<br />

Cambridge.<br />

Hanlon, R. T., Maxwell, M. R., Shashar, N., Loew, E. R., and Boyle, K. L. (1999). An ethogram of<br />

body patterning behavior in the biomedically and commercially valuable squid Loligo pealei off<br />

Cape Cod, Massachusetts. Biol. Bull. 197, 49–62.<br />

Hanlon, R. T., Smale, M. J., and Sauer, W. H. H. (2002). The mating system of the squid Loligo<br />

vulgaris reynaudii (Cephalopoda, Mollusca) off South Africa: Fighting, guarding, sneaking, mating<br />

and egg laying behavior. Bull. Mar. Sci. 71, 331–345.<br />

Hanlon, R. T., Naud, M. J., Shaw, P. W., and Havenhand, J. N. (2005). Behavioural ecology:<br />

Transient sexual mimicry leads to fertilization. Nature 430, 212.<br />

Hauser, M. D., and Nelson, D. A. (1991). ‘Intentional’ signalling in animal communication. Trends<br />

Ecol. Evol. 6, 186–189.<br />

Hawkes, K., and Bird, R. B. (2002). Showing off, handicap signaling, and the evolution of men’s<br />

work. Evol. Anthropol. 11, 58–67.<br />

Hof, D., and Hazlett, N. (2010). Low-amplitude song predicts attack in a North American wood<br />

warbler. Anim. Behav. 80, 821–828.<br />

Houston, A. I., and McNamara, J. M. (1999). Models of Adaptive Behaviour, an Approach Based on<br />

<strong>State</strong>. Cambridge <strong>University</strong> Press, Cambridge.<br />

Huber, R., and Kravitz, E. A. (1995). A quantitative study of agonistic behavior and dominance in<br />

juvenile American lobsters (Homarus americanus). Brain Behav. Evol. 46, 72–83.


3. Signaling Aggression 47<br />

Hughes, M. (2000). Deception with honest signals: Signal function for signalers and receivers. Behav.<br />

Ecol. 6, 614–623.<br />

Hurd, P. L., and Enquist, M. (2005). A strategic taxonomy of biological communication. Anim.<br />

Behav. 70, 1155–1170.<br />

Huron, D., Dahl, S., and Johnson, R. (2009). Facial expression and vocal pitch height: Evidence of an<br />

intermodal association. EMR. 4, 93–100.<br />

Jantzen, T. M., and Havenhand, J. N. (2003). Reproductive behavior in the squid Sepioteuthis australis<br />

from South Australia: Interactions on the spawning grounds. Biol. Bull. 204, 305–317.<br />

Jonsson, T., Kravitz, E. A., and Heinrich, R. (2011). Sound production during agonistic behavior of<br />

male Drosophila melanogaster. Fly (Austin) 5, 29–38.<br />

Kim, Y.-G. (1995). Status signalling games in animal contests. J. Theor. Biol. 176, 221–231.<br />

Kramer, H. G., and Lemon, R. E. (1983). Dynamics of territorial singing between neighboring song<br />

sparrows (Melospiza melodia). Behaviour 85, 198–223.<br />

Kramer, H. G., Lemon, R. E., and Morris, M. J. (1985). Song switching and agonistic stimulation in<br />

the song sparrow (Melospiza melodia): Five tests. Anim. Behav. 33, 135–149.<br />

Laidre, M. E., and Vehrencamp, S. L. (2008). Is bird song a reliable signal of aggressive intent Behav.<br />

Ecol. Sociobiol. 62, 1207–1211.<br />

Lailvaux, S. P., Reaney, L. T., and Backwell, P. R. Y. (2009). Dishonesty signalling of fighting<br />

ability and multiple performance traits in the fiddler crab Uca mjoebergi. Funct. Ecol. 23,<br />

359–366.<br />

Leimar, O., and Enquist, M. (1984). Effects of asymmetries in owner-intruder conflicts. J. Theor. Biol.<br />

111, 475–491.<br />

Logue, D. M., Abiola, I., Raines, D., Bailey, N., Zuk, M., and Cade, W. H. (2010). Does signaling<br />

mitigate the cost of agonistic interactions A test in a cricket that has lost its song. Proc. R. Soc.<br />

Lond. B 277, 2571–2575.<br />

Lorenz, K. (1963). Das sogenannte Böse. Methuen Publishing, London.<br />

Lorenz, K. (1978). Vergleichende Verhaltensforschung Grundlagen der Ethologie. Springer-Verlag,<br />

Wien.<br />

Lorenz, K. (1995). The Natural Science of the Human Species: An Introduction to Comparative<br />

Behavioral Research—The Russian Manuscript (1944–1948). MIT Press, Cambridge, MA.<br />

Lundrigan, B. (1996). Morphology of horns and fighting behavior in the family Bovidae. J. Mammal.<br />

77, 462–475.<br />

Maynard Smith, J. (1974). The theory of games and the evolution of animal conflicts. J. Theor. Biol.<br />

47, 209–221.<br />

Maynard Smith, J. (1979). Game theory and the evolution of behaviour. Proc. R. Soc. Lond. B 205,<br />

475–488.<br />

Maynard Smith, J. (1982). Do animals convey information about their intentions J. Theor. Biol. 97,<br />

1–5.<br />

Maynard Smith, J., and Harper, D. (1988). The evolution of aggression: Can selection generate<br />

variability Phil. Trans. R. Soc. Lond. B 319, 557–570.<br />

Maynard Smith, J., and Harper, D. (2003). Animal Signals. Oxford <strong>University</strong> Press, Oxford.<br />

Maynard Smith, J., and Price, G. R. (1973). The logic of animal conflict. Nature 246, 15–18.<br />

McGregor, P. K., and Dabelsteen, T. (1996). Communication networks. In “Ecology and Evolution of<br />

Acoustic Communication in Birds” (D. E. Kroodsma and E. H. Miller, eds.), pp. 409–425. Cornell<br />

<strong>University</strong> Press, Ithaca, NY.<br />

Miczek, K. A., De Almeida, R. M. M., Kravitz, E. A., Rissman, E. F., De Boer, S. F., and Raine, A.<br />

(2007). Neurobiology of escalated aggression and violence. J. Neurosci. 27, 11803–11806.<br />

Molles, L. E., and Vehrencamp, S. L. (1999). Repertoire size, repertoire overlap, and singing modes in<br />

the Banded Wren (Thryothorus pleurostictus). Auk 116, 667–689.


48 van Staaden et al.<br />

Molles, L. E., and Vehrencamp, S. L. (2001). Songbird cheaters pay a retaliation cost: Evidence for<br />

auditory conventional signals. Proc. R. Soc. Lond. B 268, 2013–2019.<br />

Moore, J. C., Obbard, D. J., Reuter, C., West, S. A., and Cook, J. M. (2009). Male morphology and<br />

dishonest signalling in a fig wasp. Anim. Behav. 78, 147–153.<br />

Moretz, J. A., and Morris, M. R. (2003). Evolutionarily labile responses to a signal of aggressive<br />

intent. Proc. R. Soc. Lond. B 270, 2271–2277.<br />

Mowbray, T. B. (1997). Swamp sparrow (Melospiza georgiana). In “The Birds of North America”<br />

(A. Poole and F. Gill, eds.), No. 509. Academy of Natural Sciences, Philadelphia.<br />

Mulligan, J. A. (1963). A Description of Song Sparrow Song Based on Instrumental Analysis<br />

Proceedings of the XIII International. Ornithological Congress. pp. 272–284.<br />

Murphy, T. G. (2008). Display of an inedible prop as a signal of aggression Adaptive significance of<br />

leaf-display by the turquoise-browed motmot, Eumomota superciliosa. Ethology 114, 16–21.<br />

Natarajan, D., de Vries, H., Saaltink, D. J., de Boer, S. F., and Koolhaas, J. M. (2009). Delineation of<br />

violence from functional aggression in mice: An ethological approach. Behav. Genet. 39, 73–90.<br />

Nice, M. M. (1943). Studies in the life history of the song sparrow. II. The behavior of the song<br />

sparrow and other passerines. Trans. Linn. Soc. N.Y. 6, 1–328.<br />

Noble, J. (2000). Talk is cheap: Evolved strategies for communication and action in asymmetrical<br />

animal contests. In “SAB00” (J.-A. Meyer, A. Berthoz, D. Floreano, H. Roitblat, and S. Wilson,<br />

eds.), pp. 481–490. MIT Press, Massachusetts.<br />

Nowicki, S., Podos, J., and Valdes, F. (1994). Temporal patterning of within-song type and betweensong<br />

type variation in song repertoires. Behav. Ecol. Sociobiol. 34, 329–335.<br />

Ohala, J. (1994). The frequency code underlies the sound-symbolic use of voice pitch. In “Sound<br />

Symbolism” (L. Hinton, J. Nichols, and J. Ohala, eds.), pp. 325–347. Cambridge <strong>University</strong> Press,<br />

Cambridge.<br />

Page, R. B., and Jaeger, R. G. (2004). Multimodal signals, imperfect information, and identification<br />

of sex in red-backed salamanders (Plethodon cinereus). Behav. Ecol. Sociobiol. 56, 132–139.<br />

Paul, S. C., and Thelen, M. H. (1983). The use of strategies and messages to alter aggressive<br />

interactions. Aggress. Behav. 9, 183–193.<br />

Peters, S., Searcy, W. A., Beecher, M. D., and Nowicki, S. (2000). Geographic variation in the<br />

organization of song sparrow repertoires. Auk 117, 936–942.<br />

Puts, D. A., Gaulin, S. J. C., and Verdolini, K. (2006). Dominance and the evolution of sexual<br />

dimorphism in human voice pitch. Evol. Hum. Behav. 27, 283–296.<br />

Raffinger, E., and Ladich, F. (2009). Acoustic threat displays and agonistic behaviour in the<br />

red-finned loach Yasuhikotakia modesta. J. Ethol. 27, 239–247.<br />

Reby, D., McComb, K., Cargnelutti, B., Darwin, C., Fitch, W. T., and Clutton-Brock, T. (2005). Red<br />

deer stags use formants as assessment cues during intrasexual agonistic interactions. Proc. R. Soc.<br />

Lond. B 272, 941–947.<br />

Rek, P., and Osiejuk, T. S. (2011). Nonpasserine bird produces soft calls and pays retaliation cost.<br />

Behav. Ecol. 22, 657–662.<br />

Saunders, A. A. (1924). Recognizing individual birds by song. Auk 41, 242–259.<br />

Scott, J. L., et al. (2010). The evolutionary origins of ritualized acoustic signals in caterpillars.<br />

Nat. Commun. 1, 4. doi: 10.1038/ncomms1002.<br />

Searcy, W. A., and Andersson, M. (1986). Sexual selection and the evolution of song. Annu. Rev.<br />

Ecol. Syst. 17, 507–533.<br />

Searcy, W. A., and Beecher, M. D. (2009). Song as an aggressive signal in songbirds. Anim. Behav.<br />

78, 1281–1292.<br />

Searcy, W. A., and Nowicki, S. (2000). Male-male competition and female choice in the evolution<br />

of vocal signaling. In “Animal Signals: Signalling and Signal Design in Animal Communication”<br />

(Y. Espmark, T. Amundsen, and G. Rosenqvist, eds.), pp. 301–315. Tapir Academic Press,<br />

Trondheim.


3. Signaling Aggression 49<br />

Searcy, W. A., and Nowicki, S. (2006). Signal interception and the use of soft song in aggressive<br />

interactions. Ethology 112, 865–872.<br />

Searcy, W. A., and Yasukawa, K. (1990). Use of the song repertoire in intersexual and intrasexual<br />

con<strong>text</strong>s by male red-winged blackbirds. Behav. Ecol. Sociobiol. 27, 123–128.<br />

Searcy, W. A., Podos, J., Peters, S., and Nowicki, S. (1995). Discrimination of song types and variants<br />

in song sparrows. Anim. Behav. 49, 1219–1226.<br />

Searcy, W. A., Nowicki, S., and Hogan, C. (2000). Song type variants and aggressive con<strong>text</strong>. Behav.<br />

Ecol. Sociobiol. 48, 358–363.<br />

Searcy, W. A., Anderson, R. C., and Nowicki, S. (2006). Bird song as a signal of aggressive intent.<br />

Behav. Ecol. Sociobiol. 60, 234–241.<br />

Simmonds, L. W., and Bailey, W. J. (1993). Agonistic communication between males of a zaprochiline<br />

katydid (Orthoptera: Tettigoniidae). Behav. Ecol. 4, 364–368.<br />

Skyrms, B. (2009). Evolution of signalling systems with multiple senders and receivers. Phil. Trans.<br />

R. Soc. B 364, 771–779.<br />

Spector, D. A. (1992). Wood-warbler song systems: A review of paruline song systems. Curr.<br />

Ornithol. 9, 199–239.<br />

Stoddard, P. K., Beecher, M. D., and Willis, M. S. (1988). Response of territorial male song sparrows<br />

to song types and variations. Behav. Ecol. Sociobiol. 22, 125–130.<br />

Stoddard, P. K., Beecher, M. D., Horning, C. L., and Willis, M. S. (1990). Strong neighbor-stranger<br />

discrimination in song sparrows. Condor 92, 1051–1056.<br />

Stoddard, P. K., Beecher, M. D., Campbell, S. E., and Horning, C. L. (1992). Song-type matching in<br />

the song sparrow. Can. J. Zool. 70, 1440–1444.<br />

Számadó, S. (2000). Cheating as a mixed strategy in a simple model of aggressive communication.<br />

Anim. Behav. 59, 221–230.<br />

Turner, C. R., Derylo, M., de Santana, C. D., Alves-Gomes, J. A., and Smith, G. T. (2007).<br />

Phylogenetic comparative analysis of electric communication signals in ghost knifefishes<br />

(Gymnotiformes: Apteronotidae). J. Exp. Biol. 210, 4104–4122.<br />

Vehrencamp, S. L. (2000). Handicap, index, and conventional signal elements of bird song.<br />

In “Animal Signals: Signalling and Signal Design in Animal Communication” (Y. Espmark,<br />

T. Amundsen, and G. Rosenqvist, eds.), pp. 277–300. Tapir Academic Press, Trondheim.<br />

Wagner, W. E. (1992). Deceptive or honest signalling of fighting ability A test of alternative<br />

hypotheses for the function of changes in call dominant frequency by male cricket frogs. Anim.<br />

Behav. 44, 449–462.<br />

Weary, D. M., Lemon, R. E., and Perrault, S. (1994). Different responses to different song types in<br />

American redstarts. Auk 111, 730–734.<br />

Wheeler, M., and De Bourcier, P. (1995). How not to murder your neighbor: Using synthetic<br />

behavioral ecology to study aggressive signaling. Adapt. Behav. 3, 273–309.<br />

Wingfield, J. C., Moore, I. T., Goymann, W., Wacker, D. W., and Sperry, T. (2005). Con<strong>text</strong>s and<br />

ethology of vertebrate aggression: Implications for the evolution of hormone-behavior interactions.<br />

In “Biology of Aggression” (R. J. Nelson, ed.), pp. 179–211. Oxford <strong>University</strong> Press, Oxford.<br />

Wong, B. B. M., and Candolin, U. (2005). How is female mate choice affected by male competition<br />

Biol. Rev. 80, 559–571.<br />

Zahavi, A. (1987). The theory of signal selection and some of its implications. In “International<br />

Symposium of Biological Evolution” (V. P. Delfino, ed.), pp. 305–327. Adriatica Editrice, Bari.


Intentionally left as blank


4<br />

Self-Structuring Properties of<br />

Dominance Hierarchies: A New<br />

Perspective<br />

Ivan D. Chase* and Kristine Seitz †<br />

*Department of Sociology, Stony Brook <strong>University</strong>, Stony Brook,<br />

New York, USA<br />

† Department of Biology, Stony Brook <strong>University</strong>, Stony Brook,<br />

New York, USA<br />

I. Introduction<br />

II. Definitions<br />

A. Dominance relationships<br />

B. Dominance hierarchies<br />

III. Animal Models<br />

A. Chickens<br />

B. Fish<br />

C. Crustaceans<br />

D. Primates<br />

IV. Factors Affecting Dominance Relationships in Pairs of Animals<br />

A. Physical differences<br />

B. Physiology<br />

C. Genetics<br />

D. Behavioral states: Winner, loser and bystander effects<br />

V. Formation of Dominance Relationships<br />

and Dominance Hierarchies in Groups<br />

A. Differences in individual attributes and hierarchy formation<br />

B. Influence of social factors on linear hierarchy formation<br />

VI. A New Approach to Explaining the Formation<br />

of Linear Hierarchies: Behavioral Processes<br />

A. Modifications of the jigsaw puzzle model<br />

B. Experimental evidence concerning animal cognitive<br />

abilities and processes of interaction<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00001-0


52 Chase and Seitz<br />

VII. Conclusion<br />

Acknowledgments<br />

References<br />

ABSTRACT<br />

Using aggressive behavior, animals of many species establish dominance hierarchies<br />

in both nature and the laboratory. Rank in these hierarchies influences<br />

many aspects of animals’ lives including their health, physiology, weight gain,<br />

genetic expression, and ability to reproduce and raise viable offspring. In this<br />

chapter, we define dominance relationships and dominance hierarchies, discuss<br />

several model species used in dominance studies, and consider factors that<br />

predict the outcomes of dominance encounters in dyads and small groups<br />

of animals. Researchers have shown that individual differences in attributes, as<br />

well as in states (recent behavioral experiences), influence the outcomes of<br />

dominance encounters in dyads. Attributes include physical, physiological, and<br />

genetic characteristics while states include recent experiences such as winning or<br />

losing earlier contests. However, surprisingly, we marshal experimental and<br />

theoretical evidence to demonstrate that these differences have significantly<br />

less or no ability to predict the outcomes of dominance encounters for animals<br />

in groups as small as three or four individuals. Given these results, we pose an<br />

alternative research question: How do animals of so many species form hierarchies<br />

with characteristic linear structures despite the relatively low predictability<br />

based upon individual differences In answer to this question, we review the<br />

evidence for an alternative approach suggesting that dominance hierarchies<br />

are self-structuring. That is, we suggest that linear forms of organization in<br />

hierarchies emerge from several kinds of behavioral processes, or sequences of<br />

interaction, that are common across many different species of animals from ants<br />

to chickens and fish and even some primates. This new approach inspires a<br />

variety of further questions for research. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

Both humans and animals use aggression in many con<strong>text</strong>s as discussed in this<br />

volume. In this chapter, we talk about aggression as it is used in the social<br />

con<strong>text</strong> of establishing dominance relationships and dominance hierarchies.<br />

A broad range of species—from insects to humans—form these types of relationships<br />

and hierarchies, and where they occur, hierarchy rank has wide-ranging<br />

and serious consequences for individuals (Addison and Simmel, 1970; Barkan


4. Self-Structuring Properties of Dominance Hierarchies 53<br />

et al., 1986; Goessmann et al., 2000; Hausfater et al., 1982; Heinze, 1990;<br />

Nelissen, 1985; Post, 1992; Savin-Williams, 1980; Vannini and Sardini, 1971;<br />

Wilson, 1975). These consequences include variation in physiology, stress,<br />

health, growth rate, access to sexual partners, ability to raise viable offspring,<br />

and even in the thickness of nerves leading from the hypothalamus to the<br />

pituitary gland (Clutton-Brock et al., 1984; Ellis, 1995; Francis et al., 1993;<br />

Holekamp and Smale, 1993; Post, 1992; Sapolsky and Share, 1994).<br />

Our discussion centers on dominance in less complex animals such as<br />

chickens, fish, and nonprimate mammals. We begin with behavioral definitions<br />

of dominance relationships and hierarchies. We then move on to the factors that<br />

predict dominance in pairs of animals including differences in both traits and<br />

behavioral states. Traits include genetic, physical, physiological, and “personality”<br />

attributes; behavioral states include the influences of winning and losing contests,<br />

as well as observing the contests of other individuals. Next, we review the<br />

research that demonstrates, very surprisingly, that factors affecting dyadic<br />

encounters are poor predictors of outcome in dominance encounters within<br />

groups of any size, even as small as three or four individuals. Consequently,<br />

researchers should not expect that findings from dominance in pairs of animals<br />

will easily generalize to small groups of animals.<br />

Finally, we suggest that rather than trying to predict the ranks of<br />

individuals in groups, a more appropriate research question is to ask how these<br />

hierarchies often come to have a linear structure (defined below) across many<br />

species. We discuss recent experimental findings that conclude that these linear<br />

hierarchies are self-structuring or self-organizing. Self-structuring hierarchies are<br />

common across a range of species. They emerge from the repeated use of several<br />

characteristic, small-scale behavioral processes, or sequences of behavior. We<br />

describe how these processes generate linear hierarchies and discuss some unresolved<br />

experimental questions suggested by these behavioral processes.<br />

II. DEFINITIONS<br />

A. Dominance relationships<br />

While researchers have proposed a variety of definitions, in this chapter, we will<br />

be using a strictly behavioral measure of dominance relationships. More specifically,<br />

we will define a dominance relationship as characterized by an asymmetry<br />

of aggressive behaviors by one animal toward another. Typically, both noncontact<br />

and contact behaviors are involved. Common noncontact type behaviors<br />

are chasing, displacement, or the threat of aggressive contact. Threat behaviors<br />

vary by species and include gill flaring in fish; certain vocalizations and gestures<br />

in primates; and short, rapid movements toward the threatened individual in a


54 Chase and Seitz<br />

variety of species. Aggressive contact behaviors also vary by species and include<br />

pecking in the case of chickens and other birds; biting in fish, rodents, and<br />

primates; and grasping with claws in crayfish, lobsters, hermit crabs, and other<br />

crustaceans.<br />

There is no established standard that defines the number of acts in a row<br />

that are recorded to determine dominance in different species. However, the<br />

general rule is to use a sufficient number such that, when it is reached, a stable<br />

relationship has been revealed with very little likelihood of the animal declared<br />

subordinate beginning to attack the one declared dominant. For example,<br />

in determining the presence of dominance relationships, Chase et al. (2002)<br />

declared that one cichlid fish was dominant over another if it bit or chased<br />

the other fish six times in a row without the other fish initiating an aggressive<br />

action in return.<br />

Other methods for determining dominance, such as recording which of<br />

two animals obtains a desired piece of food, do not necessarily reflect the kind of<br />

relatively stable social relationships that are indicated by asymmetries in aggressive<br />

behavior in pairs of animals. For example, when two monkeys are competing<br />

for a peanut, the winner may be an otherwise subordinate animal that is quicker<br />

and ready to withstand the chasing and harassing it will receive from the<br />

normally dominant animal with which it lives in order to secure the peanut.<br />

Researchers also sometimes determine dominance by observing which individual<br />

delivers the majority, rather than an uninterrupted sequence, of aggressive acts<br />

over a period of observation. A measure of dominance such as this may be<br />

appropriate in some species, such as pigeons and young children, who do not<br />

always form completely asymmetric relationships. Such measures, however, can<br />

be misleading if used when animals meet for the first time. When animals meet<br />

initially, there is often a trading back and forth of aggressive actions before one<br />

individual begins to deliver all the actions and clearly becomes dominant over<br />

the other. Figure 4.1 is a music notation graph showing the interactions of two<br />

0 0:10 0:20<br />

A<br />

Figure 4.1. Graphic display of the record of interaction between two hens using music notation.<br />

Horizontal lines represent individuals, ordered by eventual dominance rank. Each<br />

aggressive act between individuals is indicated by a vertical arrow from the line<br />

representing the initiator to the line representing the receiver. The time in minutes<br />

since the assembly of the pair appears above the graph. Letters at the ends of the<br />

horizontal lines identify the hens. See Chase (2006) for more information about the<br />

uses of music notation in visualizing interactions in groups of animals and humans.<br />

B


4. Self-Structuring Properties of Dominance Hierarchies 55<br />

chickens setting up a relationship (Chase 2006). If only the first 10 min of<br />

interactions are considered, then chicken B is dominant over A since B delivers<br />

83% of the aggressive interactions and chicken A only 17%. However, as time<br />

goes on, the back and forth actions stop, and chicken A is soon delivering<br />

all aggressive acts. If a researcher had only considered which individual initiated<br />

the majority of acts during the initial phases of the interaction, an incorrect<br />

indication of dominance would have resulted.<br />

B. Dominance hierarchies<br />

A dominance hierarchy is the overall collection, or network, of dominance<br />

relationships among the pairs of individuals in a group. In many small groups<br />

of animals and human children of around eight or ten members or less, dominance<br />

hierarchies often take a classical linear form (Addison and Simmel, 1970;<br />

Barkan et al., 1986; Goessmann et al., 2000; Hausfater et al., 1982; Heinze, 1990;<br />

Nelissen, 1985; Post, 1992; Savin-Williams, 1980; Vannini and Sardini, 1971;<br />

Wilson, 1975). In a linear hierarchy, there is one individual who dominates all<br />

the other group members, a second who dominates all but the top individual, and<br />

so on, down to the last individual who dominates no one. In larger groups, there<br />

is often the skeleton of a linear structure, but even with extensive observations,<br />

researchers do not see interactions between some pairs, especially those that<br />

seem distant in rank. In hierarchies that are not linear, there are inconsistencies<br />

in rank showing intransitive relationships (A dominates B, B dominates C, but<br />

C dominates A). The hierarchies in some animals, especially those with more<br />

complex social organization such as dolphins, chimpanzees, hyenas, baboons,<br />

and macaques, are often too complex to be simply classified as linear (Holekamp<br />

and Smale, 1993; Kummer, 1984; Möller et al., 2001, 2006; Surbeck et al., 2011;<br />

Widdig et al., 2001, among many others). Our discussion here will concentrate<br />

on those animals forming more linear hierarchies.<br />

III. ANIMAL MODELS<br />

Animal behaviorists have shown that a huge range of animals establish dominance<br />

relationships and dominance hierarchies in the wild and in the laboratory. These<br />

include insects such as fruit flies and some ants, wasps and cockroaches; crustaceans<br />

such as hermit crabs, crayfish, and lobsters; reptiles such as anoles; many<br />

species of fish, birds, and mammals; and even human preschoolers and adolescents<br />

(Addison and Simmel, 1970; Barkan et al., 1986; Clark, 1998; Goessmann et al.,<br />

2000; Hausfater et al., 1982; Heinze, 1990; Nelissen, 1985; Post, 1992; Queller<br />

et al., 2000; Savin-Williams, 1980; Vannini and Sardini, 1971; Wilson 1975).


56 Chase and Seitz<br />

In biomedical research, an animal model is usually an animal species<br />

used for research on a human disease or other condition. Dominance researchers<br />

do not often use specific species in the strict sense of models for human conditions,<br />

but instead as models for dominance in animals more broadly. The partial<br />

exception to this is that some researchers have studied dominance in primates to<br />

discover information about how dominance processes may work in human groups<br />

(see below). Below is a brief description of animals or animal groups used<br />

frequently in dominance research and a representative, but by no means<br />

comprehensive, sampling of work done with these animals.<br />

A. Chickens<br />

Chickens were among the first animals to be studied for their dominance<br />

relationships. Schjelderup-Ebbe (1922) introduced dominance hierarchies into<br />

the modern study of animal behavior and coined the term “peck order” (or<br />

Hackordnung in the original German in which he wrote). Schjelderup-Ebbe<br />

(1922) was among the first researchers to observe the highly linear structure of<br />

pecking orders. He noted that a number of factors influenced rank in the flock,<br />

including stress, prior experience, overall health, mating condition, and age. He<br />

further concluded that dominance is based, not just on the size and strength of<br />

the combatants but also on the perception of fellow flock members (Schjelderup-<br />

Ebbe, 1922).<br />

Other early researchers used chickens to explore the relationship<br />

between stress and dominance. Sactuary (1932), for example, showed that<br />

hens that mysteriously molted out of season and went out of laying condition<br />

had been relegated to the lower ranks of the flock. Thus Sactuary (1932) linked<br />

rank to both fitness (ability to produce offspring) and stress levels.<br />

Some more recent investigations in chickens have focused on the<br />

relationship between dominance, aggression, and selective breeding. These<br />

lines of inquiry began with the rise of the factory farms, in which aggression<br />

leading to deaths and lowered egg production is of great concern (Craig and<br />

Muir, 1996; Craig et al., 1965, 1969, 1975, among others).<br />

B. Fish<br />

Fish have recently become one of the most popular vertebrate models for<br />

dominance research. The fish model system is comparable to chickens, in that<br />

fish possess easily observed dominance behaviors (chases, bites, and threat<br />

behaviors), recognize members of their group as individuals, and can maintain<br />

their dominance hierarchies for extended periods of time. Fish, however, are<br />

easier to maintain in the laboratory than chickens. They have been used for a<br />

variety of studies relating to dominance which we can only briefly cover here.


One common type of study of fish explores how differences in behavioral<br />

states and individual attributes affect the outcome of dominance contests. For<br />

example, researchers have used fish to investigate the so-called winner, loser, and<br />

bystander effects (defined below) (Chase et al., 1994, 2002; Hsu and Wolf, 1999;<br />

Hsu et al., 2006; Oliveira et al., 2009, among others); prior residency, a kind of<br />

home field advantage assumed to confer benefits in social con<strong>text</strong>s (Beaugrand<br />

and Beaugrand, 1991); and the effects of differences in size and prior social<br />

experience (Beaugrand and Cotnoir, 1996).<br />

Besides studying factors affecting the outcome of contests, researchers<br />

have utilized fish in selection experiments studying the heritability of dominancerelated<br />

aggression (Bakker, 1985, 1986; Francis, 1984, 1987) and in investigations<br />

into physiological and genetic components of dominance (see Sloman and<br />

Armstrong 2002 for a review of physiological aspects).<br />

C. Crustaceans<br />

Crustaceans are a unique model system that allows researchers to study chemical<br />

signaling behaviors, and the anatomy of their nervous systems enables researchers to<br />

study the neural underpinnings of dominance relationship formation (Moore and<br />

Bergman, 2005). One of the most commonly studied chemical signals in this group<br />

of species is the release of urine during agonistic behaviors. In lobsters, this signal has<br />

been implicated in the maintenance of dominance hierarchies (Breithaupt and<br />

Atema, 2000; Karavanich and Atema, 1998), and in crayfish, it has been shown to<br />

reduce aggression in opponents during dominance bouts (Breithaupt and Eger,<br />

2002). Yeh et al. (1997) showed that changes in dominance status altered levels of<br />

serotonin in the crayfish, Procambarus clarkii. These changes caused modifications in<br />

the command neuron involved in escape behaviors in this species and were found to<br />

be reversible and linked to changes in the population of serotonin receptors.<br />

D. Primates<br />

4. Self-Structuring Properties of Dominance Hierarchies 57<br />

The dominance systems of primates are often more complex than the other<br />

model systems just discussed. As a result, their dominance behavior can more<br />

easily be generalized to humans. One of the most important lines of research in<br />

primates has shown how stress affects the hormonal responses of animals of<br />

different ranks. Sapolsky (1982) studied wild olive baboons (Papio anubis) and<br />

found that high-ranking males showed a low initial level of cortisol, but in<br />

response to stress, they had faster and larger spikes of cortisol than their less<br />

successful counterparts. Sapolsky (1982) suggested that the high ranking member’s<br />

cortisol responses might be more adaptive to their social environments,<br />

because their usual, lower cortisol levels conferred immunological and other<br />

health benefits. A general review of the influence of hierarchies on primate<br />

health can be found in Sapolsky (2005).


58 Chase and Seitz<br />

IV. FACTORS AFFECTING DOMINANCE RELATIONSHIPS<br />

IN PAIRS OF ANIMALS<br />

In attempting to predict the outcomes of dominance contests in pairs of animals,<br />

researchers have used two broad classes of variables: differences in attributes or<br />

traits and differences in behavioral conditions or states. Attributes are relatively<br />

long-lasting characteristics, while states are shorter-term conditions often influenced<br />

by behavioral events.<br />

A. Physical differences<br />

Differences in physical attributes often have a considerable impact on the<br />

outcome of dominance encounters. One common characteristic used is differences<br />

in the sizes of the organisms, which can be broken down into two<br />

categories: differences in weights and differences in lengths or heights. Researchers<br />

have found that larger, heavier animals usually dominate animals that are<br />

smaller and lighter (Frey and Miller, 1972; Houpt et al., 1978; Nakano and<br />

Furukawa-Tanaka, 1994; Knights, 1987; Lott and Galland, 1987). However,<br />

when size differences are smaller, other factors can influence the outcomes of<br />

contests. For example, in male green swordtail fish, a difference of 20–30% in the<br />

size of the lateral surface area of fish meeting in dyadic dominance contests<br />

generally resulted in the larger fish becoming dominant over the smaller<br />

(Beaugrand et al., 1996). Contests between fish with size differences of<br />

10–20%, however, showed that other factors such as prior social experiences<br />

(winning or losing) and prior residency influence the outcome of contests.<br />

Size differences below 10% do not influence dominance contests at all. Instead,<br />

the social experience (discussed below) of the fish is usually the deciding factor in<br />

dominance contests (Beaugrand et al., 1996). Similar to standard length, the<br />

effects of weight on dominance success can be ameliorated by other factors such<br />

as having won or lost a prior contest (Beacham, 1988; Schulte-Hostedde and<br />

Millar, 2002).<br />

Although size and weight are, perhaps, the physical attributes most<br />

widely studied for their effect on dominance contests, other physical features<br />

and conditions have also been implicated in dominance success. One example<br />

that has been studied extensively is the dominance badge, an area of color on the<br />

body of an animal that acts to indicate dominance to conspecifics (see Senar,<br />

1999 for a review). Other examples include the state of molt and the size of<br />

combs in chickens (Collias, 1943), the size of genital papilla in fish (Schwanck,<br />

1980), and the bill size in birds (Shaw, 1986).


4. Self-Structuring Properties of Dominance Hierarchies 59<br />

1. Behavioral profile or personality<br />

Repeatable behavioral type or personality can be defined as suites of behaviors<br />

that differ among individuals but are consistently repeatable in multiple con<strong>text</strong>s<br />

over time (Bell et al., 2009; Boon et al., 2007; Groothius and Carere, 2005;<br />

Martin and Réale, 2008; Sih et al., 2004; Sinn and Moltschaniwskyj, 2005;<br />

Svartberg et al., 2005). Researchers have demonstrated that behavioral profiles<br />

can predict the outcome of dyadic dominance encounters in a variety of species<br />

with high accuracy. For example, in fish, brown trout that scored higher in<br />

boldness were more likely to dominate those that scored lower (Sundström<br />

et al., 2004), and rainbow trout that had shorter or more proactive responses to<br />

stress were more likely to dominate those with longer or reactive responses<br />

(Øverli et al., 2004; Schjolden et al., 2005). In birds, mountain chickadees<br />

classified as high-exploring individuals (those that visited more sites within a<br />

strange area) dominated low-exploring individuals, and in great tits, fast<br />

explorers dominated slow explorers (Fox et al., 2009; Verbeek et al., 1996).<br />

Interestingly, Verbeek et al. (1999) found that the same behavioral profiles in<br />

great tits that predicted dominance in dyads gave opposite results in groups of five<br />

to eight great tits. Here, the slow explorers had higher average dominance scores.<br />

B. Physiology<br />

Whether or not physiological differences can predict the outcome of dyadic<br />

dominance encounters is an extremely vexed question. In the mid-twentieth<br />

century, researchers thought that differences in testosterone, among other<br />

hormones, were reliable determinants of dominance (For recent work see<br />

Huber et al., 1997). However, subsequent research demonstrated that the causal<br />

direction is often reversed—in many species, the ranks of individuals in hierarchies<br />

has a strong influence on the levels of their hormones and other physiologically<br />

active chemicals rather than vice versa (see, e.g., Eaton and Resko,<br />

1974; Sapolsky, 1982; Trainor and Hofmann, 2007). Further, Sloman and<br />

Armstrong (2002), in a general review, suggest that at least for fish, the physiological<br />

effects of dominance encounters in simple laboratory settings may be<br />

stronger than those observed in more complex laboratory or natural habitats.<br />

These caveats notwithstanding, there is a considerable recent literature<br />

on physiological predictors of dominance in the dyadic encounters of fish, chiefly<br />

in trout and salmon. For example, Metcalfe et al. (1995), Cutts et al. (1999), and<br />

McCarthy (2001) find that fish with higher relative metabolic rates dominate<br />

those with lower relative rates. In tests of responses to stress, Øverli et al. (2004)<br />

and Schjolden et al. (2005) report that trout with lower levels of cortisol defeat<br />

those with higher levels. In experiments in which Arctic charr are dosed with


60 Chase and Seitz<br />

L-dopa, the immediate precursor of the neurotransmitter dopamine, and trout are<br />

dosed with growth hormone, treated fish dominated control fish at significant<br />

rates (Johnsson and Björnsson, 1994; Winberg and Nilsson, 1992).<br />

We obviously need further research to untangle the complicated chains<br />

of cause and effect among various physiological variables and outcomes in<br />

dominance relationships.<br />

C. Genetics<br />

The inheritance of dominance and aggressiveness has been a topic of interest<br />

since the field’s inception. In artificial selection experiments, Craig et al. (1965)<br />

were able to produce hens with diverging dominance abilities. In dyadic contests,<br />

hens of high dominance ability usually defeated those of low dominance ability.<br />

Even based on these early findings, however, Craig et al. (1965) concluded that<br />

variations caused by interactions between genes (nonadditive genetic variation)<br />

and environmental factors were likely to be important in the inheritance of<br />

dominance. Similar studies of paradise fish (Francis, 1984, 1987), cockroaches<br />

(Moore, 1990), and deer mice (Dewsbury, 1990) confirmed that dominance<br />

could be artificially selected in a variety of species. Artificial selection, however,<br />

can only imply a genetic basis for dominance and cannot identify which genes<br />

are responsible for dominance or subordination. An alternative explanation for<br />

at least some of these results is that the social environment of mothers (including<br />

their levels of aggression and dominance ranks) can expose prenatal young to<br />

androgens that can influence their offsprings’ behavior. Such maternal influences<br />

operate independently of genotype and have been implicated in the inheritance<br />

of rank-related behavior. For example, the level of female aggression affects the<br />

amount of maternally derived testosterone in tree swallow eggs, which, in turn,<br />

influences the growth and dominance of the hatchlings (Whittingham and<br />

Schwabl, 2002). In mammals, higher ranking female hyenas (Crocuta crocuta)<br />

have higher levels of in utero androgens causing their cubs to more aggressive<br />

than those of lower ranking females (Dloniak et al., 2006).<br />

To begin to tease apart these and other influences on rank order, the<br />

newest technological advances in molecular biology are being employed to<br />

investigate which genes influence social behavior. Sociogenomics, the study of<br />

social systems at a molecular level, can offer us insights and information never<br />

before available to behavioral scientists. A variety of unique insights have arisen<br />

from this new way of studying social dominance and are revealing two major<br />

themes: one theme is that genes involved in nonsocial behaviors are often also<br />

implicated in social behaviors; the second is that genes are highly sensitive to<br />

social influences, and regulation of gene expression by social factors heavily<br />

influences behavior (Robinson et al., 2005).


4. Self-Structuring Properties of Dominance Hierarchies 61<br />

An outstanding example of the interplay between genetic expression<br />

and social factors occurs in the cichlid fish Astatolapia burtoni. In this fish,<br />

dominant males are brightly colored and actively defend territories for mating.<br />

Subordinate males are nonreproductive, move about in schools and mimic<br />

females’ cryptic coloration. Subordinate males, however, grow faster than dominant<br />

males, giving subordinates the opportunity to depose dominant males from<br />

their territories. These phenotypes are plastic and males may switch back and<br />

forth between phenotypes several times in a life span, depending on the availability<br />

of suitable territories to defend (Burmeister et al., 2005; Renn et al., 2008).<br />

Burmeister et al. (2005) investigated the neural mechanisms linking<br />

social environment to physiological changes associated with dominance. They<br />

found that when a subordinate male perceives an opportunity to move to a<br />

territory and become dominant, he begins to produce dominant coloration and<br />

some initial behavioral changes in as little as a day (Burmeister et al., 2005).<br />

It takes about 7 days, however, for males ascending to dominance to produce the<br />

same amount of gonadotropin-releasing hormone (GnRH1) as a dominant male,<br />

during which time the size of testes and GnRH1 neurons increase to sizes<br />

comparable to dominants (Burmeister et al., 2005). In A. burtoni, GnRH1 is<br />

produced by neurons in the anterior parvocellular preoptic nucleus (aPPn), the<br />

most anterior part of the preoptic area in teleosts. To study the behavioral and the<br />

genomic response to social opportunity, researchers chose to focus on the gene<br />

egr-1, which codes for a transcription factor involved in neuronal plasticity and<br />

links membrane depolarization to late-response target genes. Expression of this<br />

gene was compared in socially ascending males and dominant and subordinate<br />

males in stable hierarchies (Burmeister et al., 2005). Their results show that<br />

socially ascending males had a twofold induction of egr-1 in the aPPn, compared<br />

to the both dominant and subordinate males in stable positions. Expression levels<br />

in other parts of the brain did not differ with social status or opportunity<br />

(Burmeister et al., 2005). Stable dominant males do not show this spike in<br />

egr-1, suggesting that this change is a response to social opportunity rather<br />

than a response to dominance itself. Although socially ascending males also<br />

show a difference in physical activity (e.g., more threat displays), it is not clear<br />

whether there is a simple relationship between egr-1 expression and increased<br />

motor activity. Instead, Burmeister et al. (2005) conclude that the relationship of<br />

social con<strong>text</strong>, expression of egr-1, and activity differences have a complex<br />

relationship that cannot be adequately explained by the simple functional<br />

motor and sensory aspects of the experience (Burmeister et al., 2005).<br />

Renn et al. (2008) studied how social con<strong>text</strong> affects physiology, but in<br />

this case, a microarray was used to investigate coregulated gene sets that might<br />

differentiate dominant males, subordinate males, and brooding females. The<br />

results show that there are, indeed, gene sets that are common to each of these<br />

phenotypes, with males (both dominant and subordinate) and females having


62 Chase and Seitz<br />

the largest (16%) difference in gene expression (Renn et al., 2008). Twenty-one<br />

genes were found to be upregulated in the subordinate male phenotype, and it<br />

was hypothesized that downregulation of these genes would lead to the dominant<br />

phenotype. Additionally, subordinate males and brooding females were found to<br />

have a similar expression pattern for 16 genes, possibly suggesting a type of<br />

subordination module (Renn et al., 2008). Interestingly, although gene sets for<br />

phenotypic traits were found, results also showed that there was as much variation<br />

in gene expression between individuals of the same phenotype as there was<br />

between the phenotypes themselves. This suggests that widely variant gene<br />

expression in individuals can still yield reliable, easily identifiable phenotypes<br />

(Renn et al., 2008).<br />

Trainor and Hofmann (2007) investigated the neuropeptide hormone<br />

somatostatin, and its receptors, for possible involvement in social behavior. This<br />

hormone and its numerous receptor subtypes have been shown to play a role in<br />

the inhibition of growth hormone secretion, among other more diverse effects.<br />

The relationship between somatostatin gene expression and body size differed<br />

between dominant and subordinate individuals. In dominant males, the gene<br />

expression of one subgroup of receptors in the hypothalamus was negatively<br />

associated with body size. In subordinate fish, however, gene expression was<br />

positively correlated with body size. This suggests that growth in this animal<br />

may be socially mediated at the genetic level (Trainor and Hofmann, 2007).<br />

D. Behavioral states: Winner, loser and bystander effects<br />

In addition to differences in attributes or traits, considerable research also<br />

demonstrates that differences in states can influence the outcomes of dominance<br />

encounters in pairs of animals. Most of this research has examined what are<br />

known as winner, loser, and bystander effects. In a winner effect, an animal that<br />

has won an earlier contest with one individual has an increased probability of<br />

winning a second contest with another individual. In a loser effect, an animal<br />

that has lost a dominance encounter with one individual has an increased<br />

probability of losing a subsequent contest with another individual. In a bystander<br />

effect, an animal that has observed a dominance contest between two others<br />

alters its behavior, compared to a nonobserver, when it meets either of the<br />

animals that it observed interacting.<br />

Researchers have discovered loser effects in a broad range of species,<br />

and there is some evidence that these effects may last for several days (see, e.g.,<br />

Chase et al., 1994; Hsu et al., 2006). Winner effects seem to be less common<br />

across species and not as strong as loser effects. Further, some species seem not to<br />

have them at all (Chase et al., 1994; Rutte et al., 2006). In particular, Fuxjagera<br />

and Marlera (2010) show that winner effects can be documented in some species


4. Self-Structuring Properties of Dominance Hierarchies 63<br />

but can be nonexistent in other, closely related species. Where winner effects<br />

occur, there is some evidence that they are of much shorter duration than loser<br />

effects, lasting perhaps less than an hour or so after an individual’s initial winning<br />

experience (Bergman et al., 2003; Chase et al., 1994). Oliveira et al. (2009) have<br />

shown that winner effects can be ameliorated with antiandrogen drugs, but loser<br />

effects are unchanged. Clearly, additional work is needed in this area to elucidate<br />

the role these effects have on the formation of dominance relationships in pairs<br />

of animals.<br />

Research indicates that animals in many species are attentive observers<br />

of other individuals and that they use the information gained in their observations<br />

in shaping their future behavior with those observed. For example, a<br />

bystander fish may be less aggressive when it meets another fish it has observed<br />

winning a contest. Bystander effects have been observed in a broad range of<br />

species (see, e.g., Oliveira et al., 1998; Oliveira et al., 2001; Danchin et al., 2004;<br />

Peake and McGregor, 2004).<br />

V. FORMATION OF DOMINANCE RELATIONSHIPS<br />

AND DOMINANCE HIERARCHIES IN GROUPS<br />

Given the research that we have just reviewed, it would seem natural to assume<br />

that the same factors that strongly predict the outcomes of dominance encounters<br />

in pairs of animals by themselves should also work for dominance encounters<br />

between pairs of animals in groups. That is, the factors that predict dominance<br />

in isolated pairs should also predict dominance for socially embedded pairs.<br />

Predicting the outcome of dominance encounters for all the embedded pairs in<br />

a group would allow us to rank individuals within the dominance hierarchy and<br />

reveal the hierarchical structure. Surprisingly, while individual differences<br />

in attributes and states do have some influence on rank in hierarchies, that<br />

influence is significantly less in groups than it is in dyadic pairs. Consequently,<br />

other factors must be at work in determining individual ranks within hierarchies<br />

and in generating hierarchical structure. Unraveling that paradox—how individuals<br />

can be clearly differentiated by rank, but with that differentiation not<br />

strongly based upon individual differences—is a great challenge in the study of<br />

dominance hierarchies.<br />

In the next section, we present evidence demonstrating that individual<br />

differences can neither adequately predict the places of individuals within<br />

hierarchies nor explain their overall linear structure. Following that, we describe<br />

a new approach that we believe can account for the common formation of linear<br />

hierarchies across a variety of species.


64 Chase and Seitz<br />

A. Differences in individual attributes and hierarchy formation<br />

The prior attributes hypothesis proposes that differences in the characteristics<br />

that animals possess before forming a hierarchy predetermine their resulting<br />

hierarchy ranks. Figure 4.2 illustrates this hypothesis in graphical form. In the<br />

hypothesis, the individual ranking highest on attributes takes the top position<br />

when the hierarchy forms; the individual ranking second-highest takes the<br />

next-to-the-top position; and so on. Rank based on prior attributes could be<br />

determined by any set of characteristics: physical ones such as weight, personality<br />

ones such as aggressiveness or boldness, genetic ones such as overall genotype or<br />

specific genetic markers, social ones such as the conditions of rearing or family<br />

background, physiological ones such as various hormone levels, and so on.<br />

Prior<br />

attribute<br />

score<br />

Hierarchy<br />

rank<br />

Figure 4.2. Graphical illustration of the prior attributes hypothesis. Size indicates relative prior<br />

attribute value; larger size indicates higher rank on prior attributes. Figure adapted from<br />

Figure 24.1, p. 570 in “Dominance hierarchies” by Ivan D. Chase and W. Brent<br />

Lindquist from Oxford Handbook of Analytical Sociology ed. by Hedström, P. and Bearman,<br />

P. (2009), by permission of Oxford <strong>University</strong> Press.


4. Self-Structuring Properties of Dominance Hierarchies 65<br />

The problem in testing the prior attributes hypothesis is that although<br />

an experimenter might know some of the traits that influence dominance, he or<br />

she might not know all those involved or the size of the contribution of a specific<br />

trait to dominance outcomes. In order to get around this problem, Chase et al.<br />

(2002) designed an experiment to test the prior attributes hypothesis without<br />

knowing which attributes or the sizes of their contributions that might be<br />

involved in hierarchy formation.<br />

In their experiment, they brought together groups of four cichlid fish<br />

and let them form hierarchies, separated the fish for two weeks, which was<br />

sufficient time for them to forget one another as individuals, and then reassembled<br />

them to form second hierarchies. The plan was to let the fish form a<br />

hierarchy and then, to the extent possible, “rewind their tape,” removing all<br />

memory of recent social experience before letting them form a second hierarchy.<br />

If prior attributes, whatever they might have been, determined the ranks of the<br />

fish in the first hierarchy, the attributes, provided they were reasonably stable,<br />

should also have determined the ranks of the fish in the second hierarchies.<br />

Consequently, by the prior attributes hypothesis, the positions of the fish in the<br />

first and second hierarchies should have been the same for all, or at least most, of<br />

the groups.<br />

The results of this experiment are shown in Fig. 4.3 and Table 4.1.<br />

Instead of a high proportion of groups having identical first and second hierarchies,<br />

the experimenters found that only about a quarter (26%) of the groups did<br />

so. In nearly three-quarters of the groups, two, three, or even all four fish had<br />

different ranks in the two hierarchies. Prior attributes did, however, have a<br />

moderate influence on the ranks of the fish within the hierarchy—more groups<br />

formed identical first and second hierarchies than expected by chance alone, and<br />

there was, on average, moderate rank order correlations between the ranks of<br />

individuals in the two hierarchies. However, the lack of a high proportion of<br />

groups with identical first and second hierarchies indicated that some other<br />

factor played a substantial part in the formation of linear hierarchies and the<br />

ranks of individuals within them.<br />

One question that can be raised about the interpretation of these results<br />

is, what if the fish changed after the first hierarchy so that their attribute ranks<br />

were different before they formed their second hierarchy For example, for<br />

simplicity consider just one attribute called dominance ability. What if the<br />

rank on dominance ability before the first hierarchy had been A, B, C, D, but<br />

before they formed the second hierarchy their order had changed to give a new<br />

ranking D, C, A, B The difference in dominance ability could still have<br />

accounted for the linear hierarchies but the fish would have different ranks in<br />

the second hierarchy than the first. This question is discussed in some detail in<br />

Chase et al., (2002), which argues against this counter-explanation. In addition<br />

to that discussion, some more recent experimental work also suggests a lack of


66 Chase and Seitz<br />

1st<br />

Hierarchy<br />

2nd<br />

Hierarchy<br />

1st<br />

Hierarchy<br />

2nd<br />

Hierarchy<br />

A<br />

B<br />

C<br />

D<br />

(6)<br />

A<br />

B<br />

C<br />

D<br />

A<br />

B<br />

C<br />

D<br />

(1)<br />

B<br />

A<br />

C<br />

D<br />

A<br />

B<br />

A<br />

A<br />

B<br />

A<br />

B<br />

C<br />

C<br />

C<br />

C<br />

B<br />

D<br />

(4)<br />

D<br />

D<br />

(1)<br />

D<br />

A<br />

B<br />

C<br />

D<br />

A<br />

B<br />

C<br />

D<br />

A<br />

B<br />

C<br />

D<br />

A<br />

B<br />

C<br />

D<br />

D<br />

B<br />

A<br />

C<br />

A<br />

C1 C2 C3<br />

(1)<br />

(1)<br />

C<br />

A<br />

A<br />

B<br />

B<br />

C<br />

D<br />

D<br />

(1) (2)<br />

B<br />

A<br />

B<br />

C D<br />

C<br />

D<br />

(1) (2)<br />

D<br />

A<br />

A<br />

B<br />

C1 C2 C3<br />

C<br />

(1)<br />

(1)<br />

A<br />

C2<br />

C3<br />

C1<br />

B<br />

C<br />

A<br />

D<br />

C<br />

A<br />

D<br />

B<br />

C2<br />

C3<br />

A<br />

C1<br />

Figure 4.3. Transition pattern between the ranks of the fish in their first and second hierarchies.<br />

The total number of groups in the experiment was 22; the number of groups showing a<br />

particular transition pattern is indicated in parentheses below each pattern. Fish that<br />

have an intransitive dominance relationship (A dominates B, B dominates C, and<br />

C dominates A) share the same rank. Intransitive relationships are discussed below.


4. Self-Structuring Properties of Dominance Hierarchies 67<br />

Table 4.1. Percentage of Groups with Different Numbers of Fish<br />

Changing Ranks Between their First and Second Hierarchies<br />

(n¼22)<br />

No. of fish changing ranks<br />

Percentage of groups<br />

0 27.3<br />

2 36.4<br />

3 18.2<br />

4 18.2<br />

support for this counter-explanation. When isolated pairs of fish are tested under<br />

the same experimental conditions as the groups forming and reforming hierarchies,<br />

they have an extremely high rate (94%) of forming the same dominance<br />

relationship each time. If A dominated B when they met the first time,<br />

A virtually always dominated B the second time they met. However, at 76%,<br />

the replication rate for socially embedded pairs in the hierarchy groups was<br />

significantly less than that of isolated pairs. If the rank of the fish on attributes<br />

changed between meetings so that they did not always dominate the same fish<br />

when their hierarchies formed the second time, then likewise there should have<br />

been a similarly low rate of replication in relationships when the isolated pairs<br />

met for the second time. But this did not occur, so the changes in the relationships<br />

and ranks of the fish must be accounted for by other factors rather than<br />

changes in ranks on attributes.<br />

B. Influence of social factors on linear hierarchy formation<br />

In order to discover what factors were necessary for the formation of linear<br />

hierarchies, Chase et al. (2002) carried out a second experiment with cichlid<br />

fish. In this experiment, they set out to test that social processes, behavioral<br />

processes that could only take place in a group con<strong>text</strong>, were crucial for linear<br />

hierarchy formation and that they contributed more to linearity than prior<br />

attributes. In this experiment, they allowed groups of four and five fish to form<br />

hierarchies by two means: round-robin competition and group assembly. In roundrobin<br />

competition, the fish in a group met only as isolated pairs, out of sight of the<br />

other members of their group. The sequence of meeting was as follows: first, A and<br />

B met, then C and D, A and C, D and B, and so on. In this way, differences in<br />

individual attributes could determine which one of a pair would dominate. If, say,<br />

A was superior in dominance attributes to B, A could dominate when they met as a<br />

pair. However, social processes such as C getting information by observing the<br />

outcome of a contest between A and B and then using that information in


68 Chase and Seitz<br />

interacting with either A or B was not possible, since each pair formed a relationship<br />

in isolation from other group members. In group assembly, all the members of<br />

a group met in an aquarium at the same time. This allowed fish to use whatever<br />

social process they were capable of in forming their hierarchies.<br />

Table 4.2 shows the results of this experiment. When groups of fish<br />

established their hierarchies using round-robin competition, only about half of<br />

them formed linear hierarchies (but see McGhee and Travis 2010 for contrasting<br />

results). When they established their hierarchies using group assembly, nearly all<br />

of the hierarchies were linear. Behavior that only occurred in a group con<strong>text</strong><br />

ensured the development of linear hierarchies, while differences in individual<br />

attributes did not. However, differences in individual attributes still had some<br />

influence on the production of linear hierarchies: the proportion of linear<br />

hierarchies with round-robin competition was higher in groups of five fish than<br />

would be expected by chance alone.<br />

Although the experiments just described demonstrated that differences<br />

in the attributes of individuals were of some importance in generating linear<br />

hierarchy structures, social processes of some sort were necessary to ensure the<br />

formation of the structures. Another way to look at these findings is that, given<br />

the attributes of individuals, there was still considerable randomness in their<br />

positions in the hierarchies. It was far from total randomness, but the amount of<br />

chance in dominance rank was still substantial. In spite of this degree of<br />

randomness, the hierarchy structures themselves were almost always linear.<br />

What social processes could ensure the common formation of these linear<br />

forms of social organization in spite of the lack of predictability concerning the<br />

individuals in the structure<br />

Chase (1982) proposed that winner, loser, and bystander effects together<br />

might be the social processes largely accounting for the formation of linear hierarchies<br />

across a variety of species. The basic idea was that even if you started with a<br />

group of animals of equal prior attributes, they could eventually be differentiated in<br />

terms of dominance through feedback during the course of their interactions. For<br />

example, assume that there are both winner and loser effects for some species and<br />

that A and B have the first interaction when a group is assembled. If A defeats B, A<br />

Table 4.2. Percentage of Linear Structures Expected in Random Hierarchies and Observed in<br />

Round-Robin Competition and Group Assembly in Groups of Four and Five Fish<br />

Method of forming hierarchy<br />

Size of group<br />

Random (%) Round robin (%) Group assembly (%)<br />

4 37.5 56.2 (n¼16) 92.0 (n¼25)<br />

5 11.7 50.0 (n¼12) 90.9 (n¼11)


4. Self-Structuring Properties of Dominance Hierarchies 69<br />

increases her probability of winning her next encounter and B decreases hers. A next<br />

meets C, defeats her, and again increases her probability of winning, while B<br />

encounters D, loses, and decreases her probability further.<br />

A number of researchers have developed mathematical models and<br />

computer simulations to show that feedback from winner and loser effects, either<br />

working by themselves or together, can, at least in theory, produce highly linear<br />

hierarchies (e.g., Bonabeau et al., 1999; Dugatkin, 1997; Hemelrijk, 1999; Hock<br />

and Huber, 2006; 2007; 2009; Skvoretz and Fararo, 1996; Skvoretz et al., 1996).<br />

However, these models were not tested against actual interaction records of real<br />

animals forming dominance hierarchies. When Lindquist and Chase (2009) did<br />

evaluate three (Bonabeau et al., 1999; Dugatkin, 1997; Hemelrijk, 1999) of the<br />

most prominent of these models and simulations using detailed data records for<br />

hens establishing hierarchies (Chase 1982), they found little support for the idea<br />

that winner and loser effects were responsible for the formation of linear dominance<br />

structures. In addition, when Lindquist and Chase (2009) examined the<br />

background assumptions on which these models and simulations were based,<br />

they found little support for these assumptions in the experimental literature.<br />

Assumptions in the models and simulations include animals not remembering<br />

one another as individuals, outcomes of earlier dominance contests during<br />

hierarchy formation not influencing the outcomes of later contests, and most<br />

important, winner and loser effects actually occurring in groups forming hierarchies.<br />

In fact, the literature indicated that the actual experimental findings<br />

were in virtually all cases directly opposite to the assumptions of the models and<br />

simulations. For example, animals setting up hierarchies do remember one<br />

another as individuals—even in the case of fruit flies (Yurkovic et al., 2006).<br />

The outcome of earlier contests do influence the later ones, and perhaps most<br />

striking of all, winner and loser effects do not seem to occur in groups forming<br />

hierarchies (Brown and Colgan, 1986; Chase et al., 2003; Cheney and Seyfarth,<br />

1990; D’Eath and Keeling, 2003; D’Ettore and Heinze, 2005; Gherardi and<br />

Atema, 2005; Karavanich and Atema, 1998; Lai et al., 2005; McLeman et al.,<br />

2005; Tibbetts, 2002; Todd et al., 1967; Yurkovic et al., 2006).<br />

In particular, Chase et al. (2003) investigated several basic aspects of<br />

dominance relationships in isolated versus socially embedded pairs in groups of<br />

three and four fish. These aspects of relationships included winner and loser<br />

effects, stability of relationships over time, and the ability of pairs to replicate a<br />

relationship after a separation of two weeks (as described above). Specifically,<br />

while there was a strong loser effect in isolated pairs of fish, this effect was not<br />

above chance for socially embedded pairs. There was no winner effect in either<br />

isolated or socially embedded pairs. In addition, dominance relationships were<br />

much less stable over time for pairs within groups (a significant proportion of<br />

these relationships reversed over 24 h) compared to no relationships reversing<br />

in isolated pairs, and a significantly smaller proportion of pairs within groups


70 Chase and Seitz<br />

did not form the same relationships when they met a second time after a<br />

separation of two weeks as compared to isolated pairs. In summary, all the<br />

aspects of relationships the researchers tested either disappeared or were significantly<br />

reduced for socially embedded pairs as compared to pairs by themselves.<br />

This experiment provides a strong warning of the danger of simply assuming<br />

that experimental results for isolated pairs can be automatically generalized<br />

to animals that are part of groups—even those as small as three or four<br />

individuals.<br />

Given the lack of fit between these three prominent winner/loser<br />

models and actual data on the formation of hierarchies in real animals, and the<br />

almost total absence of experimental support for the basic assumptions of the<br />

models, it seems reasonable to suggest that winner and loser effects cannot<br />

account for the common occurrence of linear hierarchies in animal groups. But<br />

could some other models, based upon states, still satisfactorily explain linear<br />

structures For example, what about bystander states These states have also been<br />

used in models, both for animals and humans, as a way to account for linear<br />

hierarchies (e.g., see Dugatkin and Earley, 2003; Skvoretz and Fararo, 1996;<br />

Skvoretz et al., 1996). Although Lindquist and Chase (2009) did not look at<br />

bystander effects per se, they did point out that a bystander effect can often be<br />

decomposed into winner and loser effects; for example, a bystander increases its<br />

probability of winning over an individual that it has observed losing a contest<br />

and decreases its probability of defeating an individual that it has observed<br />

winning a contest. In cases of this sort, Lindquist and Chase’s (2009) results<br />

also indicate the difficulty of bystander effects in explaining the common presence<br />

of linear structures.<br />

While it is impossible to prove categorically that no differences in states<br />

for individuals could ever account for the forms of hierarchy structures, winner,<br />

loser, and bystander effects are the best candidates that have been proposed so<br />

far. Given the lack of support for them, it appears doubtful, at least to us, that<br />

differences in the states of animals can be adequate explanations for the social<br />

organization of hierarchies.<br />

VI. A NEW APPROACH TO EXPLAINING THE FORMATION<br />

OF LINEAR HIERARCHIES: BEHAVIORAL PROCESSES<br />

Given the apparent absence of support for differences in individual attributes and<br />

states as satisfactory explanations for linear hierarchies, we now review an<br />

alternative view: that the social organization of hierarchies can be explained<br />

by characteristic behavioral processes that commonly occur across many species<br />

during hierarchy formation.


4. Self-Structuring Properties of Dominance Hierarchies 71<br />

Chase’s (1982) “jigsaw puzzle” model presented the original version of<br />

this idea. The jigsaw puzzle model suggested that like the picture in a real jigsaw<br />

puzzle, a linear hierarchy forms when the “right” small pieces, in this case of<br />

social interaction, are put together in the correct manner. In this way, the model<br />

sees the dominance hierarchy as self-organizing or self-structuring. More specifically,<br />

the model indicates four possible sequences for the formation of the first<br />

two dominance relationships in subgroups of three animals making up a larger<br />

group. These four sequences, shown in Fig. 4.4, have different implications for<br />

the formation of linear hierarchies. The two patterns on the left, Double Dominance<br />

and Double Subordinance, guarantee transitive dominance relationships,<br />

regardless of the direction that the missing third relationship in those sequences<br />

takes when it fills in later. In a transitive relationship, individual X dominates<br />

individual Y, individual Y dominates individual Z, and individual X also dominates<br />

individual Z. For example, in Double Dominance, if B later comes<br />

to dominate C, the transitive relationship is A dominates B, B dominates C,<br />

and A dominates C. If C later comes to dominate B, the transitive relationship is<br />

A dominates C, C dominates B, and A dominates B. The fact that Double<br />

Dominance and Double Subordinance guarantee transitive relationships is very<br />

important because transitive relationships are the building blocks of linear<br />

hierarchies. By mathematical definition, if all the subgroups of three individuals<br />

in a larger group have transitive relationships, the hierarchy is necessarily linear.<br />

Thus the presence of only Double Dominance and Double Subordinance<br />

sequences in the subgroups of three animals (component triads) making up a<br />

larger group guarantees that a hierarchy will be linear, even before the missing<br />

third relationships in the component triads have formed.<br />

A A A A<br />

1 2<br />

1 1 2<br />

1<br />

B C B<br />

2<br />

C B C B<br />

2<br />

C<br />

Double<br />

dominance<br />

Double<br />

subordinance<br />

Bystander<br />

dominates<br />

Initial<br />

subordinate<br />

initial dominant dominates<br />

bystander<br />

Figure 4.4. The four possible sequences for the first two dominance relationships in a component<br />

triad. Arrows show the direction of dominance relationships between the members of a<br />

triad. The number 1 indicates the first relationship to form in a triad and 2 indicates the<br />

second relationship. In all triads, A is the initial dominant, B is the initial subordinate,<br />

and C is the bystander. Figure adapted from Figure 24.2, p. 574, in “Dominance<br />

hierarchies” by Ivan D. Chase and W. Brent Lindquist from Oxford Handbook of<br />

Analytical Sociology ed. by Hedström, P. and Bearman, P. (2009), by permission of<br />

Oxford <strong>University</strong> Press.


72 Chase and Seitz<br />

On the other hand, if a hierarchy is nonlinear, it contains at least one<br />

component triad with an intransitive dominance relationship; the more intransitive<br />

triads, the further the hierarchy is from linearity. In an intransitive<br />

relationship, X dominates Y, Y dominates Z, but Z dominates X. The two<br />

sequences on the right of the figure, Bystander Dominates Initial Dominant<br />

and Initial Subordinate Dominates Bystander, can lead to either transitive or<br />

intransitive relationships depending upon how the third dominance relationship<br />

eventually fills in. For example, in Bystander Dominates Initial Dominant, the<br />

relationship is transitive if C later dominates B (C dominates A, A dominates B,<br />

and C dominates B), but intransitive if B later dominates C (A dominates B,<br />

B dominates C, C dominates A). If a group had one or more Bystander Dominates<br />

Initial Dominant and Initial Subordinate Dominates Bystander sequences,<br />

the chance for a nonlinear hierarchy would be increased when the third relationships<br />

in the triads eventually formed.<br />

In a study of groups of three hens forming hierarchies, Chase (1982)<br />

found that almost all relationships were established using Double Dominance<br />

and Double Subordinance sequences. In 23 groups of three hens, 91% of the<br />

groups used Double Dominance and Double Subordinance together, while only<br />

8% used the sequences not ensuring transitivity. In a second study of 14 groups of<br />

four hens (Chase, 1982), approximately 87% of the component triads in the<br />

groups (each group of four had four subgroups of three for 56 triads altogether)<br />

used Double Subordinance and Double Dominance, while approximately 13%<br />

had sequences not guaranteeing transitivity. These results are in contrast to those<br />

expected by chance, in which each sequence would have occurred about 25% of<br />

the time (or 50% combined for the two ensuring transitivity and 50% for the two<br />

not doing so).<br />

Thus, the jigsaw puzzle model indicated that the hens established<br />

dominance relationships largely through behavioral sequences that guaranteed<br />

transitivity in their triads, and transitivity in their triads in turn guaranteed<br />

overall linear hierarchy structures. After Chase’s (1982) application of the model<br />

to hens, other researchers used the original model and some modifications of it to<br />

examine dominance interactions in a broad range of species including rhesus<br />

monkeys, Japanese macaques, Harris sparrows, crayfish, and ants (Barchas and<br />

Mendoza, 1984; Chase and Rohwer, 1987; Eaton, 1984; Goessmann et al., 2000;<br />

Heinze, personal communication; Mendoza and Barchas, 1983). In spite of the<br />

great differences among these species in phylogenetics, intelligence, and ways of<br />

making a living, all showed highly significant use of sequences ensuring transitivity,<br />

although this was somewhat lower in crayfish and large groups of Harris<br />

sparrows (Chase and Rohwer, 1987; Goessmann et al., 2000). These results<br />

suggest that behavioral sequences ensuring transitive dominance relationships<br />

may be common for many species that form dominance hierarchies. Further<br />

research to confirm this possibility would be helpful.


4. Self-Structuring Properties of Dominance Hierarchies 73<br />

A. Modifications of the jigsaw puzzle model<br />

In recent work, Lindquist and Chase (2009) reanalyzed Chase’s (1982) original<br />

data for groups of four hens forming dominance hierarchies and discovered two<br />

additional behavioral processes promoting the efficient formation of linear hierarchies<br />

in addition to those described above under the jigsaw puzzle model. The<br />

first additional process is the relative lack of back and forth fighting in pairs of<br />

animals within a group establishing a dominance hierarchy. Lindquist and Chase<br />

(2009) referred to back and forth fighting as pair flips—first A attacks B, then B<br />

attacks A, and so forth. Consider two groups forming a dominance hierarchy: in<br />

one group, there are many pair flips before they eventually form a stable linear<br />

hierarchy. In the second group, there are no counterattacks, and the group forms a<br />

stable linear hierarchy without pair flips. The formation of dominance relationships<br />

and their linear hierarchy is much more “efficient” in the second group.<br />

In their analysis, Lindquist and Chase (2009) found that the hens<br />

formed their relationships with a high level of efficiency—one approaching<br />

that of the hypothetical second group just mentioned. Of the 7257 aggressive<br />

acts recorded over the 12 h of observation for each of the 14 groups of four hens<br />

(168 h of observation, total), only 138 interactions (1.9%) involved pair flips.<br />

The second additional behavioral process was the rapid conversion of<br />

intransitive dominance relationships to transitive ones. As indicated above, the<br />

original application of the jigsaw puzzle model to the hen data showed that the<br />

great majority (87%) of the behavioral sequences in the component triads for the<br />

groups of four hens were those ensuring transitivity. However, a more detailed<br />

reanalysis of the data indicated that a few triads did initially form intransitive<br />

relationships and that several others that initially had transitive relationships<br />

later developed intransitive ones. For example, if the triad ABC initially had a<br />

transitive relationship A>B, B>C, and A>C, it would become intransitive if<br />

C reversed its relationship with A to give A>B, B>C, and C>A.<br />

In their analysis, Chase and Linquist, (2009) found that, in virtually all<br />

cases, intransitive relationships were unstable and quickly converted to transitive<br />

ones, returning hierarchies to linearity after brief episodes of nonlinearity.<br />

The average number of dominance interactions (pecks, feather pulls, etc.) that<br />

occurred among group members before an intransitive triad was converted back<br />

to a transitive one was approximately 5.2. In contrast, the average number of<br />

interactions occurring before a transitive triad was converted into an intransitive<br />

one or into a different transitive one was approximately 54.3 interactions or over<br />

10 times as many. Research by Chase and Rohwer (1987) on Harris sparrows<br />

supports these findings: they also found a strong tendency for intransitive dominance<br />

relationships to be unstable and to convert to transitive ones. Further<br />

experimental work is needed to determine whether the instability of intransitive<br />

relationships and their reformation as transitive ones are found in other species.


74 Chase and Seitz<br />

B. Experimental evidence concerning animal cognitive abilities<br />

and processes of interaction<br />

In order for animals to carry out the kinds of behavioral processes that we have<br />

just discussed, they must possess an array of quite complex cognitive abilities.<br />

These include the abilities to remember one another as individuals, to make<br />

inferences about their own interactions and the interactions of others, and to use<br />

those inferences in adjusting their future dominance behavior. We have already<br />

discussed the extensive experimental evidence concerning the ability of animals<br />

to identify and remember one another as individuals, and to make inferences<br />

about certain kinds of interactions. In particular, we know that a broad range of<br />

species can infer transitivity (Bond et al., 2003; Davis, 1992; Gillian, 1981;<br />

Grosenick et al., 2007; Lazareva, 2004; Paz-y-Mino et al., 2004; Roberts and<br />

Phelps, 1994; Steirn et al., 1995; von Fersen et al., 1991). For example, if B has<br />

dominated C, and C observes A dominating B, C will act less aggressively toward<br />

A when they meet than C will toward an animal that it has simply seen dominate<br />

another individual.<br />

As far as we are aware, however, there have been no experimental<br />

studies to show that animals can infer or act upon intransitivity. Such studies<br />

could confirm the findings of Lindquist and Chase (2009) and Chase and Rohwer<br />

(1987) mentioned above and extend our knowledge of the behavioral processes<br />

leading to the formation of linear hierarchies.<br />

VII. CONCLUSION<br />

We have pointed out that there are two types of approaches that researchers can<br />

use to explain the formation of dominance relationships and dominance hierarchies<br />

in small groups of animals. The first approach uses individuals as the unit<br />

of analysis—either concentrating on differences in their traits before hierarchy<br />

formation or differences in the states that they develop during group formation.<br />

The theoretical and experimental evidence that we have reviewed indicates that<br />

explanations based upon differences in the attributes and states of individuals<br />

often work quite well to predict the outcome of dominance encounters in<br />

isolated pairs of animals. But that evidence also demonstrates, surprisingly,<br />

that these same differences in individuals are less able to predict the outcomes<br />

of relationships in socially embedded pairs or the overall ranks of individuals in<br />

their groups.<br />

In order to resolve this problem, we have suggested that we need to ask a<br />

new research question—not what determines the ranks of individuals in hierarchies,<br />

but how linear hierarchies themselves develop. As the beginning of an<br />

answer to this question, we have discussed the support for a new approach that


4. Self-Structuring Properties of Dominance Hierarchies 75<br />

uses behavioral processes to account for hierarchy structures. For some, such an<br />

approach may seem to be a kind of “cheating,” an avoidance of discovering<br />

things about individuals that really do explain their “successes” and “failures” in<br />

winning dominance contests within hierarchies. However, because the greater<br />

complexities of groups introduce an unavoidable chance element into the predictions<br />

about individuals within hierarchies, then perforce we need an approach<br />

that does not depend upon those individuals as units of analysis. A rough analogy<br />

is the way we look at the organization of tosses of a coin. Because of randomness,<br />

we do not try to predict the outcomes of individual coin tosses. Instead we move<br />

to a higher level of the phenomenon: we say something about the organization,<br />

or form of the distribution, of a great many coin flips. The behavioral process<br />

explanation of hierarchy structure is our attempt to get around the chance<br />

elements in what individuals do in hierarchies, and to say something about the<br />

organization of hierarchies in spite of that randomness.<br />

Recognizing when individual-based and process-based approaches are<br />

best applied in studies of dominance has fundamental importance in choosing<br />

the kinds of data we collect, how we analyze those data, the explanations that we<br />

develop for hierarchies, and for the cognitive capacities, for both humans and<br />

animals, that we envision as underlying dominance behavior.<br />

Acknowledgments<br />

We thank Hronn Axelsdottir and Paul St. Denis for their help with the graphics in this chapter and<br />

Robert Huber for inviting us to participate in this volume.<br />

References<br />

Addison, W. E., and Simmel, E. C. (1970). The relationship between dominance and leadership in a<br />

flock of ewes. Bull. Psychon. Soc. 15, 303–305.<br />

Bakker, T. C. M. (1985). Two-way selection for aggression in juvenile, female and male sticklebacks<br />

(Gasterosteus aculeatus), with some notes on hormonal factors. Behaviour 93, 69–81.<br />

Bakker, T. C. M. (1986). Aggression in sticklebacks (Gasterosteus aculeatus L.): A behavior genetic<br />

study. Behaviour 98, 1–144.<br />

Barchas, P. R., and Mendoza, S. P. (1984). Emergent hierarchical relationships in rhesus macaques:<br />

An application of Chase’s model. In “Social Hierarchies: Essays Toward a Sociophysiological<br />

Perspective” (P. R. Barchas, ed.), pp. 23–44. <strong>Green</strong>wood, Westport, Conn.<br />

Barkan, C. P. L., Craig, J. L., Strahl, S. D., Stewart, A. M., and Brown, J. L. (1986). Social dominance<br />

in communal Mexican Jays Aphelocoma ultramarine. Anim. Behav. 34, 175–187.<br />

Beacham, J. L. (1988). The relative importance of body size and aggressiveness as determinants of<br />

dominance in pumpkinseed sunfish, Lepomis gibbosus. Anim. Behav. 36, 621–623.<br />

Beaugrand, J., and Beaugrand, M. (1991). Prior residency and the stability of dominance relationships<br />

in pairs of green swordtail fish Xiphophorus helleri (Pisces, Poeciliidae). Behav. Process. 24,<br />

169–175.<br />

Beaugrand, J. P., and Cotnoir, P. (1996). The role of individual differences in the formation of triadic<br />

dominance orders of male green swordtail fish (Xiphophorus helleri). Behav. Process. 38, 287–296.


76 Chase and Seitz<br />

Beaugrand, J. P., Payette, D., and Goulet, C. (1996). Conflict outcome in male green swordtail fish<br />

dyads (Xiphophorus helleri): Interaction of body size, prior dominance/subordination experience,<br />

and prior residency. Behaviour 133, 303–319.<br />

Bell, A. M., Hankison, S. J., and Laskowski, K. L. (2009). The repeatability of behavior: A metaanalysis.<br />

Anim. Behav. 77, 771–783.<br />

Bergman, D. A., Kozlowski, C., McIntyre, J. C., Huber, R., Daws, A. G., and Moore, P. A. (2003).<br />

Temporal dynamics and communication of winner-effects in the crayfish, Orconectes rusticus.<br />

Behaviour 140, 805–825.<br />

Bonabeau, E., Theraulaz, G., and Deneubourg, J. L. (1999). Dominance orders in animal societies:<br />

The self-organization hypothesis revisited. Bull. Math. Biol. 61, 727–757.<br />

Bond, A. B., Kamil, A. C., and Balda, R. P. (2003). Social complexity and transitive inference in<br />

corvids. Anim. Behav. 65, 479–487.<br />

Boon, A. K., Réale, D., and Boutin, S. (2007). The interaction between personality, offspring fitness<br />

and food abundance in North American red squirrels. Ecol. Lett. 10, 1094–1104.<br />

Breithaupt, T., and Atema, J. (2000). The timing of chemical signaling with urine in dominance<br />

fights of male lobsters (Homarus americanus). Behav. Ecol. Sociobiol. 49, 67–78.<br />

Breithaupt, T., and Eger, P. (2002). Urine makes the difference: Chemical communication in<br />

fighting crayfish made visible. J. Exp. Biol. 205, 1221–1231.<br />

Brown, J. A., and Colgan, P. W. (1986). Individual and species recognition in centrachid fishes:<br />

Evidence and hypotheses. Behav. Ecol. Sociobiol. 19, 373–379.<br />

Burmeister, S. S., Jarvis, E. D., and Fernald, R. D. (2005). Rapid behavioral and genomic responses to<br />

social opportunity. PLoS Biol. 3, e363.<br />

Chase, I. D. (1982). Dynamics of hierarchy formation: Tthe sequential development of dominance<br />

relationships. Behaviour 80, 218–240.<br />

Chase, I. D. (2006). Music notation: A new method for visualizing social interaction in animals and<br />

humans. Front. Zool. 3, 18.<br />

Chase, I. D., and Rohwer, S. (1987). Two methods for quantifying the development of dominance<br />

hierarchies in large groups with applications to Harris’ sparrows. Anim. Behav. 35, 1113–1128.<br />

Chase, I. D., Bartolomeo, C., and Dugatkin, L. A. (1994). Aggressive interactions and inter-contest<br />

interval: How long do winners keep winning Anim. Behav. 48, 393–400.<br />

Chase, I. D., Tovey, C., Spangler-Martin, D., and Manfredonia, M. (2002). Individual differences<br />

versus social dynamics in the formation of animal dominance hierarchies. Proc. Natl. Acad. Sci.<br />

USA 99, 5744–5749.<br />

Chase, I. D., Tovey, C., and Murch, P. (2003). Two’s company, three’s a crowd: Differences in<br />

dominance relationships in isolated versus socially embedded pairs of fish. Behaviour 140,<br />

1193–1217.<br />

Chase, I. D., and Lindquist, W. B. (2009). Dominance hierarchies. In “Oxford Handbook of<br />

Analytical Sociology” (P. Hedström and P. Bearman, eds.), pp. 566–591. Oxford <strong>University</strong><br />

Press, New York.<br />

Cheney, D. L., and Seyfarth, R. M. (1990). How Monkeys See the World: Inside the Mind of<br />

Another Species. <strong>University</strong> of Chicago Press, Chicago.<br />

Clark, D. C. (1998). Male mating success in the presence of a conspecific opponent in a Madagascar<br />

hissing cockroach, Gromphadorhina portentosa. Ethology 104, 877–888.<br />

Clutton-Brock, T. H., Albon, S. D., and Guinness, F. E. (1984). Maternal dominance, breeding<br />

success and birth sex-ratios in red deer. Nature 308, 358–360.<br />

Collias, N. E. (1943). Statistical analysis of factors which make for success in initial encounters<br />

between hens. Am. Nat. 77, 519–538.<br />

Craig, J. V., and Muir, W. M. (1996). Group selection for adaptation to multiple-hen cages: Beak<br />

related mortality, feathering, and body weight responses. Poult. Sci. 75, 294–302.


4. Self-Structuring Properties of Dominance Hierarchies 77<br />

Craig, J. V., Ortman, L. L., and Guhl, A. M. (1965). Genetic selection for social dominance ability in<br />

chickens. Anim. Behav. 13, 114–131.<br />

Craig, J. V., Biswas, D. K., and Guhl, A. M. (1969). Agonistic behavior influenced by strangeness,<br />

crowding and heredity in female domestic fowl (Gallus gallus). Anim. Behav. 17, 498–506.<br />

Craig, J. V., Jan, M., Polley, C. R., Bhagwat, A. L., and Dayton, A. D. (1975). Changes in relative<br />

aggressiveness and social dominance associated with selection for early egg production in chickens.<br />

Poult. Sci. 54, 1647–1658.<br />

Cutts, C. J., Metcalf, N. B., and Taylor, A. C. (1999). Competitive asymmetries in territorial juvenile<br />

Atlantic salmon, Salmo salar. Oikos 86, 479–486.<br />

D’Eath, R. B., and Keeling, L. J. (2003). Social discrimination and aggression by laying hens in large<br />

groups: From peck orders to social tolerance. Appl. Anim. Behav Sci. 84, 197–212.<br />

D’Ettore, P., and Heinze, J. (2005). Individual recognition in ant queens. Curr. Biol. 15, 2170–2174.<br />

Danchin, E., Giraldeau, L. A., and Valone, T. J. (2004). Public information: From noisy neighbors to<br />

cultural evolution. Science 305, 487–491.<br />

Davis, H. (1992). Transitive Inference in Rats (Rattus norvegicus). J. Comp. Psychol. 106, 342–349.<br />

Dloniak, S. M., French, J. A., and Holekamp, K. E. (2006). Rank-related maternal effects of<br />

androgens on behavior in wild spotted hyaenas. Nature 440, 1190–1193.<br />

Dugatkin, L. A. (1997). Winner and loser effects and the structure of dominance hierarchies. Behav.<br />

Ecol. 8(6), 583–587.<br />

Dugatkin, L. A., and Earley, R. L. (2003). Group fusion: The impact of winner, loser, and by-stander<br />

effects on hierarchy formation in large groups. Behav. Ecol. 14, 367–373.<br />

Eaton, G. G. (1984). Aggression in adult male primates: A comparison of confined Japanese<br />

macaques and free-ranging olive baboons. Int. J. Primatol. 5, 145–160.<br />

Eaton, G. G., and Resko, J. A. (1974). Plasma testosterone and male dominance in a Japanese<br />

macaque (Macaca fuscata) troop compared with repeated measures of testosterone in laboratory<br />

males. Horm. Behav. 5, 251–259.<br />

Ellis, L. (1995). Dominance and reproductive success among nonhuman animals: A cross-species<br />

comparison. Ethol. Sociobiol. 16, 257–333.<br />

Fox, R. A., Ladage, L. D., Roth, T. C. I. I., and Pravosudov, V. V. (2009). Behavioural profile predicts<br />

dominance status in mountain chickadees. Poecile gambeli. Anim. Behav. 77, 1441–1448.<br />

Francis, R. C. (1984). The effects of bidirectional selection for social dominance on agonistic<br />

behavior and sex ratios in paradise fish (Macropodus opercularis). Behavior 90, 25–44.<br />

Francis, R. C. (1987). The interaction of genotype and experience in the dominance success of<br />

paradise fish (Macropodus opercularis). Biol. Behav. 12, 1–11.<br />

Francis, R. C., Soma, K., and Fernald, R. D. (1993). Social regulation of the brain—Pituitary gonadal<br />

axis. Proc. Natl. Acad. Sci. USA 90, 7794–7798.<br />

Frey, D. F., and Miller, R. J. (1972). The establishment of dominance relationships in blue gourami,<br />

Trichogaster trichopterus (Pallas). Behaviour 42, 8–62.<br />

Fuxjagera, M. J., and Marlera, C. (2010). How and why the winner effect forms: Influences of contest<br />

environment and species differences. Behav. Ecol. 21, 37–45.<br />

Gherardi, F., and Atema, J. (2005). Memory of social partners in hermit crab dominance. Ethology<br />

111(3), 271–285.<br />

Gillian, D. J. (1981). Reasoning in the chimpanzee. II. Transitive inference. J. Exp. Psychol. Anim.<br />

Behav. Process. 7, 87–108.<br />

Goessmann, C., Hemelrijk, C., and Huber, R. (2000). The formation and maintenance of crayfish<br />

hierarchies: Behavioral and self-structuring properties. Behav. Ecol. Sociobiol. 48, 418–428.<br />

Groothius, T. G. G., and Carere, C. (2005). Avian personalities: Characterization and epigenesis.<br />

Neurosci. Biobehav. Rev. 29, 137–150.<br />

Grosenick, L., Clement, T. S., and Fernald, R. D. (2007). Fish can infer social rank by observation<br />

alone. Nature 445, 429–432.


78 Chase and Seitz<br />

Hausfater, G., Altmann, J., and Altmann, S. (1982). Long-term consistency of dominance relations<br />

among female baboons (Papio-cynocephalus). Science 217, 752–755.<br />

Heinze, J. (1990). Dominance behavior among ant females. Naturwissenschaften 77, 41–43.<br />

Hemelrijk, C. K. (1999). An individual-oriented model of the emergence of despotic and egalitarian<br />

societies. Proc.R. Soc. Lond. B 266, 361–369.<br />

Hock, K., and Huber, R. (2006). Modeling the acquisition of social rank in crayfish: Winner and loser<br />

effects and self-structuring properties. Behaviour 143, 325–346.<br />

Hock, K., and Huber, R. (2007). Effects of fighting decisions on formation and structure of dominance<br />

hierarchies. Mar. Freshw. Behav. Phy. 40, 153–169.<br />

Hock, K., and Huber, R. (2009). Models of winner and loser effects: A cost-benefit analysis. Behaviour<br />

146, 69–87.<br />

Holekamp, K. E., and Smale, L. (1993). Ontogeny of dominance in free-living spotted hyenas:<br />

Juvenile rank relations with other immature individuals. Anim. Behav. 46, 451–466.<br />

Houpt, K. A., Law, K., and Martinisi, V. (1978). Dominance hierarchies in domestic horses. Appl.<br />

Anim. Ethol. 4, 273–283.<br />

Hsu, Y., and Wolf, L. L. (1999). The winner and loser effect: Integrating multiple experiences. Anim.<br />

Behav. 57, 903–910.<br />

Hsu, Y. Y., Earley, R. L., and Wolf, L. L. (2006). Modulation of aggressive behavior by fighting<br />

experience: Mechanisms and contest outcomes. Biol. Rev. 81, 33–74.<br />

Huber, R., Smith, K., Delago, A., Isaksson, K., and Kravitz, E. A. (1997). Serotonin and aggressive<br />

motivation in crustaceans: Altering the decision to retreat. Proc. Natl. Acad. Sci. USA 94,<br />

5939–5942.<br />

Johnsson, J. I., and Björnsson, B. T. (1994). Growth hormone increases growth rate, appetite and<br />

dominance in juvenile rainbow trout, Oncorhynchus mykiss. Anim. Behav. 48, 177–186.<br />

Karavanich, C., and Atema, J. (1998). Olfactory recognition of urine signals in dominance fights<br />

between male lobster, Homarus americanus. Behaviour 135(6), 719–730.<br />

Knights, B. (1987). Agonistic behaviour and growth in the European eel Anguilla anguilla L. in<br />

relation to warm-water aquaculture. J. Fish Biol. 31, 265–276.<br />

Kummer, H. (1984). From laboratory to desert and back: A social system of hamadryas baboons.<br />

Anim. Behav. 32(4), 965–971.<br />

Lai, W.-S., Ramiro, L.-L. R., Yu, H. A., and Johnston, R. E. (2005). Recognition of familiar<br />

individuals in golden hamsters: A new method and functional neuroanatomy. J. Neurosci. 25,<br />

11239–11247.<br />

Lazareva, O. F. (2004). Transitive responding in hooded crow requires linearly ordered stimuli.<br />

J. Exp. Anal. Behav. 82, 1–19.<br />

Lindquist, W. B., and Chase, I. (2009). Data-based analysis of winner-loser models of hierarchy<br />

formation in animals. Bull. Math. Biol. 71, 556–584.<br />

Lott, D. F., and Galland, J. C. (1987). Body mass as a factor influencing dominance status in<br />

American bison cows. J. Mammal. 63, 683–685.<br />

Martin, J. G. A., and Réale, D. (2008). Temperament, risk assessment and habituation to novelty in<br />

eastern chipmunks, Tamias striatus. Anim. Behav. 75, 309–318.<br />

McCarthy, I. D. (2001). Competitive ability is related to metabolic asymmetry in juvenile rainbow<br />

trout. J. Fish Biol. 59, 1002–1014.<br />

McGhee, K. E., and Travis, J. (2010). Repeatable behavioural type and stable dominance rank in the<br />

bluefin killifish. Anim. Behav. 79, 497–507.<br />

McLeman, M. A., Mendl, M., Jones, R. B., White, R., and Wathes, C. M. (2005). Discrimination of<br />

Conspecifics by Juvenile Domestic Pigs, Sus scrofa. Anim. Behav. 70, 451–461.<br />

Mendoza, S. P., and Barchas, P. R. (1983). Behavioral processes leading to linear status hierarchies<br />

following group formation in rhesus monkeys. J. Hum. Evol. 12, 185–192.


4. Self-Structuring Properties of Dominance Hierarchies 79<br />

Metcalfe, N. B., Taylor, A. C., and Thorpe, J. E. (1995). Metabolic rate, social status and life history<br />

strategies in Atlantic salmon. Anim. Behav. 49, 431–436.<br />

Möller, L. M., Beheregaray, L. B., Allen, S. J., and Harcourt, R. G. (2006). Association patterns and<br />

kinship in female Indo-Pacific bottlenose dolphins (Tursiops aduncus) of southeastern Australia.<br />

Behav. Ecol. Sociobiol. 61, 109–117.<br />

Möller, L. M., Beheregaray, L. B., Hart, R. G., and Krützen, M. (2001). Alliance membership and<br />

kinship in wild male bottlenose dolphins (Tursiops aduncus) of southeastern Australia. Proc. R.<br />

Soc. Lond. B. 268(1479), 1941–1947.<br />

Moore, A. J. (1990). The inheritance of social dominance, mating behavior and attractiveness to<br />

mates in male Nauphoeta cinera. Anim. Behav. 42, 497–498.<br />

Moore, P. A., and Bergman, D. A. (2005). The smell of success and failure: The role of intrinsic and<br />

extrinsic chemical signals on the social behavior of crayfish. Integr. Comp. Biol. 45, 650–657.<br />

Nakano, S., and Furukawa-Tanaka, T. (1994). Intra- and interspecific dominance hierarchies and<br />

variation in foraging tactics of two species of stream-dwelling chars. Ecol. Res. 9, 9–20.<br />

Nelissen, M. H. J. (1985). Structure of the dominance hierarchy and dominance determining group<br />

factors in Melanochromic auratus (Pisces, Cichlidae). Behaviour 94, 85–107.<br />

Oliveira, R. F., McGregor, P. K., and Latruffe, C. (1998). Know thine enemy: Fighting fish gather<br />

information from observing conspecific interactions. Proc. Biol. Sci. 265, 1045–1049.<br />

Oliveira, R. F., Lopes, M., Carneiro, L. A., and Canário, A. V. M. (2001). Watching fights raises fish<br />

hormone levels. Nature 409, 475.<br />

Oliveira, R. F., Silva, A., and Canário, V. M. (2009). Why do winners keep winning Androgen<br />

mediation of winner but not loser effects in cichlid fish. Proc. R. Soc. B 276(1665), 2249–2256.<br />

Øverli, Ø., Korzan, W. J., Höglund, E., Winberg, S., Bollig, H., Watt, M., Forster, G. L.,<br />

Barton, Bruce A., Øverli, E., Renner, K. J., et al. (2004). Stress coping style predicts aggression<br />

and social dominance in rainbow trout. Horm. Behav. 45, 235–241.<br />

Paz-y-Mino, G., et al. (2004). Pinyon jays use transitive inference to predict social dominance. Nature<br />

430, 778–781.<br />

Peake, T. M., and McGregor, P. K. (2004). Information and aggression in fishes. Learn. Behav. 32,<br />

114–121.<br />

Post, W. (1992). Dominance and mating success in male boat-tailed grackles. Anim. Behav. 44,<br />

917–929.<br />

Queller, D., Zacchi, F., Cervo, R., Tupillazi, S., Henshaw, M. T., Santorelli, L. A., and<br />

Strassman, J. E. (2000). Unrelated helpers in a social insect. Nature 405, 784–787.<br />

Renn, S. C. P., Aubin-Horth, N., and Hofmann, H. A. (2008). Fish and chips: Functional genomics<br />

of social plasticity in an African cichlid fish. J. Exp. Biol. 211, 3041–3056.<br />

Roberts, W. A., and Phelps, M. T. (1994). Transitive inference in rats: A test of the spatial coding<br />

hypothesis. Psychol. Sci. 5, 368–374.<br />

Robinson, G. E., Grozinger, C. M., and Whitfield, C. W. (2005). Sociogenomics: Social life in<br />

molecular terms. Nature Rev. 6, 257–270.<br />

Rutte, C., Taborsky, M., and brinkhof, M. W. G. (2006). What sets the odds of winning and losing<br />

Trends Ecol. Evol. 21, 16–21.<br />

Sactuary, W. C. (1932). A study in avian behavior to determine the nature and persistency of the<br />

order of dominance in the domestic fowl and to relate these to certain physiological reactions.<br />

Masters Thesis at the Massachusetts <strong>State</strong> College at Amherst (unpublished).<br />

Sapolsky, R. M. (1982). The endocrine stress-response and social status in wild the baboon. Horm.<br />

Behav. 16(3), 279–292.<br />

Sapolsky, R. (2005). The influence of social hierarchy on primate health. Science 308, 648–652.<br />

Sapolsky, R. M., and Share, L. J. (1994). Rank-related differences in cardiovascular function among<br />

wild baboons: Role of sensitivity to glucocorticoids. Am. J. Primatol. 32, 261–275.


80 Chase and Seitz<br />

Savin-Williams, R. C. (1980). Dominance hierarchies in groups of middle to late adolescent males.<br />

J. Youth Adolesc. 9, 75–85.<br />

Schjelderup-Ebbe, T. (1922). Contributions to the social psychology of the domestic chicken. Social<br />

Hierarchy and Dominance, Benchmark Papers in Animal Behavior, vol. 3. pp. 7–94. Hutchinson<br />

and Ross, Inc, Dowden, Stroudsburg, PA.<br />

Schjolden, J., Stoskhus, A., and Winberg, S. (2005). Does individual variation in stress responses and<br />

agonistic behavior reflect divergent stress coping strategies in juvenile rainbow trout Physiol.<br />

Biochem. Zool. 78, 715–723.<br />

Schulte-Hostedde, A. I., and Millar, J. S. (2002). ‘Little Chipmunk’ Syndrome Male Body Size and<br />

Dominance in Captive Yellow-Pine Chipmunks (Tamias amoenus). Ethology 108, 127–137.<br />

Schwanck, E. (1980). The effect of size and hormonal state on the establishment of dominance in<br />

young males of Tilapia mariae (Pisces: Cichlidae). Behav. Processes 5, 45–53.<br />

Senar, J. C. (1999). Plumage coloration as a signal of social status. Proc. Int. Ornithol. Congr 22,<br />

1669–1686.<br />

Shaw, P. (1986). The relationship between dominance behaviour, bill size and age group in Greater<br />

Sheathbills, Chionis alba. Ibis 128, 48–56.<br />

Sih, A., Bell, A. M., Johnson, J. C., and Ziemba, R. M. (2004). Behavioral syndromes: An integrative<br />

overview. Q. Rev. Biol. 79, 241–277.<br />

Sinn, D. L., and Moltschaniwskyj, N. A. (2005). Personality traits in dumpling squid (Euprymna<br />

tasmanica): Con<strong>text</strong>-specific traits and their correlation with biological characteristics. J. Comp.<br />

Pyschol. 119, 99–110.<br />

Skvoretz, J., and Fararo, T. J. (1996). Status and participation in task droups: A dynamic network<br />

model. Am. J. Sociol. 101(5), 1366–1414.<br />

Skvoretz, J., Faust, K., and Fararo, T. J. (1996). Social structure, networks, and e-state structuralism<br />

models. J. Math. Sociol. 21, 57–76.<br />

Sloman, K. A., and Armstrong, J. D. (2002). Physiological effects of dominance hierarchies:<br />

Laboratory artefacts or natural phenomena J. Fish Biol. 61, 1–23.<br />

Steirn, J. N., Weaver, J. E., and Zentall, T. R. (1995). Transitive inference in pigeons: Simplified<br />

procedures and a test of value transfer theory. Anim. Learn. Behav. 23, 76–82.<br />

Sundström, L. F., Petersson, E., Höjesjö, J., Johnsson, J. I., and Jörvi, T. (2004). Hatchery selection<br />

promotes boldness in newly hatched brown trout (Salmo trutta): Implications for dominance.<br />

Behav. Ecol. 15, 192–198.<br />

Surbeck, M., Mundry, R., and Hohmann, G. (2011). Mothers matter! Maternal support, dominance<br />

status and mating success in male bonobos (Pan paniscus). Proc. R. Soc. Lond. B 278(1705),<br />

590–598.<br />

Svartberg, K., Tapper, I., Temrin, H., Radesäter, T., and Thorman, S. (2005). Consistency of<br />

personality traits in dogs. Anim. Behav. 69, 283–291.<br />

Tibbetts, E. A. (2002). Visual signals of individual identity in the wasp, Polistes fuscatus. Proc. R. Soc.<br />

Lond. B 269, 1423–1428.<br />

Todd, J. H., Atema, J., and Bardach, J. E. (1967). Chemical communication in social behavior of a<br />

fish, the yellow bullhead (Ictalurus natalis). Science 158, 672–673.<br />

Trainor, B. C., and Hofmann, H. A. (2007). Somatostatin and somatostatin receptor gene expression<br />

in dominant and subordinate males of an African cichlid fish. Behav. Brain Res. 179, 314–320.<br />

Vannini, M., and Sardini, A. (1971). Aggressivity and dominance in river crab Potomon fluviantile<br />

(Herbst). Monitore Zool. Ital. 5, 173–213.<br />

Verbeek, M. E. M., Boon, Anne, and Drent, P. J. (1996). Exploration, aggressive behaviour and<br />

dominance in pair-wise confrontations of juvenile male great tits. Behaviour 133, 945–963.<br />

Verbeek, M. E. M., De Goede, P., Drent, P. J., and Wiepkema, P. R. (1999). Individual behavioural<br />

characteristics and dominance in aviary groups of great tits. Behaviour 136, 23–48.


4. Self-Structuring Properties of Dominance Hierarchies 81<br />

von Fersen, L., Wynne, C. D. L., and Delius, J. D. (1991). Transitive inference formation in pigeons.<br />

J. Exp. Psychol. Anim. Behav. Process. 17, 334–341.<br />

Whittingham, L. A., and Schwabl, H. (2002). Maternal testosterone in tree swallow eggs varies with<br />

female aggression. Anim. Behav. 63, 63–67.<br />

Widdig, A., Nürnberg, P., Krawczak, M., Streich, W. J., and Bercovitch, F. B. (2001). Paternal<br />

relatedness and age proximity regulate social relationships among adult female rhesus macaques.<br />

Proc. Natl. Acad. Sci. USA 98(24), 13769–13773.<br />

Wilson, E. O. (1975). Sociobiology. Harvard <strong>University</strong> Press, Cambridge.<br />

Winberg, S., and Nilsson, G. E. (1992). Induction of social dominance by L-dopa treatment in Arctic<br />

charr. Neuroreport 3, 243–246.<br />

Yeh, S., Musolf, B. E., and Edwards, D. H. (1997). Neuronal adaptations to changes in the social<br />

dominance status of crayfish. J. Neurosci. 17(2), 697–708.<br />

Yurkovic, A., Wang, O., Basu, A. C., and Kravitz, E. A. (2006). Learning and memory associated<br />

with aggression in Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 103, 17519–127524.


Intentionally left as blank


5<br />

Neurogenomic Mechanisms of<br />

Aggression in Songbirds<br />

Donna L. Maney* and James L. Goodson †<br />

*Department of Psychology, Emory <strong>University</strong>, Atlanta, Georgia, USA<br />

† Department of Biology, Indiana <strong>University</strong>, Bloomington, Indiana, USA<br />

I. Aggression in Con<strong>text</strong><br />

II. Hormonal Mechanisms of Aggression<br />

A. Territoriality in the breeding season<br />

B. Hormones and territoriality<br />

C. Aggression outside the breeding season<br />

D. Evolution of aggression and life history strategies<br />

III. Transcriptional Activity and Neural Mechanisms<br />

of Aggression in Birds<br />

A. Transcriptional traces of aggression reveal ubiquitous<br />

vertebrate themes<br />

B. Neurochemistry and major modulators<br />

IV. A Natural Model Uniting Social Behavior, Hormones, and<br />

Genetics<br />

A. The white-throated sparrow<br />

B. Endocrine and neuroendocrine correlates of<br />

behavioral polymorphism<br />

C. Causality and “phenotypic engineering”<br />

D. Mapping the ZAL2 m<br />

V. Future Directions<br />

Acknowledgments<br />

References<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00002-2


84 Maney and Goodson<br />

ABSTRACT<br />

Our understanding of the biological basis of aggression in all vertebrates, including<br />

humans, has been built largely upon discoveries first made in birds.<br />

A voluminous literature now indicates that hormonal mechanisms are shared<br />

between humans and a number of avian species. Research on genetics mechanisms<br />

in birds has lagged behind the more typical laboratory species because the<br />

necessary tools have been lacking until recently. Over the past 30 years, three<br />

major technical advances have propelled forward our understanding of the<br />

hormonal, neural, and genetic bases of aggression in birds: (1) the development<br />

of assays to measure plasma levels of hormones in free-living individuals, or “field<br />

endocrinology”; (2) the immunohistochemical labeling of immediate early gene<br />

products to map neural responses to social stimuli; and (3) the sequencing of the<br />

zebra finch genome, which makes available a tremendous set of genomic tools for<br />

studying gene sequences, expression, and chromosomal structure in species for<br />

which we already have large datasets on aggressive behavior. This combination<br />

of hormonal, neuroendocrine, and genetic tools has established songbirds as<br />

powerful models for understanding the neural basis and evolution of aggression<br />

in vertebrates. In this chapter, we discuss the contributions of field endocrinology<br />

toward a theoretical framework linking aggression with sex steroids, explore<br />

evidence that the neural substrates of aggression are conserved across vertebrate<br />

species, and describe a promising new songbird model for studying the molecular<br />

genetic mechanisms underlying aggression. ß 2011, Elsevier Inc.<br />

I. AGGRESSION IN CONTEXT<br />

Biomedical studies of aggression and its genetic basis are most often focused on<br />

pathology, yet aggressive behaviors and related agonistic displays are essential,<br />

adaptive components of social behavior that enable animals to secure and defend<br />

food, mates, and territories. For many species, aggression is also required to<br />

protect offspring from would-be predators. Thus, given that effective aggression<br />

is often essential for gene propagation, we can expect that it will be under strong<br />

selection to meet species-typical and population-specific demands. Further, for<br />

any given species, aggression will be adaptive in some con<strong>text</strong>s but not others,<br />

and it may therefore be the case that the neural and neurogenomic mechanisms<br />

of aggression vary in relation to the functional goals of the behavior. Numerous<br />

findings support this view, including evidence that parental aggression and male–<br />

male aggression are regulated by different suites of neuroendocrine mechanisms<br />

in rodents (Gammie, 2005; Trainor et al., 2008; Veenema et al., 2007) and that<br />

neuropeptides differentially influence territorial aggression and aggressive competition<br />

for mates in songbirds (Goodson and Kabelik, 2009). Indeed, the idea


5. Neurogenomic Mechanisms of Aggression in Songbirds 85<br />

that aggression is differentially regulated across distinct functional con<strong>text</strong>s, and<br />

distinct motivational states, has been around for more than 40 years (Moyer,<br />

1968). This functional perspective suggests that ethological approaches will be<br />

particularly useful for identifying integrated suites of neurogenomic mechanisms<br />

that regulate aggression in any given con<strong>text</strong> and will provide a powerful<br />

framework for distinguishing pathology from normal, adaptive variation<br />

(Blanchard and Blanchard, 2003, 2005).<br />

In this review, we focus on aggression in the con<strong>text</strong> of competition for<br />

resources, for example, defending a breeding territory or a position in a dominance<br />

hierarchy within a social group. This type of aggression, particularly in a<br />

reproductive con<strong>text</strong>, is part of a suite of related behaviors that characterize a<br />

“life history strategy” maximizing short-term gains as opposed to longer term<br />

investments (Maynard-Smith, 1977; Trivers, 1972). Short-term relationships<br />

with mates, high aggression among same-sex individuals, and low parental care<br />

typify this strategy. At the other end of this continuum is a strategy characterized<br />

by commitment to one mate, avoidance of injury, and a high level of parental<br />

care. The two strategies are difficult to employ simultaneously, resulting in a<br />

trade-off between time spent on territorial aggression versus parenting. This<br />

trade-off has become a classic principle in ethology and is universal among<br />

vertebrates, including humans (Trivers, 1972).<br />

Disruptive selection that drives the sequestration of territorial and<br />

parental behavior into alternative strategies is most likely to act on genes with<br />

widespread effects—particularly those with multiple functions. Genes encoding<br />

the action or regulation of hormones are obvious examples of such genes (Finch<br />

and Rose, 1995; Hau, 2007; Ketterson and Nolan, 1992; McGlothlin and<br />

Ketterson, 2007; Miles et al., 2007; Moore, 1991; Nijhout, 2003; Rhen and<br />

Crews, 2002; Sinervo and Svensson, 2002). A growing literature suggests that<br />

trade-offs between parenting and territorial aggression are associated with gonadal<br />

steroids; in many species of fish, birds, rodents, and primates, including humans,<br />

high levels of circulating androgens are associated with increased intrasexual<br />

competition manifested as aggression or mating effort, whereas low levels are<br />

associated with increased parenting effort (e.g., Ketterson and Nolan, 1994;<br />

McGlothlin et al., 2007). In humans, paternal care and fatherhood have been<br />

repeatedly shown to correspond to low levels of testosterone (T) (Fleming et al.,<br />

2002; Gray, 2003; Gray et al., 2002; Storey et al., 2000; Wynne-Edwards, 2001),<br />

whereas high T levels are associated with male–male aggression and competition<br />

(Bernhardt et al., 1998; Book et al., 2001; Booth et al., 1989). These opposing<br />

strategies can be generalized as a tendency to prioritize shorter term goals (mating)<br />

versus longer term goals (parental investment); at the former end of this<br />

continuum in humans, associations have been reported between T and antisocial<br />

activities such as alcoholism, drug use, reckless driving, failure to plan ahead, risktaking,<br />

and assaults (Aromaki et al., 1999; Dabbs and Morris, 1990; Udry, 1990).


86 Maney and Goodson<br />

Strategies to balance effort toward short-term versus long-term goals may therefore<br />

involve a limited number of hormones and genes. In this review, we attempt<br />

to bring together behavior, reproductive endocrinology, and genetics by focusing<br />

on species in which all three have been characterized in some detail.<br />

Since the scientific study of behavior began, birds have been the most<br />

commonly studied animals in relation to territoriality, dominance, and agonistic<br />

communication. Their popularity primarily reflects their unique accessibility—<br />

including location and use of vocal and visual communication channels—and<br />

the fact that territorial birds are readily captured using mist nets and playback of<br />

song. For the biomedical researcher attempting to model social behavior in<br />

humans, birds may seem to represent a rather distant taxonomic group. But in<br />

fact, birds have provided the test bed for some of the most influential theories in<br />

the history of aggression research, and it is no exaggeration to say that our<br />

understanding of the hormonal mechanisms of aggression in all vertebrates,<br />

including humans, has been built in large part upon discoveries that were<br />

first made in birds (Archer, 2006; Goodson et al., 2005a,b,c; Konishi et al.,<br />

1989). For example, pioneering studies in birds established the theoretical<br />

framework currently used by researchers to understand how hormones mediate<br />

a trade-off between aggression and parenting in mammals (Wingfield et al.,<br />

1990). This theoretical framework, which has been called the “challenge<br />

hypothesis,” is based on the idea that the role of gonadal steroids in aggression<br />

is modulated by social con<strong>text</strong>. It predicts that when males are challenged in a<br />

reproductive con<strong>text</strong>, T levels rise to facilitate territorial aggression and suppress<br />

parental behavior. Since it was first proposed by Wingfield et al. (1990), the<br />

challenge hypothesis has found support in a wide variety of nonavian vertebrate<br />

taxa including fish, reptiles, and primates, including humans (reviewed by<br />

Archer, 2006). The parallels between songbirds (particularly New World sparrows)<br />

and humans with regard to the social modulation of gonadal steroids and<br />

their effects on aggressive and parental behavior are voluminous and are summarized<br />

in Table 5.1. The underlying neuroendocrine mechanisms are nearly<br />

identical in birds and humans and are based on the function of the hypothalamopituitary-gonadal<br />

(HPG) axis, which is universally recognized as being highly<br />

conserved across all vertebrates (reviewed by Adkins-Regan, 2005).<br />

Despite the contributions of avian research to our understanding of<br />

human behavior, genomic resources in birds have lagged well behind those in<br />

mammals—although this situation is now rapidly changing. In the sections that<br />

follow, we first explore the neural and endocrine literature on songbird aggression,<br />

and then describe a relatively new research program that is focused on the neurogenomics<br />

of territorial aggression in white-throated sparrows (Zonotrichia albicollis),<br />

a species that exhibits morphological and behavioral polymorphisms associated<br />

with a chromosomal inversion (Thomas et al., 2008; Thorneycroft, 1975). Importantly,<br />

the morphs differ not only in their territorial aggression, but also in parental


5. Neurogenomic Mechanisms of Aggression in Songbirds 87<br />

Table 5.1. Evidence of Shared Mechanisms of Competitive Aggression in Birds and Humans<br />

Prediction Evidence in New World sparrows Evidence in humans<br />

Males respond to competition<br />

with increased plasma T<br />

The plasma T response to<br />

challenge increases<br />

aggression<br />

Plasma T levels are lower<br />

among paternal males<br />

Aggressive dominance is<br />

correlated with plasma<br />

T levels<br />

Plasma T is associated with<br />

alternative life history<br />

strategies regarding<br />

territoriality versus<br />

parenting<br />

Wingfield (1985), Wingfield and<br />

Hahn (1994), Wingfield and<br />

Wada (1989), Wingfield et al.<br />

(1990)<br />

Archawaranon et al. (1991),<br />

Wingfield (1984b, 1994b)<br />

Wingfield (1984a), Wingfield<br />

and Farner (1978), Wingfield<br />

and Goldsmith (1990),<br />

Wingfield et al. (1990)<br />

Archawaranon and Wiley<br />

(1988), Schlinger (1987),<br />

Wiley et al. (1993)<br />

Hau (2007), Ketterson and<br />

Nolan (1992), McGlothlin<br />

et al. (2007), Schoech et al.<br />

(1998), Spinney et al. (2006),<br />

Wingfield (1984a,b,c)<br />

Meta-analysis of 23 studies in<br />

Archer (2006)<br />

Meta-analysis of 11 studies in<br />

Archer (2006)<br />

Berg and Wynne-Edwards<br />

(2001), Fleming et al.<br />

(2002), Gray et al. (2002),<br />

Storey et al. (2000)<br />

Meta-analysis of 13 studies in<br />

Archer (2006)<br />

Dabbs and Morris (1990),<br />

Dabbs et al. (1997),<br />

Daitzman and Zuckerman<br />

(1980), Gray et al. (2002),<br />

Julian and McHenry (1989)<br />

The endocrine underpinnings of competitive aggression are broadly similar in New World<br />

sparrows (here limited to the Zonotrichia, Melospiza, and Junco genera) and humans (based primarily<br />

on Archer, 2006). Only a fraction of the relevant literature is cited here.<br />

behavior, and thus this species offers an extraordinary opportunity to examine<br />

neurogenomic mechanisms that integrate aggression with other aspects of social<br />

phenotype and con<strong>text</strong>-specific behavior.<br />

II. HORMONAL MECHANISMS OF AGGRESSION<br />

A. Territoriality in the breeding season<br />

There are about 10,000 species of birds, almost half of which are songbirds.<br />

Territoriality runs the gamut, with members of some species nesting colonially or<br />

cooperatively, others defending territories of several hectares. Perhaps the beststudied<br />

territorial species are the seasonally breeding New World sparrows,<br />

which include song sparrows (Melospiza melodia), field sparrows (Spizella pusilla),<br />

white-crowned sparrows (Zonotrichia leucophrys), and dark-eyed juncos (Junco<br />

hyemalis; see Arcese et al., 2002; Chilton et al., 1995; Carey et al., 2008; Nolan<br />

et al., 2002 for reviews). In migratory populations, the males arrive at the


88 Maney and Goodson<br />

breeding grounds a week or so before the females and stake out territories<br />

containing food sources and nest sites. The females then arrive, basing their<br />

mate choices on the quality of the males as well as their territories. It is therefore<br />

important, in fact critical, for males to establish high-quality territories early in<br />

the breeding season. Once a male has attracted a mate, she will help defend the<br />

territory.<br />

The most ubiquitous and frequent behavior used for territory defense by<br />

songbirds is, not surprisingly, song. Each species’ song is distinct, and within a<br />

species there is enough variation that individuals can recognize each other’s<br />

songs (Krebs, 1971). Some species sing different types of song in different<br />

con<strong>text</strong>s; for example, the “complex song” of the field sparrow is considered<br />

more aggressive than the “simple song” (Carey et al., 2008; Nelson and Croner,<br />

1991), and the chestnut-sided warbler (Dendroica pensylvanica) sings a different<br />

song to an intruder than he does to a potential mate (Kroodsma et al., 1989; Lein,<br />

1978). Although most of the singing is done by males, females of some species do<br />

sing during agonistic encounters (e.g., Baptista et al., 1993; Falls and Kopachena,<br />

2010). Males typically choose a centrally located perch and sing loudly at regular<br />

intervals, making their presence known to would-be mates and intruders. In a<br />

now-classic study, Krebs (1976) showed that experimental removal of territorial<br />

male great tits (Parus major) resulted in rapid takeover of the vacated territories<br />

by other males; however, broadcasting a former resident’s song from a loudspeaker<br />

in his territory significantly delayed that takeover (see also Falls, 1988).<br />

Although most song is typically sung from a prominent perch in the center, it<br />

is also commonly used near territory boundaries, particularly directed at neighbors,<br />

as the territory is established. Males learn to recognize their neighbors’<br />

songs and will tolerate them at a distance; however, hearing an unfamiliar song<br />

will generally trigger an investigation and aggressive response (Falls, 1969;<br />

Goldman, 1973, Krebs, 1971; Kroodsma, 1976).<br />

In addition to song, territorial sparrows are likely to exhibit a number of<br />

other displays during territory establishment and maintenance. Birds of both<br />

sexes may puff out their feathers, in particular raising those on the head to form a<br />

crest, or quiver their wings while pointing a closed bill at the intruder<br />

(Elekonich, 2000; Nice, 1943). They may peck furiously at nearby objects. If<br />

the intruder is unfazed, the resident then resorts to more direct physical threats,<br />

flying directly over the intruder, chasing him, and eventually attacking him.<br />

Opponents may fly at each other with feet stretched forward and may even fall to<br />

the ground as they engage and struggle. Physical fights are rare, however, and<br />

generally limited to the early breeding season before territory boundaries are<br />

firmly established.<br />

Territorial responses can be studied in the field by observing naturally<br />

occurring behavior or by staging a “simulated territorial intrusion” (STI). In this<br />

procedure, experimenters place a decoy “intruder,” often accompanied by song


5. Neurogenomic Mechanisms of Aggression in Songbirds 89<br />

played through a loudspeaker, onto a resident’s territory; the resident’s behavioral<br />

response is then quantified. Taxidermic or painted models may be used as decoys,<br />

or a live, unfamiliar male in cage may be presented. The most robust responses are<br />

obtained by presenting both decoy and recorded song so that the resident receives<br />

both visual and auditory cues (Wingfield and Wada, 1989). The behavioral data<br />

that are collected typically include latency to respond, songs, flights directed at<br />

the decoy threat displays (e.g., wing quivers), distance from the decoy at the<br />

closest approach, and the time spent within a certain distance, for example 5 m,<br />

of the decoy (Wingfield, 1984b, 1985; Wingfield and Hahn, 1994).<br />

B. Hormones and territoriality<br />

In the 1970s and 1980s, John Wingfield and Donald Farner revolutionized the<br />

study of behavior in songbirds by developing methods to measure gonadal<br />

hormones in small plasma samples collected from free-living individuals<br />

(Wingfield and Farner, 1976). The ensuing research in “field endocrinology”<br />

(Wingfield et al., 1990; Walker et al., 2005) elucidated patterns of gonadal<br />

hormone secretion over the reproductive cycle in a wide variety of avian species.<br />

In general, the stages of breeding associated with high levels of aggression<br />

coincide with high plasma levels of T. In song sparrows, for example T peaks<br />

during territory establishment when agonistic encounters are most frequent<br />

(reviewed by Wingfield et al., 2001), rises again during egg-laying, and slowly<br />

declines until the incubation phase when territory disputes are rare (Fig. 5.1A).<br />

In some multiple-brooded species, for example house sparrows (Passer domesticus),<br />

competition for nest holes appears to drive an increase in plasma T during<br />

each egg-laying period (Hegner and Wingfield, 1986; Fig. 5.1B).<br />

The temporal correlation between high plasma T and territorial behavior<br />

suggests that the two are related, and experimental manipulation of either T<br />

or the competitive environment shows that the relationship is bidirectional.<br />

Male song sparrows implanted subcutaneously with T-filled silastic capsules<br />

during territory establishment showed a more aggressive response to STI than<br />

males given empty capsules, and won territories that were twice the size<br />

(Wingfield, 1984b,c). Perhaps more interesting, however, was the effect on<br />

these males’ neighbors. The residents occupying territories adjacent to the<br />

treated males also showed increases in plasma T, suggesting that having to<br />

defend their territories against their more aggressive, T-treated neighbors stimulated<br />

HPG activity. In a separate study, male song sparrows were removed from<br />

their territories, allowing new residents to take over. In this case, both the new<br />

residents and their neighbors experienced high T levels compared with unmanipulated<br />

controls (Wingfield et al., 1987). Together, these studies show not only<br />

that T increases aggression, but also that engaging in agonistic encounters<br />

increases plasma T. The HPG response is rapid; plasma T rises significantly


90 Maney and Goodson<br />

A<br />

Song sparrow<br />

monogamous<br />

Prebreeding<br />

Sexual<br />

Parental<br />

Plasma level of testosterone<br />

B<br />

Prebreeding<br />

House sparrow<br />

monogamous<br />

Sexual<br />

Parental<br />

Sexual<br />

Parental<br />

Sexual<br />

Parental<br />

C<br />

Red-winged blackbird<br />

polygynous<br />

Prebreeding<br />

Breeding<br />

Figure 5.1. Plasma testosterone (T) profiles over the breeding season in males of three passerine<br />

species. (A) In song sparrows, T peaks during territory establishment (prebreeding) and<br />

again during laying of the first clutch when females are receptive (sexual), and then falls<br />

during incubation and feeding (parental). (B) In house sparrows, T peaks during periods<br />

of intense competition for nest sites, prior to each of multiple broods. (C) In red-winged<br />

blackbirds (Agelaius phoeniceus), males provide little parental care and spend more time<br />

on territorial defense; T remains relatively high until the end of the breeding season.<br />

Redrawn from data in Wingfield (1984a) and Hegner and Wingfield (1986).<br />

within 10 min of STI onset in male song sparrows (Wingfield et al., 1987) and<br />

can remain high for several days. This prolonged hormonal response probably<br />

heightens vigilance in anticipation of a sustained challenge (Wingfield, 1994a).


C. Aggression outside the breeding season<br />

In seasonally breeding birds, the gonads regress and become quiescent during the<br />

nonbreeding season. A testis that measures more than 10 mm across during<br />

breeding may shrink to less than 1 mm in the fall. Ovaries likewise regress such<br />

that follicles are barely visible. As a result, plasma levels of gonadal steroids fall<br />

to low or even nondetectable levels (Wingfield and Farner, 1993). The gonads<br />

remain in this state until the long days of spring stimulate photoreceptors in<br />

the mediobasal hypothalamus, triggering HPG hormone secretion and gonadal<br />

recrudescence (reviewed by Dawson et al., 2001).<br />

For many species, the fall in plasma gonadal steroid levels at the end of<br />

the breeding season coincides with abandonment of breeding territories and the<br />

onset of flocking behavior. For others, quiescence of the HPG axis does not seem<br />

to affect territorial behavior at all. Both of these scenarios are considered below.<br />

1. Aggression in flocks<br />

5. Neurogenomic Mechanisms of Aggression in Songbirds 91<br />

In species that do not defend territories year-round, the conclusion of territoriality<br />

each year may give rise to flocking behavior. In flocks, birds maximize foodfinding<br />

while minimizing predation risk (Hamilton, 1971). Competition for food,<br />

roosting sites, and other resources within flocks creates many opportunities for<br />

agonistic interactions, and some of the same behaviors used to defend territories,<br />

for example, song and threat displays, are also seen in this con<strong>text</strong>. Other<br />

common aggressive displays in flocks include displacements, wherein one individual<br />

approaches another and causes it to move away, and hold-offs, wherein an<br />

individual refuses to be displaced. Each of these behaviors is easily observed in<br />

free-living groups, for example, in the vicinity of a popular food source (e.g.,<br />

Ficken et al., 1978; Harrington, 1973; Rohwer and Rohwer, 1978), or in captivity<br />

(e.g., Schlinger, 1987; Watt, et al., 1984).<br />

In some species, winter flocks adopt a highly organized social structure.<br />

Many of us are familiar, for example, with the dominance hierarchies, or “pecking<br />

order,” established by chickens (Gallus gallus domesticus; Allee, 1936, 1942).<br />

Similar social arrangements have been observed in wild and captive groups of<br />

sparrows, for example, dark-eyed juncos (Junco hyemalis; Sabine, 1959), whitethroated<br />

sparrows (Schneider, 1984; Watt et al., 1984), Harris sparrows (Zonotrichia<br />

querula; Rohwer, 1975) and to a lesser extent, song sparrows (Nice, 1943).<br />

Within a group, each pair of individuals has a stable relationship such that one is<br />

dominant to the other. The subordinate will allow the dominant to displace it,<br />

deferring access to resources, and the dominant is unlikely to tolerate the close<br />

proximity of the subordinate. When all members of the group are considered<br />

together, there is in most cases a linear order of dominance; in other words, there<br />

is an alpha bird that dominates all others, a beta that dominates all but the alpha,<br />

and so on down to a bird that is subordinate to all. Exceptions, such as triangular


92 Maney and Goodson<br />

relationships, are fairly common. Reversals, in which the hierarchy is challenged<br />

and altered, do occur, but overall the arrangement is fixed and stable (Sabine,<br />

1959). In abiding by this social structure, the members of the group avoid the<br />

potential injuries and high energy expenditure that would result from constant<br />

aggressive encounters; when the hierarchy is stable, actual fighting is extremely<br />

rare. Only newcomers are subject to aggressive behavior, which subsides as they<br />

are assimilated into the group and assume a fixed rank (Tompkins, 1933).<br />

Whereas physical contact and escalated fights are not normally required<br />

for the maintenance of a stable hierarchy, its initial establishment is associated<br />

with frequent aggressive interactions. When unfamiliar birds are forming a<br />

group, or when newcomers arrive, rank is settled by the outcome of agonistic<br />

encounters. Age and sex can predict rank in some cases. For example in whitecrowned<br />

sparrows and related species, males tend to dominate females and older<br />

birds dominate younger ones such that the hierarchy within a group follows the<br />

general rule: adult males>adult females>immature males>immature females<br />

(Keys and Rothstein, 1991). In some species, such as house sparrows and Harris<br />

sparrows, plumage characteristics can predict rank; dominant individuals have<br />

dark “bibs” that appear to serve as status signals (Møller, 1987; Rohwer, 1975).<br />

Manipulation of bib coloration does not, however, alter status; rather, it results in<br />

heightened aggression toward the altered individuals, whose new plumage coloration<br />

is inconsistent with their behavior (Rohwer and Rohwer, 1978).<br />

The relationships between dominance rank and HPG function have<br />

been explored in many species of birds. This body of work, which spans almost 75<br />

years, collectively shows that dominance rank is predicted by plasma T levels<br />

only in groups that are newly forming. In other words, when unfamiliar birds<br />

come together to form a social group, their T levels during the establishment of<br />

the hierarchy contribute toward their eventual rank. Later, after the hierarchy is<br />

settled and stable, rank is unrelated to plasma T (Baptista et al., 1987; Buchanan<br />

et al., 2010; Chase, 1982; Ramenofsky, 1984; Schlinger, 1987; Wiley et al., 1999).<br />

Given that engaging in aggressive behavior causes T to rise (Wingfield<br />

et al., 1987), we must ask whether high T leads to the acquisition of a high rank,<br />

or vice versa. Some authors have reported that short-term treatment with<br />

exogenous T can alter stable hierarchies; the newly acquired ranks were maintained<br />

in white-crowned sparrows (Baptista et al., 1987) and domestic hens<br />

(Allee et al., 1939) but not Japanese quail (Coturnix coturnix japonica; Selinger<br />

and Bermant, 1967). In the majority of such studies, T treatment affects eventual<br />

rank when it is done early during hierarchy establishment. After rank is stable,<br />

however, rank is usually unaffected by T administration (Buchanan et al., 2010;<br />

Crook and Butterfield, 1968; Lumia, 1972; Mathewson, 1961; Rohwer and<br />

Rohwer, 1978; Wiley et al., 1999). The lack of an effect of T treatment on<br />

established, stable ranks has been attributed to learning, or “social inertia”<br />

(Guhl, 1968; Wiley et al., 1999), in groups of individuals that are familiar with


5. Neurogenomic Mechanisms of Aggression in Songbirds 93<br />

each other. In house sparrows, which do not defend large breeding territories,<br />

dominance rank during the nonbreeding season is determined largely by rank at<br />

the end of the breeding season when plasma T level is falling (Buchanan et al.,<br />

2010). In other words, winners stay winners and losers stay losers (Chase, 1982).<br />

HPG activity during the breeding season may therefore contribute toward rank<br />

during the nonbreeding season. During both times of year, actual physical aggression<br />

is limited to periods of instability during which high-ranking positions up for<br />

grabs or otherwise in dispute. After the establishment of social relationships and<br />

boundaries, physical aggression is rare and plasma levels of T are much lower.<br />

2. Territoriality in the nonbreeding season<br />

Song sparrows in resident populations, despite being seasonal breeders that<br />

undergo gonadal regression in the fall, can maintain essentially the same<br />

territories year-round. Whereas the song sparrows studied by Wingfield in<br />

New York <strong>State</strong> (e.g., Wingfield, 1984a,b,c, 1985) abandon their territories<br />

and migrate at the conclusion of the breeding season, populations in Western<br />

Washington remain on the same territories, molt their feathers, and then,<br />

despite having completely regressed gonads and nondetectable levels of T,<br />

resume territorial defense (Wingfield and Hahn, 1994). Furthermore, young<br />

males just hatched the previous spring can establish new territories in the fall<br />

without an increase in T. STI during the fall induces the same behavioral<br />

responses as in spring, but without an accompanying HPG response (Soma and<br />

Wingfield, 2001; Wingfield and Hahn, 1994), and castration does not interfere<br />

with the ability to maintain a territory (Wingfield, 1994b). These results led to<br />

the hypothesis that territorial aggression and gonadal steroid secretion can<br />

become uncoupled in this and other species that defend territories outside<br />

the breeding season.<br />

To address this question, Soma et al. (1999, 2000a,b) tested whether T<br />

action is necessary for autumnal aggression in a population of song sparrows in<br />

Western Washington. To block the effects of T, they administered androgen<br />

receptor antagonists and, because T can also act via conversion to estradiol (E2),<br />

blockers of that conversion. They found that blocking the action of aromatase,<br />

an enzyme that converts T to E2, reduced territorial aggression even in the fall<br />

when plasma levels of gonadal steroids are naturally very low. This result<br />

suggested that autumnal aggression is not in fact independent of steroid hormones.<br />

This reduction was reversed by treatment with E2 (Soma et al., 2000a).<br />

Because gonads are not required for autumnal territorial defense<br />

(Wingfield, 1994b), the E2 that drives this behavior must come from a nongonadal<br />

source. One such source may be the brain itself, which contains high<br />

levels of aromatase. In song sparrows, aromatase mRNA is expressed in the brain<br />

at all times of year (Soma et al., 2003; Wacker et al., 2010) suggesting a possible


94 Maney and Goodson<br />

source of E2 available to brain tissue year-round. Aromatase activity in the<br />

ventral telencephalon, which contains the putative avian homologue of the<br />

amygdala, is reduced during molt, at which time aggression is also low (Soma<br />

et al., 2003). Brain-generated E2 may thus be an important regulator of territorial<br />

aggression in this species. To synthesize E2, the brain may make use of androgen<br />

precursors from regressed gonads or the adrenals (see Soma, 2006; Soma et al.,<br />

2008 for reviews). Alternatively, some regions of the brain contain all of the<br />

enzymes necessary to synthesize E2 de novo from cholesterol substrate (reviewed<br />

by Soma et al., 2008; Remage-Healey et al., 2010), obviating the need for a<br />

peripheral steroid synthesis altogether. The regions of the brain important for<br />

aggressive behaviors, for example the nuclei that control singing behavior, are<br />

rich in these enzymes (Remage-Healey et al., 2010). Aromatase expression is<br />

high in the ventromedial nucleus of the hypothalamus (VMH) during all times of<br />

year except during molt, when aggression is virtually absent (Wacker et al.,<br />

2010). In mice, distinct populations of VMH neurons contribute to fighting<br />

and mating behavior (Lin et al., 2011). In Section III, we will explore further<br />

the role of this region in aggression in songbirds.<br />

Like androgen release from the gonad, E2 synthesis by the brain appears to<br />

be behaviorally regulated. Remage-Healey et al. (2008)recently showed that in<br />

zebrafinches(Taeniopygia guttata), hearing conspecific song increases E2 concentrations<br />

in the auditory forebrain within minutes. Pradhan et al. (2010)showed<br />

subsequently that in nonbreeding, territorial song sparrows, exposure to STI rapidly<br />

increases activity of 3b-hydroxysteroid dehydrogenase, an enzyme necessary to<br />

synthesize E2 from androgen precursors. These findings demonstrate that the steroid<br />

environment within the brain is dynamic, sensitive to the social environment, and<br />

more independent of the gonad than previously thought. These discoveries of<br />

socially regulated, brain-generated steroid synthesis challenge the traditional view<br />

of hormone-mediated aggression and highlight the importance of songbird models<br />

to our understanding of how steroids regulate gene activity in the brain.<br />

Forty years of research on free-living sparrows has shown that aggression<br />

depends on steroid hormones. Even when at first glance it appears that aggression<br />

and steroid hormones have come “uncoupled,” for example, when resident song<br />

sparrows vigorously defend territories despite low plasma T, the evidence shows<br />

that low levels are necessary for the expression of territoriality and that the brain<br />

itself may produce sufficient amounts. The role of steroid hormones, particularly<br />

E2, in aggression seems very similar to their role in sexual receptivity (reviewed<br />

by Maney, 2010): plasma levels need not be high, and in fact seasonal peaks in<br />

plasma levels need not be associated in time with the behavior. A low level,<br />

however, is required for the behavior to be expressed. Because the frequency of<br />

aggressive behavior is clearly not always correlated directly with plasma levels of<br />

steroid hormones, it is possible that the hormones play a priming, or permissive,<br />

role and that other hormones or neurotransmitters are also important.


5. Neurogenomic Mechanisms of Aggression in Songbirds 95<br />

D. Evolution of aggression and life history strategies<br />

Research on wild songbirdshasdemonstrated a robust, two-wayrelationship between<br />

aggression and HPG activity. What does that finding tell us about the evolution and<br />

genetic control of these behaviors Because steroid hormones affect suites of behaviors,<br />

not just aggression, it is helpful to think about this issue in terms of behavioral<br />

strategies. As discussed in Section I above, investment in territory defense and matefinding<br />

defines a strategy at one end of a behavioral continuum, with investment in<br />

survival and parenting at the other (Trivers, 1972). This trade-off appears to be<br />

mediated, at least in part, by HPG activity (Ketterson and Nolan, 1994; McGlothlin<br />

et al., 2007; Wingfield et al.,1990).In specieswith maleparentalcare,Tis high during<br />

territory establishment but falls during the parental phase (Fig. 5.1A, B). Exogenous<br />

administration of T during the parental phase inhibits parental behavior and<br />

increases territorial behavior (Hegner and Wingfield, 1987; Schoech et al., 1998;<br />

Silverin, 1980). In species without male parental care, for example polygynous<br />

species in which females do the bulk of the care, T remains high in males for the<br />

duration of the season (Fig. 5.1C). In a study by Wingfield (1984c), T treatment of<br />

male song sparrows not only reduced parental care but also induced polygyny in this<br />

normally monogamous species. These males spent much of their time singing and<br />

acquired huge territories, attracting multiple females. Their failure to provision the<br />

young, however, likely reduced their overall reproductive success (Hegner and<br />

Wingfield, 1987; Silverin, 1980). Alterations in HPG function could therefore result<br />

in large, cascading effects and make an important contribution to variation in social<br />

behavior and social strategies (Ketterson and Nolan, 1992; Sinervo and Svensson,<br />

2002). When the effects of a hormone are antagonistic with respect to behavior, for<br />

example, in the case of T and parenting versus aggression, “antagonistic pleiotropy”<br />

can give rise to behavioral trade-offs (Finch and Rose, 1995). We hypothesize that<br />

suites of related genes, the expression of which is tightly governed by social cues, may<br />

act hierarchically to organize and regulate the hormonal and neural systems that<br />

promote territorial aggression and reduce parental behavior. In the next section, we<br />

explore the neural circuits that are involved and consider how evolution has shaped<br />

them in species with different behavioral strategies.<br />

III. TRANSCRIPTIONAL ACTIVITY AND NEURAL MECHANISMS<br />

OF AGGRESSION IN BIRDS<br />

A. Transcriptional traces of aggression reveal ubiquitous<br />

vertebrate themes<br />

Although it has long been known that all vertebrates share some basic features in<br />

the organization of the amygdala, hypothalamic nuclei, and associated (limbic)<br />

areas of the basal forebrain and midbrain, the extent of these similarities has


96 Maney and Goodson<br />

become much more clear in the past 10 years as investigators have combined<br />

genomic data with conventional neuroanatomical and functional approaches<br />

(e.g., using lesions and pharmacological manipulations). We now know that<br />

birds and rodents exhibit extraordinary similarities in the organization of limbic<br />

brain areas (Goodson, 2005; Newman, 1999), including distinct homologies at<br />

the subnuclear level (e.g., Goodson et al., 2004a; Kingsbury et al., 2011) and very<br />

specific similarities in the topographical patterns of transcriptional response to<br />

aggressive interactions (and other social interactions, as well), as established<br />

through the experimental induction of immediate early gene (IEG) transcripts<br />

and their protein products (Ball and Balthazart, 2001; Goodson, 2005). The IEGs<br />

most commonly used for such functional studies are c-fos and a gene variably<br />

known as zif-268, egr-1, NGFI-A, krox-24,orZenk (the latter being a name often<br />

used in birds as an acronym for the other four names; Mello et al., 1992).<br />

Experimentally induced increases in IEG mRNA can be detected by<br />

in situ hybridization (ISH) within 15–30 min. For detection of IEG proteins<br />

by immunocytochemistry (ICC), most investigators harvest brain tissue<br />

60–90 min after the experimental manipulation, such as an aggressive interaction.<br />

Because the induced IEG proteins are still elevated at 90 min and two halflives<br />

of the protein have passed (Herdegen and Leah, 1998), it is possible to<br />

determine whether the experimental manipulation may have decreased IEG<br />

activity from control levels (e.g., Bharati and Goodson, 2006; Goodson and<br />

Wang, 2006). Most behavioral neuroscientists are primarily interested in IEGs<br />

not because of what they do inside the cell, but rather because they provide a<br />

proxy marker to indicate that a cell has responded in some way to a stimulus.<br />

That response may or may not be associated with action potentials and release of<br />

neurochemicals (Herdegen and Leah, 1998), but by labeling for IEG transcripts<br />

and proteins, investigators can gain a good idea about the functional properties of<br />

different brain areas or cell groups. The actual molecular functions of IEGs are<br />

varied, but typically include the regulation of other genes involved in experience-dependent<br />

neuroplasticity (Herdegen and Leah, 1998; Mello and Ribeiro,<br />

1998), and thus IEGs probably have the ultimate effect of changing the way that<br />

cells function and behave during subsequent behavioral interactions.<br />

In rodents, resident–intruder encounters induce IEG activity in a characteristic<br />

pattern that includes the medial amygdala (MeA), medial bed nucleus<br />

of the stria terminalis (BSTm, a component of the medial extended amygdala<br />

that shares many anatomical and functional properties with the MeA), anterior<br />

hypothalamus (AH), ventrolateral lateral septum (LS), ventrolateral subdivision<br />

of the ventromedial nucleus of the hypothalamus (VMHvl; or lateral VMH in<br />

hamsters), dorsal premammillary nucleus, and the dorsal midbrain periaqueductal<br />

gray (Kollack-Walker et al., 1997; Motta et al., 2009). Notably, along with the<br />

medial preoptic area, these same brain areas are central to the regulation of most<br />

other social behaviors, including communication behaviors, parental care, pair


5. Neurogenomic Mechanisms of Aggression in Songbirds 97<br />

bonding, appetitive and consummatory sexual behavior, juvenile play, social<br />

recognition, and both same-sex and opposite-sex affiliation (Goodson, 2005;<br />

Newman, 1999). Despite the similarities, the relative amount of IEG activity<br />

across the different nodes of this “social behavior network” is distinctive for each<br />

social con<strong>text</strong>, suggesting that functional relationships across the network nodes<br />

are dynamic and con<strong>text</strong>-specific. Distinct patterns of correlated activity between<br />

network nodes in different social con<strong>text</strong>s have now been demonstrated in<br />

multiple vertebrate classes (Crews et al., 2006; Hoke et al., 2005; Yang and<br />

Wilczynski, 2007).<br />

The areas comprising the social behavior network are readily identified<br />

in birds and are anatomically and functionally conserved across amniote vertebrates,<br />

and in fact, the basic features of this network are present even in fish<br />

(Goodson, 2005; Goodson and Bass, 2002). Consistent with this conservation,<br />

territorial songbirds housed in captivity exhibit a pattern of IEG activity after<br />

STI that is virtually identical to the pattern described for rodents after a<br />

resident–intruder encounter (Goodson and Evans, 2004; Goodson et al.,<br />

2005b). This work has been conducted in animals housed in their natural<br />

habitat, and data for catecholaminergic midbrain areas are even available from<br />

animals occupying natural territories (Maney and Ball, 2003). In addition,<br />

following exposure to same-sex conspecifics through a wire barrier in a quiet<br />

room (which elicits very little overt behavior), territorial finches exhibit relatively<br />

greater Fos and/or egr-1 responses than do gregarious species in a pattern<br />

similar to aggressive encounters (Goodson et al., 2005a). Hence, at least to an<br />

extent, the IEG activity of these brain areas reflects perceptual or motivational<br />

processes, not simply activation of aggression.<br />

Although it is intuitive to interpret IEG induction as reflecting a<br />

positive relationship between a brain area and behavior, negative correlations<br />

between IEG cell counts and aggressive behavior are observed for multiple brain<br />

areas. These include the AH, both pallial and subpallial subdivisions of the LS,<br />

and the paraventricular nucleus of the hypothalamus (PVN; Fig. 5.2; Goodson<br />

et al., 2005b). This pattern of results suggests that aggression is under inhibitory<br />

control, at least by some areas. Consistent with this idea, lesions of the LS<br />

increase resident–intruder aggression in male field sparrows and pigeons<br />

(Columba livia; Goodson et al., 1999; Ramirez et al., 1988). Note, however, that<br />

such effects are not observed in some con<strong>text</strong>s, such as aggressive competition for<br />

mates in male zebra finches, a highly gregarious species (Goodson et al., 1999).<br />

The PVN is heavily interconnected with the social behavior network<br />

and plays an important role in the regulation of autonomic and pituitary activity<br />

in relation to behavioral state. A subset of cells in the PVN produce arginine<br />

vasotocin (VT; homologue and evolutionary precursor of mammalian arginine<br />

vasopressin, VP), and the percentage of those cells that express Fos is also<br />

negatively correlated with aggressive response to an STI in male song sparrows


98 Maney and Goodson<br />

In contacts<br />

In contacts<br />

In contacts<br />

A<br />

B<br />

6<br />

6<br />

6<br />

LSc.v<br />

LSc.d<br />

5<br />

r = -0.571 LSc.vl<br />

5<br />

r = -0.688 5<br />

r = -0.534<br />

P = 0.020<br />

P = 0.003<br />

P = 0.033<br />

4<br />

3<br />

2<br />

1<br />

0<br />

-1<br />

4<br />

3<br />

2<br />

1<br />

0<br />

-1<br />

4<br />

3<br />

2<br />

1<br />

0<br />

-1<br />

0 5 10 15 20 25 30 0 5 10 15 20 25 30 0 5 10 15 20 25 30<br />

D<br />

Fos-ir nuclei/100 mm 2 Fos-ir nuclei/100 mm 2 Fos-ir nuclei/100 mm 2<br />

E<br />

F<br />

6<br />

6<br />

6<br />

PVN<br />

5<br />

r = -0.632 AH LSr<br />

5<br />

r = -0.478 5<br />

r = -0.530<br />

P = 0.008<br />

P = 0.060<br />

P = 0.034<br />

4<br />

4<br />

4<br />

3<br />

3<br />

3<br />

2<br />

2<br />

2<br />

1<br />

1<br />

1<br />

0<br />

0<br />

0<br />

-1<br />

-1<br />

-1<br />

0 5 10 15 20 25 30 0 5 10 15 20 25 30 0 2 4 6 8 10<br />

Fos-ir nuclei/100 mm 2 Fos-ir nuclei/100 mm 2 Zenk-ir nuclei/100 mm 2<br />

G<br />

H<br />

I<br />

6<br />

6<br />

6<br />

LSc.vl<br />

5<br />

r = -0.693 LSc.l PVN<br />

5<br />

r = -0.551 5<br />

r = -0.573<br />

P = 0.002<br />

P = 0.026<br />

P = 0.020<br />

4<br />

4<br />

4<br />

3<br />

3<br />

3<br />

2<br />

2<br />

2<br />

1<br />

1<br />

1<br />

0<br />

0<br />

0<br />

-1<br />

-1<br />

-1<br />

0 2 4 6 8 10 0 2 4 6 8 10 0 2 4 6 8 10<br />

Zenk-ir nuclei/100 mm 2 Zenk-ir nuclei/100 mm 2 Zenk-ir nuclei/100 mm 2<br />

Figure 5.2. (A–E) Correlations between aggressive behavior and Fos-immunoreactive (-ir) cell<br />

counts in the subpallial (ventral, ventrolateral) and pallial (dorsal) zones of the caudal<br />

lateral septum (LSc.v, LSc.vl, and LSc.d; A–C, respectively), paraventricular hypothalamus<br />

(PVN; D), and anterior hypothalamus (AH; E) of male song sparrows exposed to<br />

STI (n¼16). The intruder’s cage and a speaker broadcasting song were placed adjacent<br />

to the subject’s cage. Subjects showed selective flights to the cage wall adjoining the<br />

intruder, providing a good measure of aggressive response. Data are shown as the natural<br />

log (ln) of the number of contacts with the wire barrier during a 10-min test. (F–I)<br />

Correlations between barrier contacts and Zenk-ir cell counts in the rostral LS (LSr; F),<br />

LS.vl (G), lateral zone of the LSc (LSc.l; H), and PVN (I). Cell counts are shown as the<br />

number of immunoreactive nuclei per 100 mm 2 . Modified from Goodson et al. (2005b).<br />

C


5. Neurogenomic Mechanisms of Aggression in Songbirds 99<br />

In contacts<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

-1<br />

0<br />

5<br />

r 2 = 0.305<br />

p = 0.026<br />

10 15 20 25 30 35<br />

%VT-ir neurons Fos-ir+<br />

Figure 5.3. The percentage of arginine vasotocin (VT) neurons in the PVN that express Fos after a<br />

10-min STI is negatively correlated with aggression (ln, the number of contacts with the<br />

cage wall adjoining the intruder’s cage; see Fig. 5.2 caption; n¼16) in song sparrows.<br />

Modified from Goodson and Kabelik (2009).<br />

(Fig. 5.3; Goodson and Kabelik, 2009). VT and VP are secretagogues for adrenocorticotropic<br />

hormone, and thus the lower IEG activity of the PVN VT cells<br />

in more aggressive males likely reflects a lower stress response to the encounter.<br />

Virtually identical results for VP–Fos colocalization are obtained in lab mice (Ho<br />

et al., 2010). As addressed in Section III.B below, this negative relationship<br />

between aggression and VT–Fos colocalization in the PVN accurately predicts<br />

pharmacological effects that vary in relation to the subject’s dominance status<br />

(Goodson et al., 2009b).<br />

B. Neurochemistry and major modulators<br />

Although territorial aggression in birds has been the focus of hundreds of studies,<br />

including many that address proximate endocrine mechanisms (Goodson et al.,<br />

2005c; Konishi et al.,1989), a surprisingly small number of experiments have been<br />

conducted with the goal of delineating relevant neurochemical circuits in the<br />

brain. An early study of whole-brain neurochemistry shows that dopamine, norepinephrine,<br />

and acetylcholine are all associated with aggressive behavior in male<br />

Japanese quail (Edens, 1987), and catecholaminergic midbrain nuclei show<br />

increased IEG activity in response to STI in song sparrows (Maney and Ball, 2003).<br />

Following aggressive competition for a potential mate, male zebra<br />

finches exhibit significant increases in the percentages of tyrosine hydroxylase-ir<br />

(TH-ir) cells expressing Fos within the “retrorubral” area (A8), substantial nigra<br />

(A9), ventral tegmental area (VTA; A10), and midbrain central gray (A11), but<br />

show a significant decrease in TH–Fos colocalization within the A12 neurons of<br />

the tuberal hypothalamus (Bharati and Goodson, 2006). TH is the rate-limiting


100 Maney and Goodson<br />

enzyme for catecholamine synthesis, and all of the cell groups just listed are<br />

known to be dopaminergic. Despite these results, treatments with quinpirole, a<br />

dopamine D2 receptor agonist, significantly decrease aggression during mate<br />

competition. D1 and D4 agonists are without effect, although a modest inhibition<br />

is observed with the D3 agonist 7-OH-DPAT, which may reflect weak<br />

binding to the D2 receptor (Kabelik et al., 2010). These seemingly contradictory<br />

results likely reflect the fact that courtship is displayed at a high rate during the<br />

competition tests, and given that TH–Fos colocalization in the central gray and<br />

caudal VTA correlates positively with courtship singing (Goodson et al., 2009a),<br />

the increased colocalization of TH and Fos following mate competition is likely<br />

attributable to directed singing and not the display of aggression. The number of<br />

TH-ir cells in the central gray also correlates positively with the average number<br />

of songs that male zebra finches sing to females during courtship tests. Notably,<br />

territorial finch species exhibit fewer TH-ir cells in the caudal VTA than do<br />

gregarious species such as the zebra finch, although this may reflect a lower level<br />

of affiliation rather than a negative relationship between this cell group and<br />

aggression (Goodson et al., 2009a).<br />

Dopaminergic mechanisms of song have also been examined in European<br />

starlings (Sturnus vulgaris), which sing in the con<strong>text</strong> of breeding both to<br />

attract females and to repel other males. Antagonism of D1 receptors decreases<br />

song in breeding-condition males, whereas a dopamine reuptake inhibitor facilitates<br />

it (Schroeder and Riters, 2006), and aggressive song correlates negatively<br />

with D1 receptor density in numerous areas, including the LS, BSTm, medial<br />

preoptic area, and central gray (Heimovics et al., 2009).<br />

The handful of other neurochemical manipulations that have been<br />

conducted in studies of avian aggression have focused on neuromodulators<br />

such as VT, vasoactive intestinal polypeptide (VIP), and endogenous opioids.<br />

Of these, the endogenous opioids have received the least attention, but are<br />

known to inhibit aggression in Japanese quail, at least partially via the delta<br />

receptor subtype (Kotegawa et al., 1997). These neuropeptides are each produced<br />

in multiple brain areas, and as suggested for VT, it may be the case that the<br />

different cell groups have divergent effects on behavior (a possibility that should<br />

also be considered in relation to the major neurotransmitters just discussed;<br />

Goodson and Kabelik, 2009).<br />

Both VIP and VT exert complex effects on aggression that likely reflect<br />

the modulation of stress- and anxiety-related processes. For instance, in territorial<br />

male field sparrows housed in aviaries placed in their natural habitat,<br />

intraseptal infusions of VT decrease aggression in resident–intruder tests, but<br />

selectively facilitate the spontaneous use of an agonistic song type during the<br />

dawn song period (Goodson, 1998a). No effects are observed for the multipurpose<br />

song type that is used to attract females, and VIP tends to exert an opposite<br />

pattern of effects (Goodson, 1998a,b). Interestingly, the VT and VIP systems in


5. Neurogenomic Mechanisms of Aggression in Songbirds 101<br />

the LS are both sensitive to sex steroids. Castration causes down- and upregulation<br />

of VT and VIP immunoreactivity, respectively, and T or E2 replacement<br />

reverses these effects (Aste et al., 1997; Panzica et al., 2001; Voorhuis et al., 1988;<br />

but see Wacker et al., 2008). The VT/VIP systems therefore represent a possible<br />

mechanism whereby gonadal steroids may modulate aggression.<br />

In order to determine whether VT modulates stress-related processes,<br />

and whether it does so in a manner that is integrated with its effects on agonistic<br />

behavior, Goodson and Evans (2004) examined Zenk responses to nonsocial<br />

stress alone (capture in an outdoor flight cage and restraint for intraventricular<br />

infusions), or the same nonsocial stressors followed by STI. These manipulations<br />

were conducted in male song sparrows housed in flight cages placed in their<br />

natural habitat. Subjects were infused with either vehicle or a VT V 1a receptor<br />

antagonist and were sacrificed at the completion of testing for immunolabeling of<br />

Zenk and VT. In some brain areas, nonsocial and social stimuli induced Zenk<br />

within the same subset of cells, which was discernable because (1) in vehicletreated<br />

animals, the nonsocial stressor induced a significant increase in Zenk-ir<br />

cell numbers, and subsequent exposure to the social challenge produced no<br />

further increase, but (2) blocking the Zenk response to handling with the V 1a<br />

antagonist revealed a sensitivity to the social challenge (i.e., by eliminating the<br />

ceiling effect). This “integrated” pattern of Zenk response was observed for<br />

numerous areas, including the AH, POA, lateral VMH, lateral BST, and most<br />

zones of the LS. Notably, in all of these cases, the antagonist exerted more<br />

pronounced effects in the subjects that were exposed to the nonsocial stress<br />

alone. However, in the BSTm and ventrolateral LS, Zenk responses to the social<br />

challenge were significantly greater than to the nonsocial stressor, even in<br />

vehicle-treated subjects, indicating that at least some cells in these areas are<br />

more selectively activated by social challenge. The BSTm showed particularly<br />

selective responses to the social challenge, which were completely blocked by<br />

the V 1a antagonist (Goodson and Evans, 2004).<br />

Unfortunately, VT-ir cells of the BSTm were not detectable in this study<br />

(these neurons are weakly immunoreactive in most vertebrates and may store<br />

little peptide relative to the amount being released), but the VT-immunoreactive<br />

neurons of the PVN showed significant responses to social challenge and, most<br />

interestingly, the VT–Zenk colocalization was reduced by the V 1a antagonist only<br />

in the animals exposed to the STI (Goodson and Evans, 2004). As assessed in a<br />

later study with more robust immunolabeling of VT, VT–Fos colocalization in the<br />

BSTm of male song sparrows is not increased by social challenge, whereas<br />

colocalization in the PVN is negatively correlated with aggression (Fig. 5.2;<br />

Goodson and Kabelik, 2009).<br />

In territorial estrildid finches, exposure to a same-sex conspecific<br />

through a wire barrier actually decreases VT–Fos colocalization in the BSTm,<br />

but the same manipulation increases VT–Fos colocalization in gregarious finch


102 Maney and Goodson<br />

species, and the territorial birds do show large increases in VT–Fos colocalization<br />

if they are reunited with their pair-bond partner. Conversely, socially induced<br />

VT–Fos colocalization in the BSTm is blocked in the gregarious zebra finch if the<br />

subjects are intensely subjugated by a dominant bird (Goodson and Wang,<br />

2006). Thus, the VT neurons of the BSTm exhibit an exquisite sensitivity to<br />

the valence of social stimuli, and more recent findings suggest that this valence<br />

sensitivity is not extended to nonsocial stimuli (Goodson et al., 2009c).<br />

The differential response profiles of the VT neurons in the BSTm and<br />

PVN may account for at least a portion of the con<strong>text</strong>-specificity that is observed<br />

following central infusions of VT or V 1 receptor antagonists. For instance, in<br />

zebra finches, VT promotes aggression in the con<strong>text</strong> of mate competition and a<br />

V 1a antagonist inhibits aggression (Goodson et al., 2004b). These effects are<br />

consistent with the observation that VT–Fos colocalization is increased in the<br />

BSTm during mate competition, but not in the PVN. Conversely, resident–<br />

intruder aggression is inhibited by VT infusions in territorial species, consistent<br />

with the negative correlation between aggression and VT–Fos colocalization in<br />

the PVN (Goodson and Kabelik, 2009; Goodson and Wang, 2006). The different<br />

effects on mate competition and resident–intruder aggression (or nest defense in<br />

zebra finches) can be observed in the same species (Goodson et al., 2009b;<br />

Kabelik et al., 2009), as shown for the territorial violet-eared waxbill (Uraeginthus<br />

granatina) inFig. 5.4A–B. However, in the violet-eared waxbill, males that are<br />

typically dominant do not show a behavioral response to the V 1a antagonist in<br />

standard resident–intruder tests whereas aggression is facilitated in subordinates<br />

(Fig. 5.4C; Goodson et al., 2009b). Thus, perhaps only the subordinate males<br />

activate the PVN VT neurons during aggressive encounters and this activation<br />

inhibits aggression.<br />

Figure 5.4. (A) Peripheral injections of a novel V1a antagonist that crosses the blood–brain barrier<br />

have no effect on resident–intruder aggression in male violet-eared waxbills that are<br />

aggressive and typically dominant, but aggression in the con<strong>text</strong> of mate competition is<br />

significantly reduced by the antagonist in the same males (B). (C) In males that are<br />

typically subordinate, resident–intruder aggression is disinhibited by the same treatments.<br />

Modified from Goodson et al. (2009b).


5. Neurogenomic Mechanisms of Aggression in Songbirds 103<br />

The ability to label IEG products has provided immeasurable insight<br />

into the neural basis of aggression, allowing the identification and mapping of<br />

specific circuits that respond rapidly to social stimuli. Lacking until recently were<br />

powerful genomic methods necessary for a more complete understanding of the<br />

complex protein interactions involved in social responses. The sequencing of the<br />

zebra finch genome (Warren et al., 2010) has provided unprecedented insight<br />

into what happens inside the songbird brain during agonistic encounters. Using<br />

tools such as high throughput sequencing and microarray analysis, investigators<br />

can now look at the regulation of many hundreds of genes simultaneously. In a<br />

recent gene profiling study, Mukai et al. (2009) compared the expression of more<br />

than 11,500 different gene transcripts in free-living song sparrows responding<br />

either to STI or a control intrusion by a heterospecific. For behavioral manipulations<br />

conducted during the breeding season, 67 gene transcripts were differentially<br />

expressed in the hypothalamus following exposure to an STI compared to<br />

control. During the fall, when territorial aggression seems to be independent of<br />

gonadal steroid production (reviewed by Soma, 2006; Soma et al., 2008; see also<br />

Section II), 173 transcripts were affected (Mukai et al., 2009). There were<br />

significant interactions between season and STI for 88 transcripts (Mukai et al.,<br />

2009), which may in part reflect the differential regulation of the pituitary–<br />

gonadal axis across seasons. The expression of many of the gene transcripts was<br />

not, however, affected by season and therefore may be important for the regulation<br />

of aggressive behavior itself rather than endocrine responses to aggressive<br />

encounters. This study represents the early days of genomic analysis of social<br />

behavior in free-living, natural populations and sets the standard for many more<br />

sure to follow. In the next section, we consider a songbird species that because of<br />

a natural genetic anomaly is becoming a popular model for studying the genetic<br />

mechanisms underlying aggression.<br />

IV. A NATURAL MODEL UNITING SOCIAL BEHAVIOR, HORMONES,<br />

AND GENETICS<br />

A. The white-throated sparrow<br />

The underlying genetic basis of variation in social behavior is of intense interest,<br />

yet only a handful of genes have been linked to specific social behaviors in<br />

vertebrates (reviewed by Robinson et al., 2005). Thus, there is an obvious need to<br />

identify populations, human or otherwise, in which there is clear linkage between<br />

genes and social behavior. A common wild songbird, the white-throated<br />

sparrow, offers such an opportunity. This species, in which socially monogamous<br />

pairs defend breeding territories, provision the young with food, and form flocks<br />

with stable dominance hierarchies in the winter, is a typical New World sparrow


104 Maney and Goodson<br />

in nearly all respects. What sets it apart from other songbirds is that it exhibits<br />

alternative phenotypes, defined by a plumage polymorphism, that differ in their<br />

social behavior. Both males and females can be categorized into one of two<br />

plumage morphs that differ primarily in the color of the crown stripes (Lowther,<br />

1961; Piper and Wiley, 1989; Watt, 1986; see Fig. 5.5). Behavioral studies<br />

conducted in the animals’ natural habitat have established that individuals<br />

with a white medial stripe (WS) on the crown engage in a more aggressive<br />

strategy, whereas birds with a tan medial stripe (TS) are more parental.<br />

The species represents a promising model in which to study the genetic<br />

basis of aggression because the plumage pattern segregates with the presence or<br />

absence of a structural rearrangement of chromosome 2. WS individuals are<br />

heterozygous for the rearranged chromosome (ZAL2 m ), whereas those of the TS<br />

morph are homozygous for the wild-type chromosome (ZAL2; Thorneycroft,<br />

1975). Once they molt into adult plumage the phenotype is fixed for the lifetime<br />

of the individual. Within a population, approximately half of the birds are WS<br />

(ZAL2/2 m ), whereas the other half are TS (ZAL2/2; Lowther, 1961;<br />

Thorneycroft, 1975). This balanced polymorphism is maintained in the population<br />

by disassortative mating—WS and TS birds nearly always mate with individuals<br />

of the opposite morph (Knapton and Falls, 1983; Lowther, 1961;<br />

Thorneycroft, 1975; Tuttle, 1993). This mating pattern results in a virtual<br />

absence of birds homozygous for ZAL2 m , a genotype that Thorneycroft (1975)<br />

hypothesized may be less viable due to recessive deleterious mutations. Of more<br />

than 1000 individuals genotyped, only one was found to be homozygous for<br />

ZAL2 m (Maney et al. unpublished data; Romanov et al., 2009; Thorneycroft,<br />

1975).<br />

Figure 5.5. Plumage polymorphism in white-throated sparrows. (A) Individuals of the white-stripe<br />

(WS) morph have alternating black and white stripes on the crown, bright yellow lores,<br />

and a clear white throat patch. (B) Individuals of the tan-stripe (TS) morph have<br />

alternating brown and tan stripes on the crown, duller yellow lores, and dark bars within<br />

the white throat patch. Photos by Allison Reid. Reprinted from Maney (2008).


5. Neurogenomic Mechanisms of Aggression in Songbirds 105<br />

The behavioral differences that segregate with the ZAL2 m chromosome<br />

have been well documented in field studies. Males and females of the WS morph<br />

are more aggressive, both in territorial defense and in mate-seeking, than their<br />

TS counterparts. WS males sing more in response to STI than TS males (Collins<br />

and Houtman, 1999; Kopachena and Falls, 1993a; Horton and Maney, unpublished<br />

observations) and are more likely to trespass onto the territories of other<br />

males (Tuttle, 2003). Whereas WS females sing and engage in active territorial<br />

defense independently of their mates, TS females do so only rarely (Kopachena<br />

and Falls, 1993a; Horton and Maney, unpublished observations). TS birds of<br />

both sexes feed young more often during the parental phase of the breeding<br />

season than do WS birds (Knapton and Falls, 1983; Kopachena and Falls,<br />

1993b). The relative strategies employed by the different morphs of this species<br />

therefore fall onto different ends of the behavioral continuum between territoriality<br />

and parenting (Trivers, 1972).<br />

B. Endocrine and neuroendocrine correlates of<br />

behavioral polymorphism<br />

Because the behavioral trade-off between territorial defense and parenting is<br />

clearly mediated at least in part by HPG activity in songbirds (Ketterson and<br />

Nolan, 1994; McGlothlin et al., 2007; Wingfield et al., 1990), we should immediately<br />

suspect that, in white-throated sparrows, HPG function may vary according<br />

to morph. Spinney et al. (2006) found that in free-living birds in breeding<br />

condition, WS males do have larger testes and higher levels of circulating T than<br />

TS males. This phenomenon has also been demonstrated in captive populations<br />

(Maney, 2008; Swett and Breuner, 2009). In both the field and the lab, however,<br />

the difference in plasma T disappears when birds are not in breeding condition<br />

(Maney, 2008; Spinney et al., 2006). Interestingly, morph differences in aggression<br />

appear only during the breeding season, mirroring the morph difference in<br />

circulating T. In winter flocks and in laboratory-housed birds held on short days,<br />

morph is not related to dominance rank or to aggression (Dearborn and Wiley,<br />

1993; Harrington, 1973; Piper and Wiley, 1989; Schlinger, 1987; Schwabl et al.,<br />

1988; Watt et al., 1984; Wiley et al., 1999). In contrast, when birds are held on<br />

long days and undergo gonadal recrudescence, WS birds engage in significantly<br />

more aggression than their TS cage-mates and tend to outrank them (Fig. 5.6; see<br />

also Watt et al., 1984). Morph differences in dominance and aggression may<br />

therefore depend on season and thus perhaps on differences in HPG function.<br />

Because levels of gonadal steroids differ between the morphs, the<br />

behavioral polymorphism may be driven by the effects of these steroids on the<br />

brain. To evaluate this hypothesis, Maney et al. (2005) compared the morphs<br />

with respect to the VT and VIP neuropeptide systems, which are highly steroid<br />

dependent (Aste et al., 1997; Panzica et al., 2001; Voorhuis et al., 1988). WS birds


106 Maney and Goodson<br />

Figure 5.6. Medians, IQR, and ranges for (A) aggression scores (number of aggressive acts initiated<br />

per hour) and (B) individual ranks (as percent opponents dominated) within social<br />

groups. Males were introduced in single-sex groups of six birds (three WS and three TS<br />

per group) in indoor aviaries. Aggression scores and ranks were determined 10–14 days<br />

later by observing interactions and constructing dominance matrices. During spring-like<br />

day lengths (16L:8D), WS males were (A) more aggressive and (B) outranked TS males.<br />

Rank was unrelated to morph on short days (8L:16D). The long- and short-day experiments<br />

were conducted on different individuals. Data from Horton and Maney,<br />

unpublished.<br />

had higher levels of VT-immunoreactivity in the BSTm and ventrolateral LS<br />

than TS birds. Since T is higher in WS males, this result is consistent with the<br />

idea that T may be engaging the VT system in the LS. Central administration of<br />

VT in the closely related white-throated sparrow induces agonistic song (Maney<br />

et al., 1997), suggesting that engagement of this system may be directly related to<br />

aggressive behaviors. Central administration of VIP, in contrast, reduces agonistic<br />

song in field sparrows (Goodson, 1998a); immunoreactivity for this peptide<br />

was higher in the ventrolateral LS of the TS (less aggressive) morph. VIP<br />

immunoreactivity in this region is inversely proportional to T levels (Aste<br />

et al., 1997) again supporting a possible role for gonadal sex steroids in the<br />

control of aggression in this species.


5. Neurogenomic Mechanisms of Aggression in Songbirds 107<br />

C. Causality and “phenotypic engineering”<br />

Looking for morph differences in endocrine variables in unmanipulated individuals<br />

is an important endeavor, in that significant correlations can help illuminate<br />

possible physiological causes of aggression. Such correlations alone, however, can<br />

provide only limited information on causal mechanisms. The morph difference in<br />

plasma T, for example, could be a consequence, rather than a cause, of polymorphic<br />

behavior. Either scenario would explain the observed correlations between T and<br />

social behavior (Spinney et al., 2006). As discussed in Section II above, experiments<br />

involving manipulation of T or of social con<strong>text</strong>s in songbirds have revealed causal<br />

effects in both directions. Recall that in other songbird species, free-living males<br />

treated with T defend larger territories, engage in more aggression, acquire more<br />

mates, and provide less parental care than untreated males (Hegner and Wingfield,<br />

1987; Schoech et al., 1998; Silverin, 1980; Wingfield, 1984b,c). Territorial intrusion<br />

or the presence of receptive females, however, cause release of endogenous T<br />

(Dufty and Wingfield, 1986; Moore, 1983; Wingfield and Hahn, 1994; Wingfield<br />

and Monk, 1994). A one-way causal effect of T on aggression and parenting may<br />

not completely explain polymorphic behavior in white-throated sparrows.<br />

Some authors have suggested that the role of hormones in alternative<br />

phenotypes is best studied by performing hormonal manipulations, or “phenotypic<br />

engineering” (Ketterson and Nolan, 1992; Miles et al., 2007; Zera and Harshman,<br />

2001). To test whether morph-dependent variation in territorial behavior in male<br />

white-throated sparrows is attributable entirely to variation in T, Maney et al.<br />

(2009) eliminated morph differences in T and then compared WS and TS<br />

responses to STI in the lab. Males in nonbreeding condition received silastic<br />

implants containing T, so that plasma levels in the WS and TS groups were<br />

high and equal. When presented with audio playback of conspecific male song,<br />

WS males sang significantly more often than TS males. This result suggests that<br />

WS males respond more aggressively to a territorial challenge than TS males, even<br />

when T levels are experimentally equalized between the morphs.<br />

If morph differences in social behavior are not caused simply by differences<br />

in plasma T, then our search for causal factors should turn to other aspects of HPG<br />

function, for example, steroid binding or metabolism. The list of such factors is long<br />

and includes a large number of receptors, enzymes, and binding globulins. Comparative<br />

genomic approaches are required to conduct large-scale comparisons of gene<br />

expression as well as detailed analysis of the genetic differences between the morphs.<br />

D. Mapping the ZAL2 m<br />

The early genetic work in the white-throated sparrow, done more than 35 years<br />

ago (Thorneycroft, 1975), showed definitively that morph differences are associated<br />

with a clear, tractable chromosomal rearrangement. The ZAL2 m


108 Maney and Goodson<br />

chromosome thus offers a powerful starting point for understanding the mechanisms<br />

underlying aggressive behavior in birds and other vertebrates. In essence,<br />

nature has created a genetic manipulation that allows us to identify genes that<br />

are affected by the rearrangement and therefore potentially causal for heightened<br />

aggression in the WS individuals.<br />

The identification of such genes first requires mapping of the ZAL2 m<br />

rearrangement; genes that map within it can then be evaluated as likely candidates.<br />

Using a comparative genomic approach, Thomas et al. (2008) (see also<br />

Davis et al., 2011; Huynh et al., 2010a,b) began the initial modern genomic<br />

characterization of the ZAL2 m rearrangement. By taking advantage of the<br />

genomic resources available for two other avian species, the chicken and the<br />

zebra finch, they established a comparative map of ZAL2 m and found that the<br />

chromosome contains a complex rearrangement involving not one but at least<br />

two inversions around the centromere (Fig. 5.7). The two inversions may have<br />

occurred in succession, with the second completely contained within the first.<br />

Alternatively, ZAL2 and ZAL2 m may each represent rearranged versions of an<br />

ancestral chromosome 2, having undergone rearrangement at different times.<br />

The rearrangement now spans the majority of the chromosome and could<br />

contain as many as 1000 protein-coding genes (Davis et al., 2011; Thomas<br />

et al., 2008). Thus, although the region containing the rearrangement is large,<br />

this work has delineated a finite set of genes linked to the behavioral and<br />

plumage polymorphisms in this species.<br />

2<br />

Presumed<br />

ancestral/intermediate<br />

2 m<br />

Figure 5.7. Model for the ZAL2 m rearrangement. A minimum of two pericentric inversions,<br />

represented by the pairs of dashed lines, are hypothesized to have led to the ZAL2/2 m<br />

polymorphism. ZAL2 (top) and ZAL2 m (bottom) are shown along with a hypothetical<br />

chromosomal arrangement (middle) that could be either ancestral to both the ZAL2<br />

and ZAL2 m or an intermediate arrangement. Centromeres are represented by filled<br />

circles. Dark and light boxes represent segments originating on the short and long arms<br />

of the presumed ancestral chromosome, respectively. Free recombination between the<br />

ZAL2 and ZAL2 m is limited to the tip of the short arm (hatched boxes).


5. Neurogenomic Mechanisms of Aggression in Songbirds 109<br />

Exactly how does the architecture of ZAL2 m affect the expression of the<br />

genes inside the rearrangement and the proteins they encode Inversions are<br />

hypothesized to affect gene and protein function in two main ways. First, genes at<br />

or near the breakpoints may be physically disrupted or otherwise directly affected<br />

by the breakage and subsequent change in position. So far, sequencing efforts<br />

have identified no genes physically disrupted by the ZAL2 m breakpoints (Davis<br />

et al., 2011); however, position effects may have led to functionally distinct<br />

alleles for those nearby. For example, a cluster of genes encoding bitter taste<br />

receptors has been separated by one of the breakpoints and now maps to different<br />

arms of the chromosome (Davis et al., 2010). This separation, which appears to<br />

have led to nonsynonymous variants detected between the ZAL2 and ZAL2 m ,<br />

may have implications for diet and habitat selection but is unlikely to explain<br />

morph differences in aggression and parenting.<br />

The behavioral polymorphisms in social behavior in this species are more<br />

likely related to a second important consequence of pericentric inversions, which<br />

is the suppression of recombination and subsequent genetic differentiation of the<br />

inverted region. Thorneycroft (1975) observed that pairing in the ZAL2/2 m<br />

bivalent during meiosis was limited to one arm of each chromosome. Cytogenetic<br />

mapping efforts (Davis et al., 2011; Thomas et al., 2008) suggest that single<br />

recombination events elsewhere in the chromosome would give rise to gametes<br />

with large duplications and deletions, thereby effectively preventing the inheritance<br />

of the recombined chromosomes. ZAL2 m may therefore be largely isolated<br />

from ZAL2. Population genetics studies have confirmed differentiation between<br />

the ZAL2 and ZAL2 m over the entire rearranged region as a result of suppression of<br />

recombination (Huynh et al., 2010a; Thomas et al., 2008). Alleles are shared<br />

between the haplotypes only at the tip of the short arm of ZAL2/2 m ,whichis<br />

outside the rearrangement (Fig. 5.7). The rearrangement itself contains a unique<br />

set of alleles that are not shared with ZAL2—an estimated 3000 fixed differences<br />

(Davis et al., 2011), and this set is inherited together. Thus, the lack of gene flow<br />

between the ZAL2 and ZAL2 m has provided opportunity for the evolution of<br />

functionally distinct alleles that are restricted to one arrangement or the other. In<br />

the continuing analysis of the rearrangement, we should expect to find a series of<br />

genes, inherited together as a unit in WS birds, that are functionally distinct from<br />

the ZAL2 alleles with regard to either their protein products or patterns of<br />

expression. Given the important role of the HPG axis in aggression in this and<br />

other species, the strongest candidates will be closely related to HPG function.<br />

V. FUTURE DIRECTIONS<br />

Genetic research with comparative models will ultimately show how key genes,<br />

the molecular functions of which are conserved across evolutionary divergence,<br />

relate to complex and highly derived social behaviors such as aggression.


110 Maney and Goodson<br />

The mechanisms that underlie social behaviors in less accessible species, such as<br />

humans, are best studied in species that live in societies, particularly those that<br />

can be studied in their natural habitats or under naturalistic conditions (Insel<br />

and Fernald, 2004). The existing database on avian social behavior is unparalleled—for<br />

example, for over 4000 species, we know whether they are territorial<br />

or colonial, socially monogamous or polygynous, migratory, or sedentary. We<br />

have high-quality recordings of their vocalizations. No other group of animals,<br />

invertebrate or vertebrate, has been studied with such passion and intensity.<br />

This collective database, although it could provide profound insight into the<br />

neuroendocrine basis of diverse social behaviors, is underutilized by neuroscientists<br />

because the availability of genomic tools has, until recently, been limited.<br />

The recent sequencing of the zebra finch genome (Warren et al., 2010) now<br />

makes possible unprecedented advances in our understanding of social behavior<br />

because the resulting tools are applicable to all songbirds.<br />

Advances in genomic technology, together with conservation of underlying<br />

mechanisms, will make it more and more feasible to bridge from wellcharacterized<br />

data-rich lab organisms, such as mice, to phenomena-rich wild<br />

species. These species, which include fish, lizards, songbirds, and voles, are<br />

proving to be rich resources for the analysis of social behavior and for the<br />

development of general principles (Robinson et al., 2005). Studies with these<br />

model organisms have demonstrated the power of a comparative approach—<br />

looking for neural or genetic differences among individuals with known behavioral<br />

differences (Bullock, 1984; Robinson, et al., 2005). The songbird model is<br />

preferable to more typical laboratory species in the study of social behavior<br />

because of the greater parallels with humans regarding societal structures and<br />

hormonal bases of behavioral strategies, as well as the potential to study freeliving<br />

populations under natural conditions. Our work and that of others is<br />

making these natural and powerful models of vertebrate behavior feasible for<br />

genomic and neuroendocrine analysis.<br />

Acknowledgments<br />

The authors thank Jim Thomas for his contributions to the work described in Section IV. The<br />

research from the authors’ labs was supported by NIH MH062656 to J. L. G., by NIH<br />

1R01MH082833-01, NIH 5R21MH082046-02, and NSF IOS-0723805 to D. L. M. and the Center<br />

for Behavioral Neuroscience.<br />

References<br />

Adkins-Regan, E. (2005). Hormones and Animal Social Behavior. Princeton <strong>University</strong> Press,<br />

Princeton.<br />

Allee, W. C. (1936). Analytical studies of group behavior in birds. Wilson Bull. 48, 145–151.<br />

Allee, W. C. (1942). Group organization among vertebrates. Science 95, 289–293.


5. Neurogenomic Mechanisms of Aggression in Songbirds 111<br />

Allee, W. C., Collias, N. E., and Lutherman, C. Z. (1939). Modification of the social order in flocks of<br />

hens by the injection of testosterone propionate. Physiol. Zool. 12, 412–440.<br />

Arcese, P., Sogge, M. K., Marr, A. B., and Patten, M. A. (2002). Song Sparrow (Melospiza melodia).<br />

In “The Birds of North America Online” (A. Poole, ed.). Cornell Lab of Ornithology, Ithaca. doi:<br />

10.2173/bna.704.<br />

Archawaranon, M., and Wiley, R. H. (1988). Control of aggression and dominance in whitethroated<br />

sparrows by testosterone and its metabolites. Horm. Behav. 22, 497–517.<br />

Archawaranon, M., Dove, L., and Wiley, R. H. (1991). Social inertia and hormonal control of<br />

aggression and dominance in white-throated sparrows. Behaviour 118, 42–65.<br />

Archer, J. (2006). Testosterone and human aggression: An evaluation of the challenge hypothesis.<br />

Neurosci. Biobehav. Rev. 30, 319–345.<br />

Aromaki, A. S., Lindman, R. E., and Eriksson, C. J. P. (1999). Testosterone, aggressiveness, and<br />

antisocial personality. Aggress. Behav. 25, 113–123.<br />

Aste, N., Viglietti-Panzica, C., Balthazart, J., and Panzica, G. C. (1997). Testosterone modulation of<br />

peptidergic pathways in the septo-preoptic region of male Japanese quail. Poult. Avian Biol. Rev. 8,<br />

77–93.<br />

Ball, G. F., and Balthazart, J. (2001). Ethological concepts revisited: Immediate early gene induction<br />

in response to sexual stimuli in birds. Brain Behav. Evol. 57, 252–270.<br />

Baptista, L. F., DeWolfe, B. B., and Avery-Beausoleil, L. (1987). Testosterone, aggression, and<br />

dominance in Gambel’s white-crowned sparrows. Wilson Bull. 99, 86–91.<br />

Baptista, L. F., Trail, P. W., DeWolfe, B. B., and Morton, M. L. (1993). Singing and its functions in<br />

female white-crowned sparrows. Anim. Behav. 46, 511–524.<br />

Berg, S., and Wynne-Edwards, K. E. (2001). Changes in testosterone, cortisol, and estradiol levels in<br />

men becoming fathers. Mayo Clin. Proc. 76, 582–592.<br />

Bernhardt, P. C., Dabbs, J. M., Fielden, J. A., and Lutter, C. D. (1998). Testosterone changes during<br />

vicarious experiences of winning and losing among fans at sporting events. Physiol. Behav. 65,<br />

59–62.<br />

Bharati, I. S., and Goodson, J. L. (2006). Fos responses of dopamine neurons to sociosexual stimuli in<br />

male zebra finches. Neuroscience 143, 661–670.<br />

Blanchard, D. C., and Blanchard, R. J. (2003). What can animal aggression research tell us about<br />

human aggression Horm. Behav. 44, 171–177.<br />

Blanchard, R. J., and Blanchard, D. C. (2005). Some suggestions for revitalizing aggression research.<br />

Novartis Found. Symp. 268, 4–12.<br />

Book, A. S., Starzyk, K. B., and Quinsey, V. L. (2001). The relationship between testosterone and<br />

aggression: A meta-analysis. Aggr. Viol. Behav. 6, 579–599.<br />

Booth, A., Shelley, G., Mazur, A., Tharp, G., and Kittok, R. (1989). Testosterone, and winning and<br />

losing in human competition. Horm. Behav. 23, 556–571.<br />

Buchanan, K. L., Evans, M. R., Roberts, M. L., Rowe, L., and Goldsmith, A. R. (2010). Does<br />

testosterone determine dominance in the house sparrow Passer domesticus An experimental<br />

test. J. Avian Biol. 41, 445–451.<br />

Bullock, T. H. (1984). Comparative neuroscience holds promise for quiet revolutions. Science 225,<br />

473–478.<br />

Carey, M., Burhans, D. E., and Nelson, D. A. (2008). Field Sparrow (Spizella pusilla). In “The Birds of<br />

North America Online” (A. Poole, ed.). Cornell Lab of Ornithology, Ithaca. doi: 10.2173/<br />

bna.103.<br />

Chase, I. D. (1982). Dynamics of hierarchy formation: The sequential development of dominance<br />

relationships. Behaviour 80, 218–240.<br />

Chilton, G., Baker, M. C., Barrentine, C. D., and Cunningham, M. A. (1995). White-Crowned-<br />

Sparrow (Zonotrichia leucophrys). In “The Birds of North America Online” (A. Poole, ed.). Cornell<br />

Lab of Ornithology, Ithaca. doi: 10.2173/bna.183.


112 Maney and Goodson<br />

Collins, C. E., and Houtman, A. M. (1999). Tan and white color morphs of white-throated sparrows<br />

differ in their non-song vocal responses to territorial intrusion. Condor 101, 842–845.<br />

Crews, D., Lou, W., Fleming, A., and Ogawa, S. (2006). From gene networks underlying sex<br />

determination and gonadal differentiation to the development of neural networks regulating<br />

sociosexual behavior. Brain Res. 1126, 109–121.<br />

Crook, J. H., and Butterfield, P. A. (1968). Effects of testosterone propionate and luteinizing<br />

hormone on agonistic and nest building behavior of Quelea quelea. Anim. Behav. 16, 370–384.<br />

Dabbs, J. M., Jr., and Morris, R. (1990). Testosterone, social class, and antisocial behavior in a sample<br />

of 4,462 men. Psychol. Sci. 1, 209–211.<br />

Dabbs, J. M., Jr., Strong, R., and Milun, R. (1997). Exploring the mind of testosterone: A bleeper<br />

study. J. Res. Pers. 31, 577–587.<br />

Daitzman, R., and Zuckerman, M. (1980). Disinhibitory sensation seeking, personality and gonadal<br />

hormones. Pers. Individ. Diff. 1, 103–110.<br />

Davis, J. K., Lowman, J. J., Thomas, P. J., ten Hallers, B. F. H., Koriabine, M., Huynh, L. Y.,<br />

Maney, D. L., de Jong, P. J., Martin, C. L., and NISC Comparative Sequencing Program<br />

Thomas, J. W. (2010). Evolution of a bitter taste receptor gene cluster in a New World sparrow.<br />

Genome Biol. Evol. 2, 358–370.<br />

Davis, J. K., Mittel, L. X., Lowman, J. J., Thomas, P. J., Maney, D. L., Martin, C. L., and<br />

Thomas, J. W. NISC Comparative Sequencing Program (2011). Haplotype-based genetic<br />

sequencing of a chromosomal polymorphism in the white-throated sparrow (Zonotrichia albicollis).<br />

J. Heredity (accepted with minor revision).<br />

Dawson, A., Kin, V. M., Bentley, G. E., and Ball, G. F. (2001). Photoperiodic control of seasonality<br />

in birds. J. Biol. Rhythms 16, 365–380.<br />

Dearborn, D. C., and Wiley, R. H. (1993). Prior residence has a gradual influence on the dominance<br />

in captive white-throated sparrows. Anim. Behav. 46, 39–46.<br />

Dufty, A. M., and Wingfield, J. C. (1986). The influence of social cues on the reproductive<br />

endocrinology of male brown-headed cowbirds: Field and laboratory studies. Horm. Behav. 20,<br />

222–234.<br />

Edens, F. W. (1987). Agonistic behavior and neurochemistry in grouped Japanese quail. Comp.<br />

Biochem. Physiol. A Comp. Physiol. 86, 473–479.<br />

Elekonich, M. M. (2000). Female song sparrow, Melospiza melodia, response to simulated conspecific<br />

and heterospecific intrusion across three seasons. Anim. Behav. 59, 551–557.<br />

Falls, J. B. (1969). Functions of territorial song in the white-throated sparrow. In “Bird Vocalizations”<br />

(R. A. Hinde, ed.), pp. 207–232. Cambridge <strong>University</strong> Press, Cambridge.<br />

Falls, J. B. (1988). Does song deter territorial intrusion in white-throated sparrows (Zonotrichia<br />

albicollis) Can. J. Zool. 66, 206–211.<br />

Falls, J. B., and Kopachena, J. G. (2010). In “White-Throated Sparrow (Zonotrichia albicollis), The<br />

Birds of North America Online” (A. Poole, ed.). Cornell Lab of Ornithology, Ithaca. doi:<br />

10.2173/bna.128.<br />

Ficken, R. W., Ficken, M. S., and Hailman, J. P. (1978). Differential aggression in genetically<br />

different morphs of the white-throated sparrow (Zonotrichia albicollis). Z. Tierpsychol. 46, 43–57.<br />

Finch, C. E., and Rose, M. R. (1995). Hormones and the physiological architecture of life history<br />

evolution. Quart. Rev. Biol. 70, 1–52.<br />

Fleming, A. S., Corter, C., Stallings, J., and Steiner, M. (2002). Testosterone and prolactin are<br />

associated with emotional responses to infant cries in new fathers. Horm. Behav. 42, 399–413.<br />

Gammie, S. C. (2005). Current models and future directions for understanding the neural circuitries<br />

of maternal behaviors in rodents. Behav. Cogn. Neurosci. Rev. 4, 119–135.<br />

Goldman, P. (1973). Song recognition by field sparrows. Auk 90, 103–113.<br />

Goodson, J. L. (1998a). Territorial aggression and dawn song are modulated by septal vasotocin and<br />

vasoactive intestinal polypeptide in male field sparrows (Spizella pusilla). Horm. Behav. 34, 67–77.


5. Neurogenomic Mechanisms of Aggression in Songbirds 113<br />

Goodson, J. L. (1998b). Vasotocin and vasoactive intestinal polypeptide modulate aggression in a<br />

territorial songbird, the violet-eared waxbill (Estrildidae: Uraeginthus granatina). Gen. Comp.<br />

Endocrinol. 111, 233–244.<br />

Goodson, J. L. (2005). The vertebrate social behavior network: Evolutionary themes and variations.<br />

Horm. Behav. 48, 11–22.<br />

Goodson, J. L., and Bass, A. H. (2002). Vocal-acoustic circuitry and descending vocal pathways in<br />

teleost fish: Convergence with terrestrial vertebrates reveals conserved traits. J. Comp. Neurol.<br />

448, 298–322.<br />

Goodson, J. L., and Evans, A. K. (2004). Neural responses to territorial challenge and nonsocial stress<br />

in male song sparrows: Segregation, integration, and modulation by a vasopressin V1 antagonist.<br />

Horm. Behav. 46, 371–381.<br />

Goodson, J. L., and Kabelik, D. (2009). Dynamic limbic networks and social diversity in vertebrates:<br />

From neural con<strong>text</strong> to neuromodulatory patterning. Front. Neuroendocrinol. 30, 429–441.<br />

Goodson, J. L., and Wang, Y. (2006). Valence-sensitive neurons exhibit divergent functional profiles<br />

in gregarious and asocial species. Proc. Natl. Acad. Sci. USA 103, 17013–17017.<br />

Goodson, J. L., Eibach, R., Sakata, J., and Adkins-Regan, E. (1999). Effect of septal lesions on male<br />

song and aggression in the colonial zebra finch (Taeniopygia guttata) and the territorial field<br />

sparrow (Spizella pusilla). Behav. Brain Res. 98, 167–180.<br />

Goodson, J. L., Evans, A. K., and Lindberg, L. (2004a). Chemoarchitectonic subdivisions of the<br />

songbird septum and a comparative overview of septum chemical anatomy in jawed vertebrates.<br />

J. Comp. Neurol. 473, 293–314.<br />

Goodson, J. L., Lindberg, L., and Johnson, P. (2004b). Effects of central vasotocin and mesotocin<br />

manipulations on social behavior in male and female zebra finches. Horm. Behav. 45, 136–143.<br />

Goodson, J. L., Saldanha, C. J., Hahn, T. P., and Soma, K. K. (2005a). Recent advances in behavioral<br />

neuroendocrinology: Insights from studies on birds. Horm. Behav. 48, 461–473.<br />

Goodson, J. L., Evans, A. K., Lindberg, L., and Allen, C. D. (2005b). Neuro-evolutionary patterning<br />

of sociality. Proc. R. Soc. Lond. B 272, 227–235.<br />

Goodson, J. L., Evans, A. K., and Soma, K. K. (2005c). Neural responses to aggressive challenge<br />

correlate with behavior in nonbreeding sparrows. Neuroreport 16, 1719–1723.<br />

Goodson, J. L., Kabelik, D., Kelly, A. M., Rinaldi, J., and Klatt, J. D. (2009a). Midbrain dopamine<br />

neurons reflect affiliation phenotypes in finches and are tightly coupled to courtship. Proc. Natl.<br />

Acad. Sci. USA 106, 8737–8742.<br />

Goodson, J. L., Kabelik, D., and Schrock, S. E. (2009b). Dynamic neuromodulation of aggression by<br />

vasotocin: Influence of social con<strong>text</strong> and social phenotype in territorial songbirds. Biol. Lett. 5,<br />

554–556.<br />

Goodson, J. L., Rinaldi, J., and Kelly, A. M. (2009c). Vasotocin neurons in the bed nucleus of the<br />

stria terminalis preferentially process social information and exhibit properties that dichotomize<br />

courting and non-courting phenotypes. Horm. Behav. 55, 197–202.<br />

Gray, P. B. (2003). Marriage, parenting, and testosterone variation among Kenyan Swahili men. Am.<br />

J. Phys. Anthropol. 122, 279–286.<br />

Gray, P. B., Kahlenberg, S. M., Barret, E. S., Lipson, S. F., and Ellison, P. T. (2002). Marriage and<br />

fatherhood are associated with low testosterone in males. Evol. Hum. Behav. 23, 193–201.<br />

Guhl, A. M. (1968). Social inertia and social stability in chickens. Anim. Behav. 16, 219–232.<br />

Hamilton, W. D. (1971). Geometry for the selfish herd. J. Theoret. Biol. 31, 295–311.<br />

Harrington, B. A. (1973). Aggression in winter resident and spring migrant white-throated sparrows<br />

in Massachusetts. Bird Banding 44, 314–315.<br />

Hau, M. (2007). Regulation of male traits by testosterone: Implications for the evolution of<br />

vertebrate life histories. Bioessays 292, 133–144.<br />

Hegner, R. E., and Wingfield, J. C. (1986). Behavioral and endocrine correlates of multiple brooding<br />

in the semi-colonial house sparrow Passer domesticus I. Males. Horm. Behav. 20, 294–312.


114 Maney and Goodson<br />

Hegner, R. E., and Wingfield, J. C. (1987). Effects of experimental manipulation of testosterone<br />

levels on parental investment and breeding success in male house sparrows. Auk 104, 462–469.<br />

Heimovics, S. A., Cornil, C. A., Ball, G. F., and Riters, L. V. (2009). D1-like dopamine receptor<br />

density in nuclei involved in social behavior correlates with song in a con<strong>text</strong>-dependent fashion<br />

in male European starlings. Neuroscience 159, 962–973.<br />

Herdegen, T., and Leah, J. D. (1998). Inducible and constitutive transcription factors in the<br />

mammalian nervous system: Control of gene expression by Jun, Fos and Krox, and CREB/ATF<br />

proteins. Brain Res. Rev. 28, 370–490.<br />

Ho, J. M., Murray, J. H., Demas, G. E., and Goodson, J. L. (2010). Vasopressin cell groups exhibit<br />

strongly divergent responses to copulation and male-male interactions in mice. Horm. Behav. 58,<br />

368–377.<br />

Hoke, K. L., Ryan, M. J., and Wilczynski, W. (2005). Social cues shift functional connectivity in the<br />

hypothalamus. Proc. Natl. Acad. Sci. USA 102, 10712–10717.<br />

Huynh, L. Y., Maney, D. L., and Thomas, J. W. (2010a). Chromosome-wide linkage disequilibrium<br />

caused by an inversion polymorphism in the white-throated sparrow. Heredity (in press).<br />

Huynh, L. Y., Maney, D. L., and Thomas, J. W. (2010b). Contrasting population genetic patterns<br />

within the white-throated sparrow genome (Zonotrichia albicollis). BMC Genomics 11, 96.<br />

Insel, T. R., and Fernald, R. D. (2004). How the brain processes social information: Searching for the<br />

social brain. Annu. Rev. Neurosci. 27, 697–722.<br />

Julian, T., and McHenry, P. C. (1989). Relationship of testosterone to men’s family functioning at<br />

mid-life: A research note. Aggress. Behav. 15, 281–289.<br />

Kabelik, D., Klatt, J. D., Kingsbury, M. A., and Goodson, J. L. (2009). Endogenous vasotocin exerts<br />

con<strong>text</strong>-dependent behavioral effects in a semi-naturalistic colony environment. Horm. Behav.<br />

56, 101–107.<br />

Kabelik, D., Kelly, A. M., and Goodson, J. L. (2010). Dopaminergic regulation of mate competition<br />

aggression and aromatase-Fos colocalization in vasotocin neurons. Neuropharmacology 58,<br />

117–125.<br />

Ketterson, E. D., and Nolan, V. (1992). Hormones and life histories: An integrative approach. Am.<br />

Nat. 140, S33–S62.<br />

Ketterson, E. D., and Nolan, V. (1994). Male parental behavior in birds. Annu. Rev. Ecol. Syst. 25,<br />

601–628.<br />

Keys, G. C., and Rothstein, S. I. (1991). Benefits and costs of dominance and subordinance in whitecrowned<br />

sparrows and the paradox of status signalling. Anim. Behav. 42, 899–912.<br />

Kingsbury, M. A., Kelly, A. M., Schrock, S. E., and Goodson, J. L. (2011). Mammal-like organization<br />

of the avian midbrain central gray and a reappraisal of the intercollicular nucleus. PLoS One.<br />

Knapton, R. W., and Falls, J. B. (1983). Differences in parental contribution among pair types in the<br />

polymorphic white-throated sparrow. Can. J. Zool. 61, 1288–1292.<br />

Kollack-Walker, S., Watson, S. J., and Akil, H. (1997). Social stress in hamsters: Defeat activates<br />

specific neurocircuits within the brain. J. Neurosci. 17, 8842–8855.<br />

Konishi, M., Emlen, S. T., Ricklefs, R. E., and Wingfield, J. C. (1989). Contributions of bird studies<br />

to biology. Science 246, 465–472.<br />

Kopachena, J. G., and Falls, J. B. (1993a). Aggressive performance as a behavioral correlate of<br />

plumage polymorphism in the white-throated sparrow (Zonotrichia albicollis). Behaviour 124,<br />

249–266.<br />

Kopachena, J. G., and Falls, J. B. (1993b). Re-evaluation of morph-specific variations in parental<br />

behavior of the white-throated sparrow. Wilson Bull. 105, 48–59.<br />

Kotegawa, T., Abe, T., and Tsutsui, K. (1997). Inhibitory role of opioid peptides in the regulation of<br />

aggressive and sexual behaviors in male Japanese quails. J. Exp. Zool. 277, 146–154.<br />

Krebs, J. R. (1971). Territory and breeding density in the great tit, Parus major. Ecology 52, 2–22.<br />

Krebs, J. (1976). Bird song and territorial defence. New Sci. 70, 534–536.


5. Neurogenomic Mechanisms of Aggression in Songbirds 115<br />

Kroodsma, D. E. (1976). The effect of large song repertoires on neighbor “recognition” in male song<br />

sparrows. Condor 78, 97–99.<br />

Kroodsma, D. E., Bereson, R. C., Byers, B. E., and Minear, E. (1989). Use of song types by the<br />

chestnut-sided warbler: Evidence for both intra- and inter-sexual functions. Can. J. Zool. 67,<br />

447–456.<br />

Lein, M. R. (1978). Song variation in a population of chestnut-sided warblers (Dendroica pensylvanica):<br />

Its nature and suggested significance. Can. J. Zool. 56, 1266–1283.<br />

Lin, D., Boyle, M. P., Dollar, P., Lee, H., Lein, E. S., Perona, P., and Anderson, D. J. (2011).<br />

Functional identification of an aggression locus in the mouse hypothalamus. Nature 470,<br />

221–226.<br />

Lowther, J. K. (1961). Polymorphism in the white-throated sparrow, Zonotrichia albicollis (Gmelin).<br />

Can. J. Zool. 39, 281–292.<br />

Lumia, A. R. (1972). The relationships among testosterone, conditioned aggression, and dominance<br />

in male pigeons. Horm. Behav. 3, 277–286.<br />

Maney, D. L. (2008). Endocrine and genomic architecture of life history trade-offs in an avian model<br />

of social behavior. Gen. Comp. Endocrinol. 157, 275–282.<br />

Maney, D. L. (2010). Hormonal control of sexual behavior in female nonmammalian vertebrates.<br />

In “Encyclopedia of Animal Behavior” (M. D. Breed and J. Moore, eds.), Encyclopedia of Animal<br />

Behavior,Vol. 1, pp. 697–703. Elsevier, Oxford.<br />

Maney, D. L., and Ball, G. F. (2003). Fos-like immunoreactivity in catecholaminergic brain nuclei<br />

after territorial behavior in free-living song sparrows. J. Neurobiol. 56, 163–170.<br />

Maney, D. L., Goode, C. T., and Wingfield, J. C. (1997). Intraventricular infusion of arginine<br />

vasotocin induces singing in a female songbird. J. Neuroendocrinol. 9, 487–491.<br />

Maney, D. L., Erwin, K. L., and Goode, C. T. (2005). Neuroendocrine correlates of behavioral<br />

polymorphism in white-throated sparrows. Horm. Behav. 48, 196–206.<br />

Maney, D. L., Lange, H. S., Raees, M. Q., and Sanford, S. E. (2009). Behavioral phenotypes persist<br />

after gonadal steroid manipulation in white-throated sparrows. Horm. Behav. 55, 113–120.<br />

Mathewson, S. F. (1961). Gonadotrophic hormones affect aggressive behavior in starlings. Science<br />

134, 1522–1523.<br />

Maynard-Smith, J. (1977). Parental investment: A prospective analysis. Anim. Behav. 25, 1–9.<br />

McGlothlin, J. W., and Ketterson, E. D. (2007). Hormone-mediated suites as adaptations and<br />

evolutionary constraints. Philos. Trans. R. Soc. Lond. B Biol. Sci. 170, 864–875.<br />

McGlothlin, J. W., Jawor, J. M., and Ketterson, E. D. (2007). Natural variation in a testosteronemediated<br />

trade-off between mating effort and parental effort. Am. Nat. 170, 864–875.<br />

Mello, C. V., and Ribeiro, S. (1998). ZENK protein regulation by song in the brain of songbirds.<br />

J. Comp. Neurol. 393, 426–438.<br />

Mello, C. V., Vicario, D. S., and Clayton, D. F. (1992). Song presentation induces gene expression in<br />

the songbird forebrain. Proc. Natl. Acad. Sci. USA 89, 6818–6822.<br />

Miles, D. B., Sinervo, B., Hazard, L. C., Svensson, E. I., and Costa, D. (2007). Relating endocrinology,<br />

physiology and behaviour using species with alternative mating strategies. Funct. Ecol. 21,<br />

653–665.<br />

Møller, A. P. (1987). Variation in badge size in male house sparrows Passer domesticus: Evidence for<br />

status signalling. Anim. Behav. 35, 1637–1644.<br />

Moore, M. C. (1983). Effect of female displays on the endocrine physiology and behavior of male<br />

white-crowned sparrows, Zonotrichia leucophrys. J. Zool. 199, 137–148.<br />

Moore, M. C. (1991). Application of organization-activation theory to alternative male reproductive<br />

strategies: A review. Horm. Behav. 25, 154–179.<br />

Motta, S. C., Goto, M., Gouveia, F. V., Baldo, M. V. C., Canteras, N. S., and Swanson, L. W. (2009).<br />

Dissecting the brain’s fear system reveals the hypothalamus is critical for responding in subordinate<br />

conspecific intruders. Proc. Natl. Acad. Sci. 106, 4870–4875.


116 Maney and Goodson<br />

Moyer, K. E. (1968). Kinds of aggression and their physiological basis. Commun. Behav. Biol. 2(A),<br />

65–87.<br />

Mukai, M., Replogle, K., Drnevich, J., Wang, G., Wacker, D., Band, M., Clayton, D. F., and<br />

Wingfield, J. C. (2009). Seasonal differences of gene expression profiles in song sparrow (Melospiza<br />

melodia) hypothalamus in relation to territorial aggression. PLoS One 4, e8182.<br />

Nelson, D. A., and Croner, L. J. (1991). Song categories and their functions in the field sparrow<br />

(Spizella pusilla). Auk 108, 42–52.<br />

Newman, S. W. (1999). The medial extended amygdala in male reproductive behavior: A node in<br />

the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877, 242–257.<br />

Nice, M. M. (1943). Studies in the Life History of the Song Sparrow. II. The Behaviour of the Song<br />

Sparrow and Other Passerines. Dover, New York.<br />

Nijhout, F. H. (2003). Development and evolution of adaptive polyphenisms. Evol. Dev. 5, 9–18.<br />

Nolan, Jr., V., Ketterson, E. D., Cristol, D. A., Rogers, C. M., Clotfelter, E. D., Titus, R. C., Schoech,<br />

S. J., and Snajdr, E. (2002). Dark-eyed Junco (Junco hyemalis). In “The Birds of North America<br />

Online” (A. Poole, ed.). Cornell Lab of Ornithology, Ithaca. doi:10.2173/bna.716.<br />

Panzica, G. C., Aste, N., Castagna, C., Viglietti-Panzica, C., and Balthazart, J. (2001). Steroidinduced<br />

plasticity in the sexually dimorphic vasotocinergic innervation of the avian brain:<br />

Behavioral implications. Brain Res. Rev. 37, 178–200.<br />

Piper, W. H., and Wiley, R. H. (1989). Correlates of dominance in wintering white-throated<br />

sparrows: Age, sex and location. Anim. Behav. 37, 298–310.<br />

Pradhan, D. S., Newman, A. E., Wacker, D. W., Wingfield, J. C., Schlinger, B. A., and Soma, K. K.<br />

(2010). Aggressive interactions rapidly increase androgen synthesis in the brain during the nonbreeding<br />

season. Horm. Behav. 57, 381–389.<br />

Ramenofsky, M. (1984). Agonistic behavior and endogenous plasma hormones in male Japanese<br />

quail. Anim. Behav. 32, 698–708.<br />

Ramirez, J. M., Salas, C., and Portavella, M. (1988). Offense and defense after lateral septal lesions in<br />

Columba livia. Int. J. Neurosci. 41, 241–250.<br />

Remage-Healey, L., Maidment, N. T., and Schlinger, B. A. (2008). Forebrain steroid levels fluctuate<br />

rapidly during social interactions. Nat. Neurosci. 11, 1327–1334.<br />

Remage-Healey, L., London, S. E., and Schlinger, B. A. (2010). Birdsong and the neural production<br />

of steroids. J. Chem. Neuroanat. 39, 72–81.<br />

Rhen, T., and Crews, D. (2002). Variation in reproductive behaviour within a sex: Neural systems<br />

and endocrine activation. J. Neuroendocrinol. 14, 517–531.<br />

Robinson, G. E., Grozinger, C. M., and Whitfield, C. W. (2005). Sociogenomics: Social life in<br />

molecular terms. Nat. Rev. Genet. 6, 257–270.<br />

Rohwer, S. (1975). The social significance of avian winter plumage variability. Evolution 29,<br />

593–610.<br />

Rohwer, S., and Rohwer, F. C. (1978). Status signalling in Harris sparrows: Experimental deceptions<br />

achieved. Anim. Behav. 26, 1012–1022.<br />

Romanov, M. N., Tuttle, E. M., Houck, M. L., Modi, W. S., Chemnick, L. G., Korody, M. L., et al.<br />

(2009). The value of avian genomics to the conservation of wildlife. BMC Genomics 10, S10.<br />

Sabine, W. S. (1959). The winter society of the Oregon Junco: Intolerance, dominance, and the<br />

pecking order. Condor 61, 110–135.<br />

Schlinger, B. A. (1987). Plasma androgens and aggressiveness in captive winter white-throated<br />

sparrows (Zonotrichia albicollis). Horm. Behav. 21, 203–210.<br />

Schneider, K. C. J. (1984). Dominance, predation, and optimal foraging in white-throated sparrow<br />

flocks. Ecology 65, 1820–1827.<br />

Schoech, S. J., Ketterson, E. D., Nolan, V., Sharp, P. J., and Buntin, J. D. (1998). The effect of<br />

exogenous testosterone on parental behavior, plasma prolactin, and prolactin binding sites in the<br />

dark-eyed juncos. Horm. Behav. 34, 1–10.


5. Neurogenomic Mechanisms of Aggression in Songbirds 117<br />

Schroeder, M. B., and Riters, L. V. (2006). Pharmacological manipulations of dopamine and opioids<br />

have differential effects on sexually motivated song in male European starlings. Physiol. Behav. 88,<br />

575–584.<br />

Schwabl, H., Ramenofsky, M., Schwabl-Benzinger, I., Farner, D. S., and Wingfield, J. C. (1988).<br />

Social status, circulating levels of hormones, and competition for food in winter flocks of the<br />

white-throated sparrow. Behaviour 107, 107–121.<br />

Selinger, H. E., and Bermant, G. (1967). Hormonal control of aggressive behavior in Japanese quail<br />

(Coturnix coturnix japonica). Behavior 28, 255–268.<br />

Silverin, B. (1980). Effects of long-acting testosterone treatment on free living pied flycatchers,<br />

Ficedula hypoleuca, during the breeding season. Anim. Behav. 28, 906–912.<br />

Sinervo, B., and Svensson, E. (2002). Correlational selection and the evolution of genomic<br />

architecture. Heredity 89, 329–338.<br />

Soma, K. K. (2006). Testosterone and aggression: Berthold, birds and beyond. J. Neuroendocrinol. 18,<br />

543–551.<br />

Soma, K. K., and Wingfield, J. C. (2001). Dehydroepiandrosterone in songbird plasma: Seasonal<br />

regulation and relationship to territorial aggression. Gen. Comp. Endocrinol. 123, 144–155.<br />

Soma, K. K., Sullivan, K., and Wingfield, J. C. (1999). Combined aromatase inhibitor and antiandrogen<br />

treatment decreases territorial aggression in a wild songbird during the nonbreeding<br />

season. Gen. Comp. Endocrinol. 115, 442–453.<br />

Soma, K. K., Tramontin, A. D., and Wingfield, J. C. (2000a). Oestrogen regulates male aggression in<br />

the non-breeding season. Proc. R. Soc. Lond. B Biol. Sci. 267, 1089–1096.<br />

Soma, K. K., Sullivan, K. A., Tramontin, A. D., Saldanha, C. J., Schlinger, B. A., and Wingfield, J. C.<br />

(2000b). Acute and chronic effects of an aromatase inhibitor on territorial aggression in breeding<br />

and nonbreeding male song sparrows. J. Comp. Physiol. A 186, 759–769.<br />

Soma, K. K., Schlinger, B. A., Wingfield, J. C., and Saldanha, C. J. (2003). Brain aromatase, 5-alpha<br />

reductase, and 5-beta reductase change seasonally in wild male song sparrows: Relationship to<br />

aggressive and sexual behavior. J. Neurobiol. 56, 209–221.<br />

Soma, K. K., Scotti, M. A., Newman, A. E., Charlier, T. D., and Demas, G. E. (2008). Novel<br />

mechanisms for neuroendocrine regulation of aggression. Front. Neuroendocrinol. 29, 476–489.<br />

Spinney, L. H., Bentley, G. E., and Hau, M. (2006). Endocrine correlates of alternative phenotypes in<br />

the white-throated sparrow (Zonotrichia albicollis). Horm. Behav. 50, 762–771.<br />

Storey, A. E., Walch, C. J., Quinton, R. L., and Wynne-Edwards, K. E. (2000). Hormonal correlates<br />

of paternal responsiveness in new and expectant fathers. Evol. Hum. Behav. 21, 79–95.<br />

Swett, M. B., and Breuner, C. W. (2009). Plasma testosterone correlates with morph type across<br />

breeding substages in male white-throated sparrows. Physiol. Biochem. Zool. 82, 572–579.<br />

Thomas, J. W., Cáceres, M., Lowman, J. J., Morehouse, C. B., Short, M. E., Baldwin, E. L.,<br />

Maney, D. L., and Martin, C. L. (2008). The chromosomal polymorphism linked to variation in<br />

social behavior in the white-throated sparrow (Zonotrichia albicollis) is a complex rearrangement<br />

that suppresses recombination. Genetics 179, 1455–1468.<br />

Thorneycroft, H. B. (1975). A cytogenetic study of the white-throated sparrow, Zonotrichia albicollis.<br />

Evolution 29, 611–621.<br />

Tompkins, G. (1933). Individuality and territoriality as displayed in winter by three passerine species.<br />

Condor 35, 98–106.<br />

Trainor, B. C., Finy, M. S., and Nelson, R. J. (2008). Paternal aggression in a biparental mouse:<br />

Parallels with maternal aggression. Horm. Behav. 53, 200–207.<br />

Trivers, R. L. (1972). Parental investment and sexual selection. In “Sexual Selection and the Descent<br />

of Man” (B. Campbell, ed.), pp. 139–179. Aldine, Chicago.<br />

Tuttle, E. M. (1993). Mate choice and stable polymorphism in the white-throated sparrow. Ph.D,<br />

Dissertation, <strong>State</strong> <strong>University</strong> of New York at Albany.


118 Maney and Goodson<br />

Tuttle, E. M. (2003). Alternative reproductive strategies in the white-throated sparrow: Behavioral<br />

and genetic evidence. Behav. Ecol. 14, 425–432.<br />

Udry, J. R. (1990). Biosocial models of adolescent problem behaviors. Social Biol. 37, 1–10.<br />

Veenema, A. H., Bredewold, R., and Neumann, I. D. (2007). Opposite effects of maternal separation<br />

on intermale and maternal aggression in C57BL/6 mice: Link to hypothalamic vasopressin and<br />

oxytocin immunoreactivity. Psychoneuroendocrinology 32, 437–450.<br />

Voorhuis, T. A. M., Kiss, J. Z., de Kloet, E. R., and de Wied, D. (1988). Testosterone-sensitive<br />

vasotocin-immunoreactive cells and fibers in the canary brain. Brain Res. 442, 139–146.<br />

Wacker, D. W., Schlinger, B. A., and Wingfield, J. C. (2008). Combined effects of DHEA and<br />

fadrozole on aggression and neural VIP immunoreactivity in the non-breeding male song sparrow.<br />

Horm Behav. 53, 287–294.<br />

Wacker, D. W., Wingfield, J. C., Davis, J. E., and Meddle, S. L. (2010). Seasonal changes<br />

in aromatase and androgen receptor, but not estrogen receptor mRNA expression in the brain<br />

of the free-living male song sparrow, Melospiza melodia morphna. J. Comp. Neurol. 518,<br />

3819–3835.<br />

Walker, B. G., Boersma, P. D., and Wingfield, J. C. (2005). Field endocrinology and conservation<br />

biology. Integr. Comp. Biol. 45, 12–18.<br />

Warren, W. C., Clayton, D. F., Ellegren, H., Arnold, A. P., Hillier, L. W., Künstner, A., et al. (2010).<br />

The genome of a songbird. Nature 464, 757–762.<br />

Watt, D. J. (1986). Plumage brightness index for white-throated sparrows. J. Field Ornithol. 57,<br />

105–113.<br />

Watt, D. J., Ralph, C. J., and Atkinson, C. T. (1984). The role of plumage polymorphism in<br />

dominance relationships of the white-throated sparrow. Auk 101, 110–120.<br />

Wiley, R. H., Piper, W. H., Archawaranon, M., and Thompson, E. W. (1993). Singing in relation to<br />

social dominance and testosterone in white-throated sparrows. Behaviour 127, 175–190.<br />

Wiley, R. H., Steadman, L., Chawick, L., and Wollerman, L. (1999). Social inertia in white-throated<br />

sparrows results from recognition of opponents. Anim. Behav. 57, 453–463.<br />

Wingfield, J. C. (1984a). Environmental and endocrine control of reproduction in the song sparrow,<br />

Melospiza melodia I. Temporal organization of the breeding cycle. Gen. Comp. Endocrinol. 56,<br />

406–416.<br />

Wingfield, J. C. (1984b). Environmental and endocrine control of reproduction in the song sparrow,<br />

Melospiza melodia II. Agonistic interactions as environmental information stimulating secretion<br />

of testosterone. Gen. Comp. Endocrinol. 56, 417–424.<br />

Wingfield, J. C. (1984c). Androgens and mating systems: Testosterone-induced polygyny in normally<br />

monogamous birds. Auk 101, 665–671.<br />

Wingfield, J. C. (1985). Short-term changes in plasma levels of hormones during establishment and<br />

defense of a breeding territory in male song sparrows, Melospiza melodia. Horm. Behav. 19,<br />

174–187.<br />

Wingfield, J. C. (1994a). Control of territorial aggression in a changing environment. Psychoneuroendocrinology<br />

19, 709–721.<br />

Wingfield, J. C. (1994b). Regulation of territorial behavior in the sedentary song sparrow, Melospiza<br />

melodia morphna. Horm. Behav. 28, 1–15.<br />

Wingfield, J. C., and Farner, D. S. (1976). Avian endocrinology—Field investigations and methods.<br />

Condor 78, 570–573.<br />

Wingfield, J. C., and Farner, D. S. (1978). The endocrinology of a naturally breeding population of<br />

white-crowned sparrow, Zonotrichia leucophrys pugetensis. Physiol. Zool. 51, 188–205.<br />

Wingfield, J. C., and Farner, D. S. (1993). Endocrinology of reproduction in wild species. In “Avian<br />

Biology” (D. S. Farner, J. King, and K. Parkes, eds.), pp. 163–327. Academic Press, San Diego.


5. Neurogenomic Mechanisms of Aggression in Songbirds 119<br />

Wingfield, J. C., and Goldsmith, A. R. (1990). Plasma levels of prolactin and gonadal steroids in<br />

relation to multiple-brooding and renesting in free-living populations of the song sparrow,<br />

Melospiza melodia. Horm. Behav. 24, 89–103.<br />

Wingfield, J. C., and Hahn, T. P. (1994). Testosterone and territorial behavior in sedentary and<br />

migratory sparrows. Anim. Behav. 47, 77–89.<br />

Wingfield, J. C., and Monk, D. (1994). Behavioral and hormonal responses of male song sparrows to<br />

estradiol-treated females during the non-breeding season. Horm. Behav. 28, 146–154.<br />

Wingfield, J. C., and Wada, M. (1989). Male-male interactions increase both luteinizing hormone<br />

and testosterone in the song sparrow, Melospiza melodia: Specificity, time course and possible<br />

neural pathways. J. Comp. Physiol. A 166, 189–194.<br />

Wingfield, J. C., Ball, G. F., Dufty, A. M., Jr., Hegner, R. E., and Ramenofsky, M. (1987). Testosterone<br />

and aggression in birds: Tests of the “challenge hypothesis”. Am. Sci. 75, 602–608.<br />

Wingfield, J. C., Hegner, R. F., Dufty, A. M. J., and Ball, G. F. (1990). The challenge hypothesis:<br />

Theoretical implications for patterns of testosterone secretion, mating systems and breeding<br />

strategies. Am. Nat. 136, 829–846.<br />

Wingfield, J. C., Lynn, S. E., and Soma, K. K. (2001). Avoiding the ‘costs’ of testosterone: Ecological<br />

bases of hormone-behavior interactions. Brain Behav. Evol. 57, 239–251.<br />

Wynne-Edwards, K. E. (2001). Hormonal changes in mammalian fathers. Horm. Behav. 40,<br />

139–145.<br />

Yang, E. J., and Wilczynski, W. (2007). Social experience organizes parallel networks in sensory and<br />

limbic forebrain. Dev. Neurobiol. 67, 285–303.<br />

Zera, A. J., and Harshman, L. G. (2001). The physiology of life history trade-offs in animals. Annu.<br />

Rev. Ecol. Syst. 32, 95–126.


Intentionally left as blank


6<br />

Genetics of Aggression in Voles<br />

Kyle L. Gobrogge 1 and Zuoxin W. Wang<br />

Department of Psychology and Program in Neuroscience, Florida <strong>State</strong><br />

<strong>University</strong>, Tallahassee, Florida, USA<br />

I. Introduction<br />

II. The Prairie Vole Model<br />

III. Neural Correlates<br />

IV. Neural Circuitry<br />

V. Neurochemical Regulation of Selective Aggression<br />

A. Neuropeptides<br />

B. Dopamine<br />

C. Steroid hormones<br />

D. Classical neurotransmitters<br />

VI. Molecular Genetics of Selective Aggression<br />

VII. Drug-induced Aggression<br />

VIII. Conclusions and Future Directions<br />

Acknowledgments<br />

References<br />

ABSTRACT<br />

Prairie voles (Microtus ochrogaster) are socially monogamous rodents that form<br />

pair bonds—a behavior composed of several social interactions including attachment<br />

with a familiar mate and aggression toward conspecific strangers.<br />

1 Present address: Department of Neurobiology, Harvard Medical School, Boston,<br />

Massachusetts, USA<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00003-4


122 Gobrogge and Wang<br />

Therefore, this species has provided an excellent opportunity for the study of pair<br />

bonding behavior and its underlying neural mechanisms. In this chapter, we<br />

discuss the utility of this unique animal model in the study of aggression and<br />

review recent findings illustrating the neurochemical mechanisms underlying<br />

pair bonding-induced aggression. Implications of this research for our understanding<br />

of the neurobiology of human violence are also discussed. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

Mating induces aggression in several organisms throughout the animal kingdom.<br />

Within species, patterns of inter- and intrasexual aggression vary as a function of<br />

monogamy, parental investment, and group structure. In the wild, the appropriate<br />

coordination of social behavior is necessary for survival and reproductive<br />

success. How organisms make decisions about which behavior to display in the<br />

natural environment remains an important area of biological investigation. To<br />

address these questions, previous work has relied on using traditional laboratory<br />

rodents. However, these animals do not readily display certain types of social<br />

behaviors and thus are not appropriate for some investigations. For example,<br />

laboratory rats and mice do not exhibit strong social bonds between mates, and<br />

males typically do not display paternal behavior or female-directed aggression.<br />

Because mating naturally induces pair bonding, aggression, and biparental behavior<br />

in the socially monogamous prairie vole (Microtus ochrogaster), this<br />

species represents a unique animal model to study the underlying neural mechanisms<br />

regulating social behavior associated with a monogamous life strategy.<br />

In this chapter, we begin by describing the prairie vole model and<br />

reviewing the neural correlates of pair bonding behavior. We focus on the<br />

neuropeptides arginine vasopressin (AVP) and oxytocin; neurotransmitters dopamine<br />

(DA), gamma-aminobutyric acid (GABA), and glutamate; and steroid<br />

hormones testosterone and estrogen in the regulation of aggression. We highlight<br />

the molecular genetics underlying courtship and aggression associated with<br />

monogamous pair bonding in voles and humans. Finally, we speculate on the<br />

potential for translation, of aggression studies in prairie voles, for research<br />

examining the etiology of violence in human populations—with a particular<br />

emphasis on the interactions between drug abuse and social behavior.<br />

II. THE PRAIRIE VOLE MODEL<br />

Voles are microtine (Microtus) rodents that are taxonomically and genetically<br />

similar, yet show remarkable differences in their social behavior (Young and<br />

Wang, 2004; Young et al., 2008, 2011a). These animals have provided an


6. Genetics of Aggression in Voles 123<br />

excellent opportunity for comparative studies examining social behaviors associated<br />

with different life strategies. For example, prairie (M. ochrogaster) and pine<br />

(M. pinetorum) voles are highly social and monogamous, whereas meadow<br />

(M. pennsylvanicus) and montane (M. montanus) voles are asocial and promiscuous<br />

(Dewsbury, 1987; Insel and Hulihan, 1995; Jannett, 1982). In the laboratory,<br />

prairie and pine voles are biparental, with both parents equally caring for their<br />

young, while meadow and montane voles are primarily maternal and males do<br />

not stay in the natal nest after female parturition (McGuire and Novak, 1984,<br />

1986; Oliveras and Novak, 1986). Following mating, prairie voles develop pair<br />

bonds between mates (Fig. 6.1A; Young and Wang, 2004) and males even display<br />

aggression selectively toward conspecific strangers but not toward their familiar<br />

partner (Fig. 6.1B; Aragona et al., 2006; Gobrogge et al., 2007, 2009; Winslow<br />

et al., 1993)—behaviors that are not exhibited by promiscuous meadow or<br />

montane voles (Insel et al., 1995; Lim et al., 2004). Interestingly, these vole<br />

species do not differ in their nonsocial behavior (Tamarin, 1985), further<br />

indicating associations between species-specific social behavior and life strategy<br />

(Carter et al., 1995; Insel et al., 1998; Wang and Aragona, 2004; Young and<br />

Wang, 2004; Young et al., 1998). Therefore, prairie voles represent a unique<br />

model system to dissect the neural mechanisms underlying ethologically relevant<br />

social behavior.<br />

One behavioral index of pair bonding is selective aggression, which is<br />

more prominent in male than in female prairie voles. It has been suggested that<br />

selective aggression is a behavioral trait associated with mate guarding that is<br />

important for pair bonding (Carter et al., 1995). Selective aggression is studied<br />

using a resident–intruder test (RIT) (Fig. 6.1C; Gobrogge et al., 2007, 2009;<br />

Winslow et al., 1993; Wang et al., 1997a). A conspecific intruder is introduced<br />

into the male resident cage and their behavioral interactions are videotaped for<br />

5–10 min (Aragona et al., 2006; Gobrogge et al., 2007, 2009; Wang et al., 1997a;<br />

Winslow et al., 1993). Subject’s behavioral interactions with the intruder are<br />

recorded and the frequency of aggressive behaviors including attacks, bites,<br />

chases, defensive/offensive upright postures, offensive sniffs, threats, and retaliatory<br />

attacks are calculated as a composite score (Gobrogge et al., 2007, 2009) as<br />

well as the duration of affiliative side-by-side physical contact (Gobrogge et al.,<br />

2007, 2009; Winslow et al., 1993). It is important to note that both offensive and<br />

defensive types of aggression are critical components of selective aggression in<br />

male prairie voles (Wang et al., 1997a; Winslow et al., 1993).<br />

Selective aggression is associated with mating, as cohabitation in the<br />

absence of mating does not induce this behavior in male prairie voles (Insel et al.,<br />

1995; Wang et al., 1997a; Winslow et al., 1993). Selective aggression is also<br />

enduring (Aragona et al., 2006; Gobrogge et al., 2007, 2009; Insel et al., 1995)<br />

and lasts for at least 2 weeks after partner preference formation (Aragona et al.,<br />

2006; Gobrogge et al., 2007, 2009), even in the absence of continuous exposure


124 Gobrogge and Wang<br />

A<br />

B<br />

C<br />

Contact time (min/3 h)<br />

D<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Partner<br />

Stranger<br />

6 h Cohab<br />

*<br />

24 h Mating<br />

Aggression (# attacks/10 min)<br />

E<br />

120<br />

80<br />

40<br />

0<br />

Naive<br />

a<br />

Stranger<br />

Paired<br />

a<br />

b<br />

Partner Stranger Stranger<br />

female male<br />

g<br />

Partner<br />

F<br />

Stranger<br />

Density Fos-ir (#/mm 3 )<br />

2000<br />

1600<br />

1200<br />

800<br />

400<br />

0<br />

AH<br />

Partner<br />

Stranger<br />

*<br />

MeA<br />

*<br />

AH<br />

OT<br />

MeA<br />

AcA<br />

OT<br />

Figure 6.1. Neural correlates of selective aggression. (A) After 24 h, but not 6 h, of mating, male<br />

and female prairie voles display significantly more time in side-by-side physical contact<br />

with an opposite sex familiar partner than with a stranger. (B) Sexually naïve male<br />

prairie voles (Naïve) do not display aggression toward a stranger female, whereas 2 weeks<br />

of sexual and social experience (Paired) induces selective aggression toward both male<br />

and female strangers but not toward familiar female partners. (C) Photo depicts a pair<br />

bonded male prairie vole (left) preparing to attack a—sexually receptive—stranger<br />

female prairie vole (right). (D) Stereological estimates reveal a significantly higher<br />

density of Fos-ir cells in the anterior hypothalamus (AH) and medial amygdala<br />

(MeA) of pair bonded males displaying aggression toward a stranger female (Stranger)<br />

compared to males displaying affiliation toward their familiar female partner (Partner).<br />

(E) Photomicrographs showing Fos-ir (dark nuclear staining) in the AH and MeA from<br />

pair bonded males re-exposed to their familiar female partner (Partner) or to a stranger<br />

female (Stranger). F: fornix; OT: optic tract. Bars indicate meansstandard error of the<br />

mean. Bars with different Greek letters differ significantly from each other. *p


6. Genetics of Aggression in Voles 125<br />

et al., 2007, 2009; Wang et al., 1997a). Together, these reliably expressed and<br />

measurable agonistic behaviors make the prairie vole an excellent model for<br />

investigation of the neural mechanisms underlying naturally occurring aggression<br />

associated with monogamy. It should be mentioned that although female<br />

aggression has been less studied in voles, female prairie voles exhibit similar<br />

aggressive behavior as males and this behavior is influenced by a female’s social<br />

and sexual experience (Bowler et al., 2002).<br />

III. NEURAL CORRELATES<br />

With considerable overlap of brain areas involved in several forms of social<br />

(Newman, 1999) and agonistic (Table 6.1) behaviors, there is a significant<br />

amount of ambiguity regarding which brain areas may be involved in the<br />

regulation of selective aggression. Using a neuronal activation marker of an<br />

immediate early gene, c-fos, previous studies in voles have examined neuronal<br />

Table 6.1. Summary of Brain Areas Implicated in Aggression<br />

Brain area Species References<br />

Anterior hypothalamus (AH) Human Sano et al. (1966), Ramamurthi (1988)<br />

Prairie vole Gobrogge et al. (2007, 2009), Gobrogge and<br />

Wang (2009)<br />

Rat<br />

Veening et al. (2005), Kruk (1991), Bermond<br />

et al. (1982), Kruk et al. (1984), Adams et al.<br />

(1993), Roeling et al. (1993), Haller et al.<br />

(1998), Veenema et al. (2006), Motta et al.<br />

(2009)<br />

Syrian hamster Delville et al. (2000), Ferris and Potegal (1988),<br />

Caldwell and Albers (2004), Ferris et al. (1997,<br />

1989), Albers et al. (2006), Harrison et al.<br />

(2000b), Jackson et al. (2005), Grimes et al.<br />

(2007), Ricci et al. (2009), Schwartzer et al.<br />

(2009), Schwartzer and Melloni (2010a,b)<br />

Lateral septum (LS) Rat Veenema et al. (2010)<br />

Medial amygdala (MeA) Human Ramamurthi (1988)<br />

Prairie vole Wang et al. (1997a), Gobrogge and Wang (2009)<br />

Rat Koolhaas et al. (1990)<br />

Nucleus accumbens (NAcc) Prairie vole Aragona et al. (2006)<br />

Ventromedial hypothalamus<br />

(VMH)<br />

Mouse Choi et al. (2005), Lin et al. (2011)<br />

Brain structures involved in aggression, across species, with corresponding references to ground<br />

brain area acronyms used throughout the chapter.


126 Gobrogge and Wang<br />

activity associated with aggression (Wang et al., 1997a), maternal (Katz et al.,<br />

1999) and paternal behavior (Kirkpatrick et al., 1994), mating (Curtis and<br />

Wang, 2003; Lim and Young, 2004), anxiety (Stowe et al., 2005), spatial learning<br />

(Kuptsov et al., 2005), chemosensory processing (Hairston et al., 2003; Tubbiola<br />

and Wysocki, 1997), social experience (Cushing et al., 2003a; Kramer et al.,<br />

2006), or pharmacological challenges (Curtis and Wang, 2005b; Cushing et al.,<br />

2003b; Gingrich et al., 1997). Within these studies, however, typically only one<br />

type of behavior was investigated. Little focus was aimed at examining other<br />

forms of social behavior, including affiliation or general social olfactory processing.<br />

Consequently, there is considerable overlap in brain–behavior relationships<br />

among these studies leading to ambiguity as to which brain areas regulate<br />

selective aggression. Nevertheless, in an early study, male prairie voles displayed<br />

aggression toward a male intruder following 24 h of mating, but not following<br />

24 h of cohabitation with a female without mating (Wang et al., 1997a).<br />

However, despite their differences in sociosexual experience and in aggressive<br />

behavior, both types of male exposure led to equal levels of Fos-ir (immunoreactivity)<br />

expression in some brain areas, such as the bed nucleus of the stria<br />

terminalis (BNST). Males that mated for 24 h and displayed high levels of<br />

aggression toward either a male or a female intruder showed increased levels of<br />

Fos-ir expression in the medial amygdala (MeA; Wang et al., 1997a), compared<br />

to males that cohabitated with a female without mating, implicating the MeA as<br />

a brain area associated with the display of mating-induced selective aggression<br />

(Fig. 6.1D and E).<br />

In a more recent study, several brain areas including the BNST, medial<br />

preoptic area (MPOA), paraventricular nucleus (PVN), and lateral septum (LS)<br />

showed higher levels of Fos expression in pair bonded males that had experienced<br />

an RIT compared to pair bonded males not exposed to a social intruder<br />

(Gobrogge et al., 2007). However, no group differences in Fos expression across<br />

these brain areas were found among males that were exposed to different social<br />

stimuli or displaying different patterns of social behavior, including aggression or<br />

affiliation toward intruders. These data suggest that the increased neuronal<br />

activation in these brain regions is probably due to olfactory stimulation or<br />

general arousal associated with exposure to a conspecific, but such a response is<br />

nonselective. A unique pattern of Fos expression was found in the anterior<br />

hypothalamus (AH), in which exposure to a conspecific stranger, either male<br />

or female, induced a significant increase in AH-Fos over those reexposed to their<br />

familiar partner (Fig. 6.1D and E; Gobrogge et al., 2007). This increase in Fos<br />

staining may indicate a stimulus-specific response. The AH appears to be more<br />

responsive to chemosensory, tactile, and/or visual cues associated with conspecific<br />

strangers, but not familiar partners (Gobrogge et al., 2007). These data<br />

indicate that the increased neuronal activation in the AH may be involved in<br />

aggressive behavior displayed by pair bonded male prairie voles (Gobrogge et al.,


6. Genetics of Aggression in Voles 127<br />

2007). This notion is corroborated by previous research documenting a critical<br />

role of the hypothalamus in regulating aggression across several mammalian<br />

species. For example, the ventromedial hypothalamus (VMH) in mice (Choi<br />

et al., 2005; Lin et al., 2011) and AH in rats (Veening et al., 2005) is responsive<br />

to conspecific chemosensory cues, which elicit aggressive behavior. Electrical<br />

stimulation applied directly to the AH induces attack toward conspecifics in rats<br />

(Kruk, 1991) and other animals (Albert and Walsh, 1984; Siegel et al., 1999).<br />

Interestingly, in humans, surgical lesioning of the AH reduces physical violence<br />

(Ramamurthi, 1988; Sano et al., 1966). In summary, data from vole studies<br />

demonstrate that activation of the MeA and AH is associated with the display<br />

of selective aggression (Gobrogge et al., 2007; Wang et al., 1997a).<br />

IV. NEURAL CIRCUITRY<br />

To directly evaluate the neural circuitry programming selective aggression, we<br />

performed a series of tract tracing experiments focusing on the AH and MeA.<br />

Intra-AH injections of an anterograde tracer, biotinylated dextran amine<br />

(BDA), resulted in BDA-ir staining in several brain regions. The AH projected<br />

to areas involved in processing chemosensory cues including the MeA; areas<br />

important for regulating social behavior including the LS, BNST, MPOA, VMH,<br />

and dorsal raphe (DR); and areas coordinating motor output, such as the<br />

periaqueductal gray (Gobrogge and Wang, 2009). The AH also projected to<br />

brain areas implicated in evaluating incentive salience including the medial<br />

prefrontal cortex (mPFC), nucleus accumbens (NAcc), and ventral pallidum<br />

(VP), as well as areas involved in memory formation and consolidation including<br />

hippocampal regions CA3 and the dentate gyrus (Gobrogge and Wang, 2009).<br />

Further, site-specific injections of a retrograde tracer, fluorogold (FG), into the<br />

LS, NAcc, or MeA resulted in FG-ir staining in the AH, indicating reciprocal<br />

connections between the AH and regions involved in motivation and chemosensory<br />

communication (Gobrogge and Wang, 2009).<br />

Fos-ir staining was also used to assess neuronal activation, in this circuit,<br />

associated with the display of pair bonding behavior. Males displaying aggression<br />

toward an unfamiliar female showed a significantly higher density of Fos-ir in the<br />

AH and MeA relative to males displaying affiliation toward their female partner<br />

(Fig. 6.1D and E), replicating our previous findings (Gobrogge et al., 2007; Wang<br />

et al., 1997a). Interestingly, we identified a MeA-AH-LS circuit that was activated<br />

when males were displaying aggression and a DR-AH circuit that was<br />

recruited when males were displaying affiliation (Gobrogge and Wang, 2009).<br />

The identification of these two neural circuits indicates a specific neuronal<br />

framework associated with the choice between affiliation (DR-AH circuit) and<br />

aggression (MeA-AH-LS circuit) in pair bonded male prairie voles.


128 Gobrogge and Wang<br />

V. NEUROCHEMICAL REGULATION OF SELECTIVE AGGRESSION<br />

Previous work has primarily focused on partner preference formation and documented<br />

a growing list of neurochemicals, including oxytocin (OT), AVP, corticotropin<br />

releasing hormone, DA, GABA, and glutamate, as well as their<br />

interactions in the regulation of pair bonding behavior (Aragona et al., 2003;<br />

Curtis and Wang, 2005a,b; Carter et al., 1995; DeVries et al., 1995; Gingrich<br />

et al., 2000; Lim and Young, 2004; Liu and Wang, 2003; Lim et al., 2004; Liu<br />

et al., 2001; Smeltzer et al., 2006; Wang et al., 1998, 1999; Williams et al., 1992,<br />

1994; Winslow et al., 1993). Importantly, data from several studies indicate a<br />

select subset of neurochemicals in the regulation of selective aggression.<br />

A. Neuropeptides<br />

Comparative approaches have been utilized in studies examining neuroendocrine<br />

mechanisms regulating social behavior in voles (Insel, 2010). Studies have<br />

focused on examining the central AVP system, a nine amino acid neuropeptide<br />

with diverse forebrain projections, across monogamous and promiscuous vole<br />

species. AVP is an antidiuretic hormone and has been shown to stimulate three<br />

structurally distinct receptors V1a, V1b, and V2, each activating very specific<br />

second messenger systems (Michell et al., 1979). Classically, AVP was first<br />

described as a primary homeostatic factor controlling kidney water reabsorption,<br />

blood volume/pressure, and vasodilatation in the peripheral nervous system.<br />

AVP and its receptors have been shown to be widely expressed in the central<br />

nervous system (Thibonnier, 1992), within specific brain regions (Johnson et al.,<br />

1993).<br />

The V1a AVP receptor subtype (V1aR), in particular, has been extensively<br />

studied in the regulation of social behavior (Insel et al., 1994) including<br />

aggression (Albers et al., 2006; Cooper et al., 2005; Ferris et al., 2006; Winslow<br />

et al., 1993). V1aRs are directly coupled to stimulatory (s) Gq-11 proteins<br />

(Thibonnier et al., 1993). Stimulation of these G-proteins leads to activation<br />

of adenylate cyclase, cAMP, protein kinase C, and phospholipases C, A 2 , and D<br />

(Raggenbass et al., 1991; Thibonnier, 1992; Thibonnier et al., 1992, 1994)<br />

enhancing calcium influx through L-type calcium channels (Son and Brinton,<br />

2001). Such activation enhances learning and memory in the aging brain (Deyo<br />

et al., 1989; Yamada et al., 1996) via direct effects on gene expression (Murphy<br />

et al., 1991).<br />

Because AVP regulates species-specific social behaviors such as aggression<br />

(Ferris et al., 1984; Ryding et al., 2008), it was hypothesized that the<br />

organization of central AVP systems may differ between monogamous and<br />

promiscuous vole species (Bamshad et al., 1993b; Insel and Shapiro, 1992). To<br />

test this hypothesis, the distribution pattern of AVP cells, fibers, and receptors


6. Genetics of Aggression in Voles 129<br />

were mapped in the vole brain. In all vole species examined, AVP-ir neurons<br />

were found in several brain regions, including the PVN and SON (supraoptic<br />

nucleus) of the hypothalamus, the BNST, MeA, AH, and MPOA (Bamshad<br />

et al., 1993b; Gobrogge et al., 2007; Wang, 1995; Wang et al., 1996). AVP-ir<br />

fibers were localized in the LS, lateral habenular nucleus, diagonal band, BNST,<br />

MPOA, and MeA (Bamshad et al., 1993b; Wang et al., 1996). Overall, AVP<br />

distribution patterns were highly conserved between monogamous and promiscuous<br />

vole species (Wang, 1995; Wang et al., 1996). Dramatic species differences<br />

in the distribution patterns and regional densities of V1aRs, however, were<br />

observed between vole species exhibiting different life strategies (Hammock<br />

and Young, 2002). For example, prairie voles have higher densities of V1aRs in<br />

the BNST, VP, central and basolateral nuclei of the amygdala, and accessory<br />

olfactory bulb, whereas montane voles exhibit a higher density of V1aRs in the<br />

LS and mPFC (Insel et al., 1994; Lim et al., 2004; Smeltzer et al., 2006; Wang<br />

et al., 1997c; Young et al., 1997). Further, prairie and pine voles exhibit similar<br />

patterns of V1aR binding, which differ from that of promiscuous meadow and<br />

montane voles (Insel et al., 1994; Lim et al., 2004). Species differences in V1aR<br />

distribution are stable across the lifespan (Wang et al., 1997b,c) and are receptorspecific,<br />

as no species differences are found in either the benzodiazepine or opiate<br />

receptor systems (Insel and Shapiro, 1992). In addition, monogamous prairie and<br />

promiscuous meadow voles differ in central AVP activity during mating and<br />

reproduction (Bamshad et al., 1993a; Wang et al., 1994). Because of these<br />

anatomical and functional differences, central AVP was thought to underlie<br />

selective aggression in male prairie voles (Winslow et al., 1993).<br />

Among the neuropeptides underlying aggression (Miczek et al., 2007;<br />

Siever, 2008), AVP, and its homolog vasotocin, have been found to regulate<br />

several forms of aggression across species (Caldwell et al., 2008; Riters and<br />

Panksepp, 1997) and diverse taxa (Backstrom and Winberg, 2009; Goodson,<br />

2008). In humans, central AVP correlates with aggressive behavior (Coccaro<br />

et al., 1998) and mediates anger (Thompson et al., 2004). Thus, the central AVP<br />

system may have evolved to be primed by a wide variety of experiences to induce<br />

aggression, when appropriate, in social animals (Donaldson and Young, 2008).<br />

Because central AVP underlies territorial aggression in other rodents (Ferris<br />

et al., 1984), AVP was proposed to regulate mating-induced aggression in prairie<br />

voles. In a pharmacological study, injections of a V1aR antagonist (V1aR Ant)<br />

into the lateral ventricle of male prairie voles blocked selective aggression<br />

induced by mating whereas injections of AVP-induced aggression toward an<br />

intruder in the absence of mating (Winslow et al., 1993). These effects were<br />

neuropeptide specific, as intracerebroventricular (ICV) infusion of an OT receptor<br />

antagonist had no effect on mating-induced aggression (Winslow et al.,<br />

1993). Importantly, developmental exposure to either AVP in male prairie<br />

voles (Stribley and Carter, 1999) or OT in female prairie voles (Bales and


130 Gobrogge and Wang<br />

Carter, 2003) facilitates aggression in adulthood. Together, these data highlight<br />

a critical role of neuropeptide regulation of prairie vole aggression. However, the<br />

action site and release dynamics of neuropeptides in the regulation of selective<br />

aggression were unclear.<br />

In a more recent study, it was found that the display of selective aggression<br />

was associated with increased neuronal activation in the AH, specifically in<br />

neurons expressing AVP (Fig. 6.2A andB;Gobrogge et al., 2007). In a previous<br />

study in Syrian hamsters (Mesocricetus auratus), aggression has also been shown to<br />

be associated with an increase in AVP-ir/Fos-ir double-labeled cells in the nucleus<br />

circularis (NC), medial SON (mSON), and surrounding areas ventral to the fornix<br />

in the AH (Delville et al., 2000). Our data, combined with previous results from<br />

other species, suggest that the AH may be a brain area in which AVP regulates<br />

selective aggression. Indeed, AH-AVP has been implicated in various forms of<br />

aggressive behavior. In Syrian hamsters, for example, blockade of V1aRs in the<br />

AH diminished offensive aggression (Caldwell and Albers, 2004; Ferris and<br />

Potegal, 1988) whereas intra-AH administration of a V1aR agonist enhanced<br />

aggressive behavior (Caldwell and Albers, 2004; Ferris et al.,1997). More recently,<br />

the density of V1aRs in the AH has been found to increase significantly in Syrian<br />

hamsters after their display of offensive aggression (Albers et al., 2006).<br />

Because selective aggression was associated with neuronal activation in<br />

the AH, specifically in AVP-containing neurons (Fig. 6.2AandB;Gobrogge et al.,<br />

2007), we tested the hypothesis that selective aggression is associated with AH-<br />

AVP release. In vivo brain microdialysis, with ELISA, was performed on male<br />

prairie voles that were pair bonded for 2 weeks. Pair bonded males displayed<br />

significantly higher levels of aggression toward novel females but more side-byside<br />

affiliation with their familiar female partner (Gobrogge et al., 2009). ELISA<br />

analysis indicated that exposure to a stranger female, compared to a familiar<br />

partner, increased AH-AVP release (Fig. 6.2C), which is further confirmed by<br />

correlation analyses indicating that AH-AVP release was coupled positively with<br />

aggression and negatively with affiliation (Gobrogge et al.,2009). Moreover, intra-<br />

AH administration of AVP at a high (500 ng/side), but not a low (5 ng/side), dose<br />

in sexually naïve males induced aggression toward a novel female, and this effect<br />

was mediated by V1aR as concurrent administration of a 10-fold higher dose of a<br />

V1aR Ant blocked AVP-induced aggression (Fig. 6.2D; Gobrogge et al., 2009).<br />

Further, intra-AH infusions of a V1aR Ant (5 mg/side), in pair bonded males,<br />

blocked aggression and facilitated social affiliation toward novel females<br />

(Fig. 6.2D; Gobrogge et al.,2009). Thus, AH-AVP is both necessary and sufficient<br />

to regulate mating-induced selective aggression in male prairie voles.<br />

Prior research has shown that the social environment has a significant<br />

impact on signaling and structural components of AVP systems in the brain. In<br />

marmoset monkeys, for example, prefrontal V1aR increases during fatherhood<br />

(Kozorovitskiy et al., 2006). In hamsters, several social and drug paradigms have


6. Genetics of Aggression in Voles 131<br />

A B C<br />

10<br />

200<br />

b<br />

F<br />

AVP-ir/Fos-ir cells (density)<br />

Aggression (# attacks/10 min)<br />

8<br />

6<br />

4<br />

2<br />

0<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Control Partner Stranger<br />

female<br />

Naive<br />

a<br />

a<br />

CSF<br />

a<br />

a<br />

AVP AVP+<br />

V1aR Ant<br />

Paired<br />

b<br />

Stranger<br />

male<br />

D E F<br />

b<br />

*<br />

CSF V1aR Ant<br />

Naive<br />

AH<br />

OT<br />

AH<br />

Paired<br />

AVP release (% baseline)<br />

Aggression (# attacks/10 min)<br />

150<br />

100<br />

50<br />

0<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Partner<br />

*<br />

Stranger<br />

female<br />

*<br />

Control AAV-V1aR<br />

Figure 6.2. Vasopressin (AVP) regulation of selective aggression. (A) Pair bonded male prairie voles<br />

that display aggression toward female or male strangers exhibit a significantly higher<br />

density of AVP-ir/Fos-ir double-labeled cells in the AH relative to pair bonded males<br />

re-exposed to their female partner or to males not exposed to any social stimulus<br />

(Control). (B) Photomicrograph of AVP-ir cell bodies and fibers (brown cytoplasmic<br />

staining), Fos-ir (dark nuclear staining), or AVP-ir/Fos-ir double labeled cells (insert) in<br />

the anterior hypothalamus (AH). (C) In vivo brain microdialysis reveals a significant<br />

increase in AH-AVP release in pair bonded male prairie voles displaying aggression<br />

toward a stranger female compared to males reexposed to their female partner. (D) Intra-<br />

AH AVP microinfusion, in sexually naïve males (Naïve), is sufficient to induce aggression<br />

toward a stranger female compared to control males infused with cerebral spinal<br />

fluid (CSF). AH-AVP-induced aggression is blocked with concurrent administration of<br />

AVP with a V1aR Ant. Pair bonded males (Paired) display aggression toward stranger<br />

females, which is blocked by intra-AH infusion of a V1aR Ant. (E) Pair bonded males<br />

(Paired) exhibit a significantly higher density of AVP-V1a receptor (V1aR) binding in<br />

the AH relative to sexually naïve males (Naïve). (F) Sexually naïve males infused with<br />

an adeno-associated virus expressing the V1aR gene (AAV-V1aR) in the AH exhibit<br />

enhanced aggression toward stranger females relative to males infused with the<br />

LacZ-gene (Control). F, fornix; OT, optic tract. Bars indicate means standard error<br />

of the mean. Bars with different Greek letters differ significantly from each other.<br />

*p


132 Gobrogge and Wang<br />

significantly increased the number of AVP mRNA labeled cells in the BNST<br />

(Wang et al., 1994). Because social isolation increases the density of V1aRs in<br />

the AH to regulate offensive aggression in golden hamsters (Albers et al., 2006),<br />

we tested the hypothesis that the density of AH-V1aR changes with pair bonding<br />

experience to engage selective aggression. Pair bonded males showed higher<br />

densities of V1aR binding, site specifically, in the AH (Fig. 6.2E), with no<br />

change in OT receptor binding, demonstrating that pair bonding experience<br />

induces a neural plastic reorganization of V1aRs in a region- and receptorspecific<br />

manner (Gobrogge et al., 2009). To determine whether this increase in<br />

V1aR density in the AH, following pair bonding, was directly related to the<br />

emergence of aggression toward novel females, we used viral vector mediated<br />

gene transfer to artificially elevate V1aR density in the AH. Males that received<br />

intra-AH infusions of the AAV-V1aR displayed higher levels of aggression<br />

toward a novel female compared to control males that received infusions of the<br />

LacZ-gene (Fig. 6.2F; Gobrogge et al., 2009). Similar viral vector mediated<br />

increases in V1aR expression in the VP in voles (Lim et al., 2004; Pitkow<br />

et al., 2001) and LS in mice (Bielsky et al., 2005) enhanced affiliation. In male<br />

rats, intermale aggression correlates with AVP release in the LS while AVP<br />

release in the BNST is inversely related to aggression levels (Veenema et al.,<br />

2010). Together, these data support the notion that region-specific AVP functioning<br />

regulates specific types of social behaviors, and multiple brain regions<br />

serve as a circuit in which AVP coordinates a range of adaptive behaviors<br />

important for reproductive success.<br />

B. Dopamine<br />

Anatomically, central DA is divided into three distinct pathways: nigrostriatal,<br />

incertohypothalamic, and mesocorticolimbic. DA cell bodies projecting from<br />

the substantia nigra synapse in the dorsal striatum and comprise the nigrostriatal<br />

path (Swanson, 1982). Incertohypothalamic paths extend from DA cell bodies of<br />

the A12–14 cell groups and project to the MPOA and PVN (Cheung et al.,<br />

1998). The mesocorticolimbic path represents DA cell bodies originating in the<br />

ventral tegmental area (VTA; Fig. 6.3C) projecting to the mPFC and NAcc<br />

(Fig. 6.3C; Carr and Sesack, 2000; Swanson, 1982). In addition, DA cells in the<br />

AH (Fig. 6.3B) project to forebrain areas including the striatum, LS, NAcc, and<br />

mPFC (Alcaro et al., 2007; Lindvall and Stenevi, 1978; Maeda and Mogenson,<br />

1980).<br />

DA preferentially binds to two families of receptors: D1-like and D2-<br />

like. Both types of DA receptors are found in the mPFC, NAcc, LS, and MeA<br />

(Boyson et al., 1986). D1-like receptors are directly coupled to both stimulatory<br />

(s) Ga and Ga olf proteins (Neve et al., 2004). Stimulation of these G-proteins<br />

leads to activation of adenylate cyclase, cAMP, and protein phosphatase-1


6. Genetics of Aggression in Voles 133<br />

A B C<br />

TH-ir/Fos-ir cells (density)<br />

20<br />

15<br />

10<br />

5<br />

0<br />

a<br />

a<br />

b<br />

Control Partner Stranger<br />

female<br />

b<br />

Stranger<br />

male<br />

D E F<br />

OT<br />

AH<br />

F<br />

TH-NAcc<br />

TH-VTA<br />

DAR binding (optical density)<br />

200<br />

160<br />

120<br />

80<br />

40<br />

0<br />

D1R<br />

*<br />

Naive<br />

Paired<br />

D2R<br />

Naive<br />

NAcc<br />

Paired<br />

Aggression (# attacks /10 min)<br />

40<br />

30<br />

20<br />

10<br />

0<br />

a<br />

b<br />

CSF CSF D2R Ant D1R Ant<br />

partner stranger stranger stranger<br />

b<br />

a<br />

Figure 6.3. Dopamine (DA) regulation of selective aggression. (A) Pair bonded male prairie voles<br />

that display aggression toward female or male strangers exhibit a significantly higher<br />

density of TH-ir/Fos-ir double-labeled cells in the AH relative to pair bonded males reexposed<br />

to their female partner or to males not exposed to any social stimulus (Control).<br />

(B) Photomicrograph of TH-ir cell bodies and fibers (brown cytoplasmic staining), Fos-ir<br />

(dark nuclear staining), or TH-ir/Fos-ir double labeled cells (insert) in the anterior<br />

hypothalamus (AH). (C) Photomicrograph of TH-ir fibers in the nucleus accumbens<br />

(NAcc) and neurons in the ventral tegmental area (VTA). (D, E) Pair bonded males<br />

(Paired) exhibit a significantly higher density of DA D1-like (D1R), but not D2-like<br />

(D2R), receptor binding in the NAcc compared to sexually naïve males (Naïve). (F)<br />

Pair bonded males, infused with CSF in the NAcc, display aggression toward a stranger<br />

female but not toward their female partner. Concurrent infusion of CSF with a DA D1R<br />

antagonist (D1R Ant), but not D2R antagonist (D2R Ant), in the NAcc is sufficient to<br />

attenuate pair bonding-induced aggression toward a stranger female. F, fornix; OT, optic<br />

tract. Bars indicate meansstandard error of the mean. Bars with different Greek letters<br />

differ significantly from each other. *p


134 Gobrogge and Wang<br />

Because mesocorticolimbic DA underlies partner preference formation<br />

(Aragona and Wang, 2009; Aragona et al., 2003; Gingrich et al., 2000; Gobrogge<br />

et al., 2008; Wang et al., 1999), studies focused on examining the potential role<br />

of central DA regulating selective aggression (Gobrogge et al., 2008). Pair<br />

bonded male prairie voles have significantly higher levels of DA D1-type receptors<br />

(D1Rs), but not D2-type receptors (D2Rs), in the NAcc (Fig. 6.3D and E;<br />

Aragona et al., 2006). Males that cohabitated with a female for 24 h, with or<br />

without mating, did not exhibit an increase in D1Rs in the NAcc, supporting the<br />

idea that upregulation of NAcc D1Rs, after pair bonding, may directly regulate<br />

selective aggression. To test this notion, pair bonded male prairie voles were<br />

injected with a D1R antagonist (D1R Ant) into the NAcc. NAcc-D1R, not D2R<br />

(D2R Ant), antagonism was sufficient to block selective aggression in pair<br />

bonded male prairie voles (Fig. 6.3F; Aragona et al., 2006). In other work,<br />

both brief and extended cohabitation with unfamiliar conspecifics in female<br />

prairie voles significantly increased the number of DA-ergic cells in the BNST<br />

and MeA (Cavanaugh and Lonstein, 2010) and blocking D2Rs, during development,<br />

decreased aggression-related behavior including infanticide in adult<br />

female, but not male, prairie voles (Hostetler et al., 2010). Further, pair bonded<br />

male prairie voles—displaying aggression toward either male or female intruders,<br />

had a significantly higher density of cells in the AH that were double-labeled for<br />

tyrosine hydroxylase-ir (TH—rate-limiting enzyme in DA biosynthesis) and Fosir<br />

than males not exposed to a social stimulus or males that were re-exposed to<br />

their familiar female partner (Fig. 6.3A and B), implicating AH-DA involvement<br />

in selective aggression (Gobrogge et al., 2007).<br />

C. Steroid hormones<br />

Physical aggression is significantly more common in males than females and<br />

these behavioral sex differences have been observed across many species<br />

(Gatewood et al., 2006). Research describing biological contributions underlying<br />

these sex differences has focused primarily on steroid hormones (Gatewood et al.,<br />

2006). Several studies have examined the role of androgens in the development<br />

of aggressive behavior, both organizationally (e.g., treatment with prenatal<br />

testosterone) and activationally (e.g., treatment with postnatal testosterone).<br />

Previous research has found that prenatal androgen exposure increases the<br />

behavioral expression of adult aggression (Michard-Vanhee, 1988; Vale et al.,<br />

1972). Although organizational and activational influences of androgen on<br />

aggression have been noted, some inconsistent results have been reported. For<br />

example, castration in male rats (Koolhaas et al., 1990) and male prairie voles<br />

(Demas et al., 1999) has no affect on aggression. Thus, circulating testosterone,<br />

alone, cannot solely contribute to the expression of aggressive behavior in all<br />

rodent species. However, these findings do not rule out the possible effects of


6. Genetics of Aggression in Voles 135<br />

testosterone having organizational influences on other neurochemical systems in<br />

the brain—which, together, may regulate aggression in adulthood. Thus,<br />

neurochemical–steroid hormone interactions—underlying aggression—have<br />

also been examined. For example, AVP administered directly into the MeA<br />

facilitates territorial aggression in male rats and is sufficient to block the effects of<br />

castration on reducing aggression (Koolhaas et al., 1990). Further, castration<br />

(Bermond et al., 1982), but not ovariectomy (Kruk et al., 1984), decreases the<br />

excitability of neurons in the AH—blocking electrically induced aggression,<br />

which in castrates can be reversed by testosterone treatment (Bermond et al.,<br />

1982). Therefore, circulating testosterone may be acting as a potent neuromodulator—interacting<br />

with neurochemicals, like AVP, to regulate aggression.<br />

Additional evidence demonstrating steroid hormone–neurotransmitter<br />

interactions exists in research investigating central DA. For example, 75% of<br />

TH-ir expressing cells in the hamster posterior MeA contain androgen receptors,<br />

are DA-ergic (i.e., they do not co-label with DA beta hydroxylase), and are<br />

highly influenced by gonadal hormones compared to TH-ir cells found in the<br />

anterior MeA (Asmus and Newman, 1993; Asmus et al., 1992). Interestingly,<br />

this same group of TH-ir cells is found in the posterior MeA and BNST in male<br />

prairie voles, which appears to be influenced by testosterone (Northcutt et al.,<br />

2007) and activated after mating and social experience (Northcutt and Lonstein,<br />

2009). Together, these data suggest interactions between steroid hormones and<br />

central DA, in areas such as the MeA, in processing chemosensory cues related to<br />

social communication.<br />

D. Classical neurotransmitters<br />

In addition to the effects of neurotransmitters and hormones on aggression,<br />

neuromodulators such as GABA and glutamate have also been shown to be<br />

involved in the display of agonistic behavior. For example, microinjection of a<br />

GABA antagonist (Adams et al., 1993; Roeling et al., 1993) with concurrent<br />

treatment of a glutamate agonist (Haller et al., 1998) in the AH facilitates attack<br />

behavior in rodents—with higher doses having greater behavioral effects. Further,<br />

reverse microdialysis infusion with a glutamate agonist and a GABA-A<br />

antagonist into the AH of rats, recently having experienced an agonistic<br />

encounter, also facilitates aggression (Haller et al., 1998). Finally, it is worth<br />

mentioning that neuromodulators in the VTA, which provides the major<br />

source of DA projections to the NAcc (Fig. 6.3C) and mPFC, are also involved<br />

in pair bonding behavior. Glutamate and GABA receptor blockade in<br />

the VTA, which alters DA activity in the NAcc, induces partner preference<br />

formation in the absence of mating in male prairie voles (Curtis and Wang,<br />

2005b).


136 Gobrogge and Wang<br />

VI. MOLECULAR GENETICS OF SELECTIVE AGGRESSION<br />

Monogamous male prairie voles carry several repetitive microsatellite DNA<br />

sequences in the promoter region of the V1aR gene that are not found in<br />

promiscuous male voles (Hammock and Young, 2002, 2004; Young, 1999).<br />

These genetic differences directly contribute to species differences in social<br />

organization (Landgraf et al., 2003; Lim et al., 2004; Pitkow et al., 2001). Further,<br />

mice carrying a transgene coding for the prairie vole V1aR exhibit central V1aR<br />

patterns similar to prairie voles and, when injected with AVP, display enhanced<br />

social affiliation (Young et al., 1999). Male voles injected in the VP, with a virus<br />

expressing V1aR, display enhanced partner preference in the absence of mating<br />

(Lim et al., 2004; Pitkow et al., 2001). Interestingly, individual variability in the<br />

genetic sequences coding V1aRs has revealed remarkable within species differences<br />

in the strength of monogamous pair bonds in prairie voles (Hammock and<br />

Young, 2005; Ophir et al., 2008).<br />

Because prairie voles carry varying lengths of DNA to code V1aRs<br />

(Hammock and Young, 2005), this genomic predisposition enables their brain<br />

to dynamically express V1aRs after sociosexual experience. This genetic loading<br />

distinguishes prairie voles from traditional laboratory rodents that lack this<br />

genetic makeup. Future work would benefit from comparing aggression levels<br />

between male prairie voles carrying long versus short versions of the promoter<br />

region encoding the V1aR gene. In humans, polymorphisms in the promoter<br />

region encoding V1aR are associated with differences in sociosexual bonding<br />

behaviors (Prichard et al., 2007; Walum et al., 2008) including altruism (Israel<br />

et al., 2008) and deficits in social communication observed in individuals with<br />

autistic spectrum disorders (Israel et al., 2008; Kim et al., 2002; Meyer-<br />

Lindenberg et al., 2008). To date, no study has examined potential associations<br />

between the V1aR gene and patterns of aggression in humans. Thus, it would be<br />

interesting to examine polymorphisms in the V1aR gene in patients with a<br />

lifetime history of pathological violence, as the V1aR system may harbor susceptibility<br />

genes underlying extreme forms of aggression, increasing the prevalence<br />

of homicide and suicide in human populations.<br />

VII. DRUG-INDUCED AGGRESSION<br />

The prairie vole has been established as an animal model for depression related to<br />

social separation (Bosch et al., 2009; Grippo et al., 2007). Further, chronic metal<br />

ingestion—a potential model for autism—produces social avoidance in male, but<br />

not female, prairie voles exposed to unfamiliar same-sex conspecific strangers<br />

(Curtis et al., 2010), suggesting a developmental mechanism underlying the social


6. Genetics of Aggression in Voles 137<br />

deficits associated with autistic spectrum disorders in humans. Recently, prairie<br />

voles have also been utilized to examine the effects of drugs of abuse on pair<br />

bonding behavior (Gobrogge et al., 2009; Liu et al., 2010; Young et al., 2011b).<br />

Drug addiction is a significant problem for many humans because drug<br />

abuse has such a powerful control over social behavior essential for survival<br />

(Kelley and Berridge, 2002; Nesse and Berridge, 1997; Panksepp et al., 2002). In<br />

humans, substance abuse has been associated with weapon-related violence and<br />

homicide (Hagelstam and Hakkanen, 2006; Madan et al., 2001; Spunt et al.,<br />

1998), intimate partner aggression, including partner-directed physical and<br />

psychological aggression (Chermack et al., 2008; O’Farrell and Fals-Stewart,<br />

2000), sexual (El-Bassel et al., 2001), and child abuse (Haapasalo and<br />

Hamalainen, 1996; Mokuau, 2002; Walsh et al., 2003). Collectively, drugrelated<br />

violence leads to family system dysfunction and incarceration (Krug<br />

et al., 2002), creating significant societal concerns. While aggression research<br />

in humans has provided valuable information regarding relationships between<br />

drug abuse and violence, animal models have been used to examine neural<br />

mechanisms underlying drug-induced aggression.<br />

Drug use can override neurobiological programs to activate maladaptive<br />

forms of agonistic behavior, engaging inappropriate types of physical aggression<br />

(Swartz et al., 1998) such as domestic violence (Moore et al., 2008) and intimate<br />

partner homicide (Farooque et al., 2005). As a result, chronic drug abuse can<br />

cause permanent neural reorganization (Nestler and Aghajanian, 1997; White<br />

and Kalivas, 1998), impairing the adaptive—social brain (Panksepp et al., 2002),<br />

leading to the display of maladaptive social behavior (Wise, 2002). Multiple<br />

studies have demonstrated that aggression may be altered shortly after drug<br />

exposure and that the directionality of these effects depends on drug, dose, and<br />

individual differences between subjects.<br />

Repeated exposure to several drugs of abuse, during adolescence or<br />

adulthood, persistently enhances agonistic behaviors, specifically those associated<br />

with offensive aggression. For example, Syrian hamsters treated during<br />

adolescence with cocaine (DeLeon et al., 2002a; Harrison et al., 2000a; Jackson<br />

et al., 2005; Knyshevski et al., 2005a,b; Melloni et al., 2001) or anabolic-androgenic<br />

steroids (AASs) (DeLeon et al., 2002b; Harrison et al., 2000b; Melloni and<br />

Ferris, 1996; Melloni et al., 1997) display enhanced offensive aggression in<br />

adulthood. Interestingly, these drug experiences reorganize AVP (Grimes et al.,<br />

2007; Harrison et al., 2000b; Jackson et al., 2005), DA (Ricci et al., 2009;<br />

Schwartzer et al., 2009), and GABA (Schwartzer et al., 2009) signaling in<br />

the AH.<br />

For example, when compared with nonaggressive sesame oil-treated<br />

control males, aggressive AAS-treated males exhibit significant neuroplastic<br />

changes in the AH including increased AVP-ir fiber density and AVP content<br />

(Harrison et al., 2000b), an increase in TH-ir cell and fiber density (Ricci et al.,


138 Gobrogge and Wang<br />

2009), enhanced DA-D2R expression (Schwartzer et al., 2009), a higher number<br />

of GAD 67 -ir cells (Schwartzer et al., 2009), and decreased GABA A receptor<br />

expression (Schwartzer et al., 2009). Further, pharmacological blockade of D2<br />

(Schwartzer and Melloni, 2010a,b), but not D5 (Schwartzer and Melloni,<br />

2010b), DA receptors in the AH abolishes these effects. Together, results from<br />

this work suggests that AVP and DA signaling facilitates aggression by GABA<br />

inhibition in the AH of AAS-treated male Syrian hamsters.<br />

As noted above, previous work has shown that exposure to drugs of<br />

abuse, such as cocaine, enhances male–male aggression by reorganizing the AH-<br />

AVP system in hamsters (Jackson et al., 2005). Therefore, we tested the hypothesis<br />

that amphetamine (AMPH), another commonly abused psychostimulant,<br />

would act in a similar fashion in affecting male-to-female aggression in prairie<br />

voles. Because our recent data revealed that repeated AMPH treatment—(1 mg/<br />

kg) for 3 consecutive days—induces a conditioned place preference (Aragona<br />

et al., 2007) and blocks mating-induced partner preference (Fig. 6.4A; Liu et al.,<br />

2010), this treatment regimen was used. To examine the selectivity of AMPHinduced<br />

aggression, males treated with saline or AMPH were tested for aggression<br />

toward an unfamiliar female or a familiar female (that cohabitated with a<br />

male across a wire mesh screen for 24 h without mating). Compared with salinetreated<br />

controls, AMPH-treated males displayed significantly higher levels of<br />

aggression toward either familiar or unfamiliar females (Fig. 6.4B), indicating<br />

that AMPH exposure induces generalized aggression, rather than being selective<br />

to novel females. This AMPH treatment also induced an increase in the density<br />

of AVP-V1aR binding in the AH, but not MPOA, relative to saline control<br />

males (Fig. 6.4C). Further, intra-AH infusions of CSF containing the V1aR Ant,<br />

but not CSF alone, diminished AMPH-induced aggression toward novel females<br />

(Fig. 6.4D). These data suggest that repeated exposure to AMPH can induce<br />

female-directed aggression and that this behavior is mediated by AH-AVP.<br />

Interestingly, these behavioral effects coincide with upregulation of D1Rs in<br />

the NAcc (Liu et al., 2010) and V1aRs in the AH (Gobrogge et al., 2009)—<br />

which both facilitate aggression toward novel females (Aragona et al., 2006;<br />

Gobrogge et al., 2009); indicating that drugs of abuse can hijack neuroplasticity<br />

evolved to maintain monogamous pair bonds.<br />

VIII. CONCLUSIONS AND FUTURE DIRECTIONS<br />

Prairie voles have provided an excellent model system to study the neurobiology<br />

of ethologically meaningful aggression associated with monogamous pair bonds.<br />

Aggression can be easily manipulated under laboratory conditions and reliably<br />

expressed following mating and social cohabitation. Several brain areas: MeA,<br />

AH, and NAcc, work in a neural circuit to regulate selective aggression via AVP


6. Genetics of Aggression in Voles 139<br />

A<br />

B<br />

Side-by-Side contact (min/3 h)<br />

100<br />

80<br />

60<br />

40<br />

20<br />

** **<br />

Partner<br />

Stranger<br />

Frequency of aggression<br />

40<br />

30<br />

20<br />

10<br />

Familiar<br />

Unfamiliar<br />

a<br />

b<br />

0<br />

Intact<br />

0.0 1.0 5.0<br />

AMPH (mg/kg)<br />

0<br />

Saline<br />

AMPH<br />

C<br />

V1aR binding (optical density)<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

Saline<br />

AMPH<br />

*<br />

D<br />

Frequency of aggression<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

*<br />

0<br />

MPOA<br />

AH<br />

0<br />

CSF<br />

V1aR Ant<br />

Figure 6.4. Drug experience impairs pair bonding behavior. (A) Pair bonded (Intact) and saline<br />

treated (0.0) male prairie voles, receiving 3-day—once daily; repeated injections, spend<br />

significantly more time in physical side-by-side contact with their familiar female<br />

partner than with an unfamiliar stranger female. Pair bonded males injected (i.p.,<br />

intraperitoneal) with 1.0 or 5.0 mg/kg amphetamine (AMPH) spent equal amounts of<br />

time in physical side-by-side contact with their female partner as with a stranger female.<br />

(B, C) Repeated AMPH exposure, in sexually naïve males, increases aggression toward<br />

both familiar and unfamiliar females and enhances the density of vasopressin (AVP)<br />

receptor (V1aR) expression in the anterior hypothalamus (AH) but not in the medial<br />

preoptic area (MPOA). (D) Site-specific microinfusion of an AVP-V1aR antagonist<br />

(V1aR Ant) into the AH, of males receiving 3-day repeated AMPH exposure (i.p.),<br />

significantly decreases AMPH-induced aggression toward novel females relative to<br />

males receiving intra-AH infusions of cerebral spinal fluid (CSF). Bars indicate meansstandard<br />

error of the mean. Bars with different Greek letters differ significantly from<br />

each other. *p


140 Gobrogge and Wang<br />

Although male-to-male aggression has been studied in a variety of<br />

mammals, we know surprisingly little about male-to-female aggression and its<br />

underlying neuromechanisms. Interestingly, pair bonded male prairie voles naturally<br />

display aggression toward conspecific females but not toward their female<br />

partner and, therefore, selective aggression allows for investigation of the neurobiology<br />

of male-to-female aggression. Data have demonstrated that this form of<br />

selective aggression is mediated by elevated AVP release and increased V1aR<br />

expression in the AH—priming male prairie voles to respond aggressively to<br />

novel females. In addition, data have also shown that the same AH-AVP system<br />

mediates generalized, female-directed aggression induced by AMPH. Together<br />

with previous research from other animals (Ferris et al., 1989, 1997; Grimes et al.,<br />

2007; Harrison et al., 2000b; Jackson et al., 2005; Veenema et al., 2006), these<br />

data demonstrate a unique point of convergence in the mammalian brain (Choi<br />

et al., 2005; Motta et al., 2009). The AH-AVP system is highly conserved and<br />

functions to control different forms of aggression to maintain a wide range of<br />

resources important for reproductive success. These highly evolved neuropeptide<br />

systems appear to be extremely vulnerable to drugs of abuse, as our data show that<br />

hypothalamic AVP controls both naturally occurring as well as drug-facilitated<br />

female-directed aggression, suggesting that psychostimulant drugs, like AMPH,<br />

are capable of switching adaptive (functional) forms of aggression (e.g., mate<br />

guarding) to aberrant (dysfunctional) forms of violent behavior (e.g., partnerdirected<br />

aggression). Together, these data demonstrate the utility of the prairie<br />

vole model for evaluation of the effects of drug abuse on neural systems<br />

controlling adaptive forms of aggression—such as mate guarding.<br />

Finally, because other neurochemicals, such as DA (Aragona et al.,<br />

2006) and serotonin (Villalba et al., 1997), also regulate selective aggression in<br />

prairie voles, offensive aggression related to drug experience (Tidey and Miczek,<br />

1992) and AVP/5-HT interactions mediate aggression in other rodents (Ferris<br />

et al., 1997; Veenema et al., 2006), future studies should examine potential<br />

neurochemical interactions in the regulation of selective aggression. By understanding<br />

the basic neuroendocrinology of pair bonding in prairie voles, we may<br />

eventually be able to better clarify the neural chemistry of mental health deficits<br />

associated with aberrations in social behavior in patients suffering from drug<br />

addiction or pathological violence.<br />

Acknowledgments<br />

The authors would like to thank Benjamin William Tyson for critically reading this manuscript.<br />

The work reviewed in this chapter was supported by National Institutes of Health grants MHF31-<br />

79600 to K. L. G., MHR01-58616, MHR01-89852, DAR01-19627, and DAK02-23048 to Z. X. W.,<br />

and NIH Program Training Grant T32 NS-07437.


6. Genetics of Aggression in Voles 141<br />

References<br />

Adams, D. B., Boudreau, W., Cowan, C. W., Kokonowski, C., Oberteuffer, K., and Yohay, K. (1993).<br />

Offense produced by chemical stimulation of the anterior hypothalamus of the rat. Physiol. Behav.<br />

53, 1127–1132.<br />

Albers, E. H., Dean, A., Karom, M. C., Smith, D., and Huhman, K. L. (2006). Role of V1a<br />

vasopressin receptors in the control of aggression in Syrian hamsters. Brain Res. 1073–1074,<br />

425–430.<br />

Albert, D. J., and Walsh, M. L. (1984). Neural systems and the inhibitory modulation of agonistic<br />

behavior: A comparison of mammalian species. Neurosci. Biobehav. Rev. 8, 5–24.<br />

Alcaro, A., Huber, R., and Panksepp, J. (2007). Behavioral functions of the mesolimbic dopaminergic<br />

system: An affective neuroethological perspective. Brain Res. Rev. 56, 283–321.<br />

Aragona, B. J., and Wang, Z. (2009). Dopamine regulation of social choice in a monogamous rodent<br />

species. Front. Behav. Neurosci. 3, 15.<br />

Aragona, B. J., Liu, Y., Curtis, J. T., Stephan, F. K., and Wang, Z. (2003). A critical role for nucleus<br />

accumbens dopamine in partner-preference formation in male prairie voles. J. Neurosci. 23,<br />

3483–3490.<br />

Aragona, B. J., Liu, Y., Yu, Y. J., Curtis, J. T., Detwiler, J. M., Insel, T. R., and Wang, Z. (2006).<br />

Nucleus accumbens dopamine differentially mediates the formation and maintenance of monogamous<br />

pair bonds. Nat. Neurosci. 9, 133–139.<br />

Aragona, B. J., Detwiler, J. M., and Wang, Z. (2007). Amphetamine reward in the monogamous<br />

prairie vole. Neurosci. Lett. 418, 190–194.<br />

Asmus, S. E., and Newman, S. W. (1993). Tyrosine hydroxylase neurons in the male hamster<br />

chemosensory pathway contain androgen receptors and are influenced by gonadal hormones.<br />

J. Comp. Neurol. 331, 445–457.<br />

Asmus, S. E., Kincaid, A. E., and Newman, S. W. (1992). A species-specific population of tyrosine<br />

hydroxylase-immunoreactive neurons in the medial amygdaloid nucleus of the Syrian hamster.<br />

Brain Res. 575, 199–207.<br />

Backstrom, T., and Winberg, S. (2009). Arginine-vasotocin influence on aggressive behavior and<br />

dominance in rainbow trout. Physiol. Behav. 96, 470–475.<br />

Bales, K. L., and Carter, C. S. (2003). Sex differences and developmental effects of oxytocin on<br />

aggression and social behavior in prairie voles (Microtus ochrogaster). Horm. Behav. 44, 178–184.<br />

Bamshad, M., Novak, M. A., and De Vries, G. J. (1993a). Sex and species differences in the<br />

vasopressin innervation of sexually naive and parental prairie voles, Microtus ochrogaster and<br />

meadow voles, Microtus pennsylvanicus. J. Neuroendocrinol. 5, 247–255.<br />

Bamshad, M., Novak, M. A., and deVries, A. C. (1993b). Sex and species differences in the<br />

vasopressin innervation of sexually naive and parental prairie voles (Microtus ochrogaster) and<br />

meadow voles (Microtus pennsylvanicus). J. Neuroendocrinol. 5, 247–255.<br />

Bermond, B., Mos, J., Meelis, W., van der Poel, A. M., and Kruk, M. R. (1982). Aggression induced<br />

by stimulation of the hypothalamus: Effects of androgens. Pharmacol. Biochem. Behav. 16, 41–45.<br />

Bielsky, I. F., Hu, S. B., Ren, X., Terwilliger, E. F., and Young, L. J. (2005). The V1a vasopressin<br />

receptor is necessary and sufficient for normal social recognition: A gene replacement study.<br />

Neuron 47, 503–513.<br />

Bosch, O. J., Nair, H. P., Ahern, T. H., Neumann, I. D., and Young, L. J. (2009). The CRF system<br />

mediates increased passive stress-coping behavior following the loss of a bonded partner in a<br />

monogamous rodent. Neuropsychopharmacology 34, 1406–1415.<br />

Bowler, C. M., Cushing, B. S., and Carter, C. S. (2002). Social factors regulate female-female<br />

aggression and affiliation in prairie voles. Physiol. Behav. 76, 559–566.<br />

Boyson, S. J., McGonigle, P., and Molinoff, P. B. (1986). Quantitative autoradiographic localization<br />

of the D1 and D2 subtypes of dopamine receptors in rat brain. J. Neurosci. 6, 3177–3188.


142 Gobrogge and Wang<br />

Caldwell, H. K., and Albers, H. E. (2004). Effect of photoperiod on vasopressin-induced aggression in<br />

Syrian hamsters. Horm. Behav. 46, 444–449.<br />

Caldwell, H. K., Lee, H. J., Macbeth, A. H., and Young, W. S., 3rd (2008). Vasopressin: Behavioral<br />

roles of an “original” neuropeptide. Prog. Neurobiol. 84, 1–24.<br />

Carr, D. B., and Sesack, S. R. (2000). Projections from the rat prefrontal cortex to the ventral<br />

tegmental area: Target specificity in the synaptic associations with mesoaccumbens and mesocortical<br />

neurons. J. Neurosci. 20, 3864–3873.<br />

Carter, C. S., DeVries, A. C., and Getz, L. L. (1995). Physiological substrates of mammalian<br />

monogamy: The prairie vole model. Neurosci. Biobehav. Rev. 19, 303–314.<br />

Cavanaugh, B. L., and Lonstein, J. S. (2010). Social novelty increases tyrosine hydroxylase immunoreactivity<br />

in the extended olfactory amygdala of female prairie voles. Physiol. Behav. 100,<br />

381–386.<br />

Chermack, S. T., Murray, R. L., Walton, M. A., Booth, B. A., Wryobeck, J., and Blow, F. C. (2008).<br />

Partner aggression among men and women in substance use disorder treatment: Correlates of<br />

psychological and physical aggression and injury. Drug Alcohol Depend. 98, 35–44.<br />

Cheung, S., Ballew, J. R., Moore, K. E., and Lookingland, K. J. (1998). Contribution of dopamine<br />

neurons in the medial zona incerta to the innervation of the central nucleus of the amygdala,<br />

horizontal diagonal band of Broca and hypothalamic paraventricular nucleus. Brain Res. 808,<br />

174–181.<br />

Choi, G. B., Dong, H. W., Murphy, A. J., Valenzuela, D. M., Yancopoulos, G. D., Swanson, L. W.,<br />

and Anderson, D. J. (2005). Lhx6 delineates a pathway mediating innate reproductive behaviors<br />

from the amygdala to the hypothalamus. Neuron 46, 647–660.<br />

Coccaro, E. F., Kavoussi, R. J., Hauger, R. L., Cooper, T. B., and Ferris, C. F. (1998). Cerebrospinal<br />

fluid vasopressin levels: Correlates with aggression and serotonin function in personality-disordered<br />

subjects. Arch. Gen. Psychiatry 55, 708–714.<br />

Cooper, M. A., Karom, M., Huhman, K. L., and Albers, H. E. (2005). Repeated agonistic encounters<br />

in hamsters modulate AVP V1a receptor binding. Horm. Behav. 48, 545–551.<br />

Curtis, J. T., and Wang, Z. (2003). Forebrain c-fos expression under conditions conducive to pair<br />

bonding in female prairie voles (Microtus ochrogaster). Physiol. Behav. 80, 95–101.<br />

Curtis, J. T., and Wang, Z. (2005a). Glucocorticoid receptor involvement in pair bonding in female<br />

prairie voles: The effects of acute blockade and interactions with central dopamine reward<br />

systems. Neuroscience 134, 369–376.<br />

Curtis, J. T., and Wang, Z. (2005b). Ventral tegmental area involvement in pair bonding in male<br />

prairie voles. Physiol. Behav. 86, 338–346.<br />

Curtis, J. T., Hood, A. N., Chen, Y., Cobb, G. P., and Wallace, D. R. (2010). Chronic metals<br />

ingestion by prairie voles produces sex-specific deficits in social behavior: An animal model of<br />

autism. Behav. Brain Res. 213, 42–49.<br />

Cushing, B. S., Mogekwu, N., Le, W. W., Hoffman, G. E., and Carter, C. S. (2003a). Cohabitation<br />

induced Fos immunoreactivity in the monogamous prairie vole. Brain Res. 965, 203–211.<br />

Cushing, B. S., Yamamoto, Y., Hoffman, G. E., and Carter, C. S. (2003b). Central expression of c-Fos<br />

in neonatal male and female prairie voles in response to treatment with oxytocin. Brain Res. Dev.<br />

Brain Res. 143, 129–136.<br />

DeLeon, K. R., Grimes, J. M., Connor, D. F., and Melloni, R. H., Jr. (2002a). Adolescent cocaine<br />

exposure and offensive aggression: Involvement of serotonin neural signaling and innervation in<br />

male Syrian hamsters. Behav. Brain Res. 133, 211–220.<br />

DeLeon, K. R., Grimes, J. M., and Melloni, R. H., Jr. (2002b). Repeated anabolic-androgenic steroid<br />

treatment during adolescence increases vasopressin V(1A) receptor binding in Syrian hamsters:<br />

Correlation with offensive aggression. Horm. Behav. 42, 182–191.<br />

Delville, Y., De Vries, G. J., and Ferris, C. F. (2000). Neural connections of the anterior hypothalamus<br />

and agonistic behavior in golden hamsters. Brain Behav. Evol. 55, 53–76.


6. Genetics of Aggression in Voles 143<br />

Demas, G. E., Moffatt, C. A., Drazen, D. L., and Nelson, R. J. (1999). Castration does not inhibit<br />

aggressive behavior in adult male prairie voles (Microtus ochrogaster). Physiol. Behav. 66, 59–62.<br />

DeVries, A. C., DeVries, M. B., Taymans, S., and Carter, C. S. (1995). Modulation of pair bonding in<br />

female prairie voles (Microtus ochrogaster) by corticosterone. Proc. Natl. Acad. Sci. USA 92,<br />

7744–7748.<br />

Dewsbury, D. A. (1987). The comparative psychology of monogamy. Nebr. Symp. Motiv. 35, 1–50.<br />

Deyo, R. A., Straube, K. T., and Disterhoft, J. F. (1989). Nimodipine facilitates associative learning<br />

in aging rabbits. Science 243, 809–811.<br />

Donaldson, Z. R., and Young, L. J. (2008). Oxytocin, vasopressin, and the neurogenetics of sociality.<br />

Science 322, 900–904.<br />

El-Bassel, N., Witte, S. S., Wada, T., Gilbert, L., and Wallace, J. (2001). Correlates of partner<br />

violence among female street-based sex workers: Substance abuse, history of childhood abuse, and<br />

HIV risks. AIDS Patient Care STDS 15, 41–51.<br />

Farooque, R. S., Stout, R. G., and Ernst, F. A. (2005). Heterosexual intimate partner homicide:<br />

Review of ten years of clinical experience. J. Forensic Sci. 50, 648–651.<br />

Ferris, C. F., and Potegal, M. (1988). Vasopressin receptor blockade in the anterior hypothalamus<br />

suppresses aggression in hamsters. Physiol. Behav. 44, 235–239.<br />

Ferris, C. F., Albers, H. E., Wesolowski, S. M., Goldman, B. D., and Luman, S. E. (1984). Vasopressin<br />

injected into the hypothalamus triggers a stereotypic behavior in golden hamsters. Science 224,<br />

521–523.<br />

Ferris, C. F., Axelson, J. F., Martin, A. M., and Roberge, L. F. (1989). Vasopressin immunoreactivity<br />

in the anterior hypothalamus is altered during the establishment of dominant/subordinate relationships<br />

between hamsters. Neuroscience 29, 675–683.<br />

Ferris, C. F., Melloni, R. H., Jr., Koppel, G., Perry, K. W., Fuller, R. W., and Delville, Y. (1997).<br />

Vasopressin/serotonin interactions in the anterior hypothalamus control aggressive behavior in<br />

golden hamsters. J. Neurosci. 17, 4331–4340.<br />

Ferris, C. F., Lu, S. F., Messenger, T., Guillon, C. D., Heindel, N., Miller, M., Koppel, G., Robert<br />

Bruns, F., and Simon, N. G. (2006). Orally active vasopressin V1a receptor antagonist, SRX251,<br />

selectively blocks aggressive behavior. Pharmacol. Biochem. Behav. 83, 169–174.<br />

Gatewood, J. D., Wills, A., Shetty, S., Xu, J., Arnold, A. P., Burgoyne, P. S., and Rissman, E. F.<br />

(2006). Sex chromosome complement and gonadal sex influence aggressive and parental behaviors<br />

in mice. J. Neurosci. 26, 2335–2342.<br />

Gingrich, B. S., Huot, R. L., Wang, Z., and Insel, T. R. (1997). Differential Fos expression following<br />

microinjection of oxytocin or vasopressin in the prairie vole brain. Ann. N. Y. Acad. Sci. 807,<br />

504–505.<br />

Gingrich, B., Liu, Y., Cascio, C., Wang, Z., and Insel, T. R. (2000). Dopamine D2 receptors in the<br />

nucleus accumbens are important for social attachment in female prairie voles (Microtus ochrogaster).<br />

Behav. Neurosci. 114, 173–183.<br />

Gobrogge, K. L., and Wang, Z. (2009). Neural Circuitry Underlying Pair Bonding-induced Aggression<br />

in Monogamous Male Prairie Voles. Society for Neuroscience Abstracts #377.4 Chicago, IL.<br />

Gobrogge, K. L., Liu, Y., Jia, X., and Wang, Z. (2007). Anterior hypothalamic neural activation and<br />

neurochemical associations with aggression in pair-bonded male prairie voles. J. Comp. Neurol.<br />

502, 1109–1122.<br />

Gobrogge, K. L., Liu, Y., and Wang, Z. (2008). Dopamine regulation of pair bonding in monogamous<br />

prairie voles. In “The Neurobiology of the Parental Brain” (R. Bridges, ed.), pp. 345–358. Elsevier,<br />

San Diego, CA.<br />

Gobrogge, K. L., Liu, Y., Young, L. J., and Wang, Z. (2009). Anterior hypothalamic vasopressin<br />

regulates pair-bonding and drug-induced aggression in a monogamous rodent. Proc. Natl. Acad.<br />

Sci. USA 106, 19144–19149.


144 Gobrogge and Wang<br />

Goodson, J. L. (2008). Nonapeptides and the evolutionary patterning of sociality. Prog. Brain Res.<br />

170, 3–15.<br />

Grimes, J. M., Ricci, L. A., and Melloni, R. H., Jr. (2007). Alterations in anterior hypothalamic<br />

vasopressin, but not serotonin, correlate with the temporal onset of aggressive behavior during<br />

adolescent anabolic-androgenic steroid exposure in hamsters (Mesocricetus auratus). Behav.<br />

Neurosci. 121, 941–948.<br />

Grippo, A. J., Cushing, B. S., and Carter, C. S. (2007). Depression-like behavior and stressor-induced<br />

neuroendocrine activation in female prairie voles exposed to chronic social isolation. Psychosom.<br />

Med. 69, 149–157.<br />

Haapasalo, J., and Hamalainen, T. (1996). Childhood family problems and current psychiatric<br />

problems among young violent and property offenders. J. Am. Acad. Child Adolesc. Psychiatry<br />

35, 1394–1401.<br />

Hagelstam, C., and Hakkanen, H. (2006). Adolescent homicides in Finland: Offence and offender<br />

characteristics. Forensic Sci. Int. 164, 110–115.<br />

Hairston, J. E., Ball, G. F., and Nelson, R. J. (2003). Photoperiodic and temporal influences on<br />

chemosensory induction of brain fos expression in female prairie voles. J. Neuroendocrinol. 15,<br />

161–172.<br />

Haller, J., Abraham, I., Zelena, D., Juhasz, G., Makara, G. B., and Kruk, M. R. (1998). Aggressive<br />

experience affects the sensitivity of neurons towards pharmacological treatment in the hypothalamic<br />

attack area. Behav. Pharmacol. 9, 469–475.<br />

Hammock, E. A., and Young, L. J. (2002). Variation in the vasopressin V1a receptor promoter and<br />

expression: Implications for inter- and intraspecific variation in social behaviour. Eur. J. Neurosci.<br />

16, 399–402.<br />

Hammock, E. A., and Young, L. J. (2004). Functional microsatellite polymorphism associated with<br />

divergent social structure in vole species. Mol. Biol. Evol. 21, 1057–1063.<br />

Hammock, E. A., and Young, L. J. (2005). Microsatellite instability generates diversity in brain and<br />

sociobehavioral traits. Science 308, 1630–1634.<br />

Harrison, R. J., Connor, D. F., Nowak, C., and Melloni, R. H., Jr. (2000a). Chronic low-dose cocaine<br />

treatment during adolescence facilitates aggression in hamsters. Physiol. Behav. 69, 555–562.<br />

Harrison, R. J., Connor, D. F., Nowak, C., Nash, K., and Melloni, R. H., Jr. (2000b). Chronic<br />

anabolic-androgenic steroid treatment during adolescence increases anterior hypothalamic vasopressin<br />

and aggression in intact hamsters. Psychoneuroendocrinology 25, 317–338.<br />

Hostetler, C. M., Harkey, S. L., and Bales, K. L. (2010). D2 antagonist during development decreases<br />

anxiety and infanticidal behavior in adult female prairie voles (Microtus ochrogaster). Behav.<br />

Brain Res. 210, 127–130.<br />

Insel, T. R. (2010). The challenge of translation in social neuroscience: A review of oxytocin,<br />

vasopressin, and affiliative behavior. Neuron 65, 768–779.<br />

Insel, T. R., and Hulihan, T. J. (1995). A gender-specific mechanism for pair bonding: Oxytocin and<br />

partner preference formation in monogamous voles. Behav. Neurosci. 109, 782–789.<br />

Insel, T. R., and Shapiro, L. E. (1992). Oxytocin receptor distribution reflects social organization in<br />

monogamous and polygamous voles. Proc. Natl. Acad. Sci. USA 89, 5981–5985.<br />

Insel, T. R., Wang, Z., and Ferris, C. F. (1994). Patterns of brain vasopressin receptor distribution<br />

associated with social organization in microtine rodents. J. Neurosci. 14, 5381–5392.<br />

Insel, T. R., Preston, S., and Winslow, J. T. (1995). Mating in the monogamous male: Behavioral<br />

consequences. Physiol. Behav. 57, 615–627.<br />

Insel, T. R., Winslow, J. T., Wang, Z., and Young, L. J. (1998). Oxytocin, vasopressin, and the<br />

neuroendocrine basis of pair bond formation. Adv. Exp. Med. Biol. 449, 215–224.


6. Genetics of Aggression in Voles 145<br />

Israel, S., Lerer, E., Shalev, I., Uzefovsky, F., Reibold, M., Bachner-Melman, R., Granot, R.,<br />

Bornstein, G., Knafo, A., Yirmiya, N., and Ebstein, R. P. (2008). Molecular genetic studies of<br />

the arginine vasopressin 1a receptor (AVPR1a) and the oxytocin receptor (OXTR) in human<br />

behaviour: From autism to altruism with some notes in between. Prog. Brain Res. 170, 435–449.<br />

Jackson, D., Burns, R., Trksak, G., Simeone, B., DeLeon, K. R., Connor, D. F., Harrison, R. J., and<br />

Melloni, R. H., Jr. (2005). Anterior hypothalamic vasopressin modulates the aggression-stimulating<br />

effects of adolescent cocaine exposure in Syrian hamsters. Neuroscience 133, 635–646.<br />

Jannett, F. (1982). Nesting patterns of adult voles, Microtus montanus, in field populations.<br />

J. Mammal. 63, 495–498.<br />

Johnson, A. E., Audigier, S., Rossi, F., Jard, S., Tribollet, E., and Barberis, C. (1993). Localization and<br />

characterization of vasopressin binding sites in the rat brain using an iodinated linear AVP<br />

antagonist. Brain Res. 622, 9–16.<br />

Katz, L. F., Ball, G. F., and Nelson, R. J. (1999). Elevated Fos-like immunoreactivity in the brains of<br />

postpartum female prairie voles, Microtus ochrogaster. Cell Tissue Res. 298, 425–435.<br />

Kelley, A. E., and Berridge, K. C. (2002). The neuroscience of natural rewards: Relevance to<br />

addictive drugs. J. Neurosci. 22, 3306–3311.<br />

Kim, S. J., Young, L. J., Gonen, D., Veenstra-VanderWeele, J., Courchesne, R., Courchesne, E.,<br />

Lord, C., Leventhal, B. L., Cook, E. H., Jr., and Insel, T. R. (2002). Transmission disequilibrium<br />

testing of arginine vasopressin receptor 1A (AVPR1A) polymorphisms in autism. Mol. Psychiatry<br />

7, 503–507.<br />

Kirkpatrick, B., Kim, J. W., and Insel, T. R. (1994). Limbic system fos expression associated with<br />

paternal behavior. Brain Res. 658, 112–118.<br />

Knyshevski, I., Connor, D. F., Harrison, R. J., Ricci, L. A., and Melloni, R. H., Jr. (2005a). Persistent<br />

activation of select forebrain regions in aggressive, adolescent cocaine-treated hamsters. Behav.<br />

Brain Res. 159, 277–286.<br />

Knyshevski, I., Ricci, L. A., McCann, T. E., and Melloni, R. H., Jr. (2005b). Serotonin type-1A<br />

receptors modulate adolescent, cocaine-induced offensive aggression in hamsters. Physiol. Behav.<br />

85, 167–176.<br />

Koolhaas, J. M., Van den Brink, T. H. C., Roozendaal, B., and Boorsma, F. (1990). Medial amygdala<br />

and aggressive behavior: Interaction between testosterone and vasopressin. Aggress. Behav. 16,<br />

223–229.<br />

Kozorovitskiy, Y., Hughes, M., Lee, K., and Gould, E. (2006). Fatherhood affects dendritic spines and<br />

vasopressin V1a receptors in the primate prefrontal cortex. Nat. Neurosci. 9, 1094–1095.<br />

Kramer, K. M., Choe, C., Carter, C. S., and Cushing, B. S. (2006). Developmental effects of oxytocin<br />

on neural activation and neuropeptide release in response to social stimuli. Horm. Behav. 49,<br />

206–214.<br />

Krug, E. G., Dahlberg, L. L., Mercy, J. A., Zwi, A. B., and Lozito, R. (2002). World Report on<br />

Violence and Health. World Health Organization, Geneva.<br />

Kruk, M. R. (1991). Ethology and pharmacology of hypothalamic aggression in the rat. Neurosci.<br />

Biobehav. Rev. 15, 527–538.<br />

Kruk, M. R., Van der Laan, C. E., Mos, J., Van der Poel, A. M., Meelis, W., and Olivier, B. (1984).<br />

Comparison of aggressive behaviour induced by electrical stimulation in the hypothalamus of<br />

male and female rats. Prog. Brain Res. 61, 303–314.<br />

Kuptsov, P. A., Pleskacheva, M. G., Voronkova, D. N., Lipp, H. P., and Anokhin, K. V. (2005).<br />

Features of the c-Fos gene expression along the hippocampal rostro-caudal axis in common voles<br />

after rapid spatial learning. Zh. Vyssh. Nerv. Deiat. Im. I P Pavlova 55, 231–240.<br />

Landgraf, R., Frank, E., Aldag, J. M., Neumann, I. D., Sharer, C. A., Ren, X., Terwilliger, E. F.,<br />

Niwa, M., Wigger, A., and Young, L. J. (2003). Viral vector-mediated gene transfer of the vole<br />

V1a vasopressin receptor in the rat septum: Improved social discrimination and active social<br />

behaviour. Eur. J. Neurosci. 18, 403–411.


146 Gobrogge and Wang<br />

Lim, M. M., and Young, L. J. (2004). Vasopressin-dependent neural circuits underlying pair bond<br />

formation in the monogamous prairie vole. Neuroscience 125, 35–45.<br />

Lim, M. M., Wang, Z., Olazabal, D. E., Ren, X., Terwilliger, E. F., and Young, L. J. (2004). Enhanced<br />

partner preference in a promiscuous species by manipulating the expression of a single gene.<br />

Nature 429, 754–757.<br />

Lin, D., Boyle, M. P., Dollar, P., Lee, H., Lein, E. S., Perona, P., and Anderson, D. J. (2011).<br />

Functional identification of an aggression locus in the mouse hypothalamus. Nature 470,<br />

221–226.<br />

Lindvall, O., and Stenevi, U. (1978). Dopamine and noradrenaline neurons projecting to the septal<br />

area in the rat. Cell Tissue Res. 190, 383–407.<br />

Liu, Y., and Wang, Z. (2003). Nucleus accumbens oxytocin and dopamine interact to regulate pair<br />

bond formation in female prairie voles. Neuroscience 121, 537–544.<br />

Liu, Y., Curtis, J. T., and Wang, Z. (2001). Vasopressin in the lateral septum regulates pair bond<br />

formation in male prairie voles (Microtus ochrogaster). Behav. Neurosci. 115, 910–919.<br />

Liu, Y., Aragona, B. J., Young, K. A., Dietz, D. M., Kabbaj, M., Mazei-Robison, M., Nestler, E. J., and<br />

Wang, Z. (2010). Nucleus accumbens dopamine mediates amphetamine-induced impairment of<br />

social bonding in a monogamous rodent species. Proc. Natl. Acad. Sci. USA 107, 1217–1222.<br />

Madan, A., Beech, D. J., and Flint, L. (2001). Drugs, guns, and kids: The association between<br />

substance use and injury caused by interpersonal violence. J. Pediatr. Surg. 36, 440–442.<br />

Maeda, H., and Mogenson, G. J. (1980). An electrophysiological study of inputs to neurons of the<br />

ventral tegmental area from the nucleus accumbens and medial preoptic-anterior hypothalamic<br />

areas. Brain Res. 197, 365–377.<br />

McGuire, B., and Novak, M. (1984). A comparison of maternal behavior in the meadow vole<br />

(Microtus pennsylvanicus), prairie vole (M. ochrogaster) and pine vole (M. pinetorum). Anim.<br />

Behav. 32, 1132–1141.<br />

McGuire, B., and Novak, M. (1986). Parental care and its relation to social organization in the<br />

montane vole. J. Mammal. 67, 305–311.<br />

Melloni, R. H., Jr., and Ferris, C. F. (1996). Adolescent anabolic steroid use and aggressive behavior<br />

in golden hamsters. Ann. N. Y. Acad. Sci. 794, 372–375.<br />

Melloni, R. H., Jr., Connor, D. F., Hang, P. T., Harrison, R. J., and Ferris, C. F. (1997). Anabolicandrogenic<br />

steroid exposure during adolescence and aggressive behavior in golden hamsters.<br />

Physiol. Behav. 61, 359–364.<br />

Melloni, R. H., Jr., Connor, D. F., Todtenkopf, M. S., DeLeon, K. R., Sanyal, P., and Harrison, R. J.<br />

(2001). Repeated cocaine treatment activates flank marking in adolescent female hamsters.<br />

Physiol. Behav. 73, 561–570.<br />

Meyer-Lindenberg, A., Kolachana, B., Gold, B., Olsh, A., Nicodemus, K. K., Mattay, V., Dean, M.,<br />

and Weinberger, D. R. (2008). Genetic variants in AVPR1A linked to autism predict amygdala<br />

activation and personality traits in healthy humans. Mol. Psychiatry 10, 968–975.<br />

Michard-Vanhee, C. (1988). Aggressive behavior induced in female mice by an early single injection<br />

of testosterone is genotype dependent. Behav. Genet. 18, 1–12.<br />

Michell, R. H., Kirk, C. J., and Billah, M. M. (1979). Hormonal stimulation of phosphatidylinositol<br />

breakdown with particular reference to the hepatic effects of vasopressin. Biochem. Soc. Trans. 7,<br />

861–865.<br />

Miczek, K. A., de Almeida, R. M., Kravitz, E. A., Rissman, E. F., de Boer, S. F., and Raine, A. (2007).<br />

Neurobiology of escalated aggression and violence. J. Neurosci. 27, 11803–11806.<br />

Mokuau, N. (2002). Culturally based interventions for substance use and child abuse among native<br />

Hawaiians. Public Health Rep. 117(Suppl. 1), S82–S87.<br />

Moore, T. M., Stuart, G. L., Meehan, J. C., Rhatigan, D. L., Hellmuth, J. C., and Keen, S. M. (2008).<br />

Drug abuse and aggression between intimate partners: A meta-analytic review. Clin. Psychol. Rev.<br />

28, 247–274.


6. Genetics of Aggression in Voles 147<br />

Motta, S. C., Goto, M., Gouveia, F. V., Baldo, M. V., Canteras, N. S., and Swanson, L. W. (2009).<br />

Dissecting the brain’s fear system reveals the hypothalamus is critical for responding in subordinate<br />

conspecific intruders. Proc. Natl. Acad. Sci. USA 106, 4870–4875.<br />

Murphy, T. H., Worley, P. F., and Baraban, J. M. (1991). L-type voltage-sensitive calcium channels<br />

mediate synaptic activation of immediate early genes. Neuron 7, 625–635.<br />

Nesse, R. M., and Berridge, K. C. (1997). Psychoactive drug use in evolutionary perspective. Science<br />

278, 63–66.<br />

Nestler, E. J., and Aghajanian, G. K. (1997). Molecular and cellular basis of addiction. Science 278,<br />

58–63.<br />

Neve, K. A., Seamans, J. K., and Trantham-Davidson, H. (2004). Dopamine receptor signaling.<br />

J. Recept. Signal Transduct. Res. 24, 165–205.<br />

Newman, S. W. (1999). The medial extended amygdala in male reproductive behavior. A node in<br />

the mammalian social behavior network. Ann. N. Y. Acad. Sci. 877, 242–257.<br />

Northcutt, K. V., and Lonstein, J. S. (2009). Social contact elicits immediate-early gene expression<br />

in dopaminergic cells of the male prairie vole extended olfactory amygdala. Neuroscience 163,<br />

9–22.<br />

Northcutt, K. V., Wang, Z., and Lonstein, J. S. (2007). Sex and species differences in tyrosine<br />

hydroxylase-synthesizing cells of the rodent olfactory extended amygdala. J. Comp. Neurol. 500,<br />

103–115.<br />

O’Farrell, T. J., and Fals-Stewart, W. (2000). Behavioral couples therapy for alcoholism and drug<br />

abuse. J. Subst. Abuse Treat. 18, 51–54.<br />

Oliveras, D., and Novak, M. (1986). A comparison of paternal behavior in the meadow vole,<br />

Microtus pennsylvanicus, the pine vole, Microtus pinetorum, and prairie vole, Microtus ochrogaster.<br />

Anim. Behav. 34, 519–526.<br />

Ophir, A. G., Wolff, J. O., and Phelps, S. M. (2008). Variation in neural V1aR predicts sexual fidelity<br />

and space use among male prairie voles in semi-natural settings. Proc. Natl. Acad. Sci. USA 105,<br />

1249–1254.<br />

Panksepp, J., Knutson, B., and Burgdorf, J. (2002). The role of brain emotional systems in addictions:<br />

A neuro-evolutionary perspective and new ‘self-report’ animal model. Addiction 97, 459–469.<br />

Pitkow, L. J., Sharer, C. A., Ren, X., Insel, T. R., Terwilliger, E. F., and Young, L. J. (2001).<br />

Facilitation of affiliation and pair-bond formation by vasopressin receptor gene transfer into the<br />

ventral forebrain of a monogamous vole. J. Neurosci. 21, 7392–7396.<br />

Prichard, Z. M., Mackinnon, A. J., Jorm, A. F., and Easteal, S. (2007). AVPR1A and OXTR<br />

polymorphisms are associated with sexual and reproductive behavioral phenotypes in humans.<br />

Hum. Mutat. 28, 1150. Mutation in brief no. 981. Online.<br />

Raggenbass, M., Goumaz, M., Sermasi, E., Tribollet, E., and Dreifuss, J. J. (1991). Vasopressin<br />

generates a persistent voltage-dependent sodium current in a mammalian motoneuron. J. Neurosci.<br />

11, 1609–1616.<br />

Ramamurthi, B. (1988). Stereotactic operation in behaviour disorders. Amygdalotomy and hypothalamotomy.<br />

Acta Neurochir. Suppl. 44, 152–157.<br />

Ricci, L. A., Schwartzer, J. J., and Melloni, R. H., Jr. (2009). Alterations in the anterior hypothalamic<br />

dopamine system in aggressive adolescent AAS-treated hamsters. Horm. Behav. 55, 348–355.<br />

Riters, L. V., and Panksepp, J. (1997). Effects of vasotocin on aggressive behavior in male Japanese<br />

quail. Ann. N. Y. Acad. Sci. 807, 478–480.<br />

Roeling, T. A., Kruk, M. R., Schuurmans, R., and Veening, J. G. (1993). Behavioural responses of<br />

bicucculline methiodide injections into the ventral hypothalamus of freely moving, socially<br />

interacting rats. Brain Res. 615, 121–127.<br />

Ryding, E., Lindstrom, M., and Traskman-Bendz, L. (2008). The role of dopamine and serotonin in<br />

suicidal behaviour and aggression. Prog. Brain Res. 172, 307–315.


148 Gobrogge and Wang<br />

Sano, K., Yoshioka, M., Ogashiwa, M., Ishijima, B., and Ohye, C. (1966). Postero-medial hypothalamotomy<br />

in the treatment of aggressive behaviors. Confin. Neurol. 27, 164–167.<br />

Schwartzer, J. J., and Melloni, R. H., Jr. (2010a). Anterior hypothalamic dopamine D2 receptors<br />

modulate adolescent anabolic/androgenic steroid-induced offensive aggression in the Syrian<br />

hamster. Behav. Pharmacol. 21, 314–322.<br />

Schwartzer, J. J., and Melloni, R. H., Jr. (2010b). Dopamine activity in the lateral anterior hypothalamus<br />

modulates AAS-induced aggression through D2 but not D5 receptors. Behav. Neurosci. 124,<br />

645–655.<br />

Schwartzer, J. J., Ricci, L. A., and Melloni, R. H., Jr. (2009). Interactions between the dopaminergic<br />

and GABAergic neural systems in the lateral anterior hypothalamus of aggressive AAS-treated<br />

hamsters. Behav. Brain Res. 203, 15–22.<br />

Siegel, A., Roeling, T. A., Gregg, T. R., and Kruk, M. R. (1999). Neuropharmacology of brainstimulation-evoked<br />

aggression. Neurosci. Biobehav. Rev. 23, 359–389.<br />

Siever, L. J. (2008). Neurobiology of aggression and violence. Am. J. Psychiatry 165, 429–442.<br />

Smeltzer, M. D., Curtis, J. T., Aragona, B. J., and Wang, Z. (2006). Dopamine, oxytocin, and<br />

vasopressin receptor binding in the medial prefrontal cortex of monogamous and promiscuous<br />

voles. Neurosci. Lett. 394, 146–151.<br />

Son, M. C., and Brinton, R. D. (2001). Regulation and mechanism of L-type calcium channel<br />

activation via V1a vasopressin receptor activation in cultured cortical neurons. Neurobiol.<br />

Learn. Mem. 76, 388–402.<br />

Spunt, B., Brownstein, H. H., Crimmins, S. M., Langley, S., and Spanjol, K. (1998). Alcohol-related<br />

homicides committed by women. J. Psychoactive Drugs 30, 33–43.<br />

Stowe, J. R., Liu, Y., Curtis, J. T., Freeman, M. E., and Wang, Z. (2005). Species differences in<br />

anxiety-related responses in male prairie and meadow voles: The effects of social isolation. Physiol.<br />

Behav. 86, 369–378.<br />

Stribley, J. M., and Carter, C. S. (1999). Developmental exposure to vasopressin increases aggression<br />

in adult prairie voles. Proc. Natl. Acad. Sci. USA 96, 12601–12604.<br />

Swanson, L. W. (1982). The projections of the ventral tegmental area and adjacent regions:<br />

A combined fluorescent retrograde tracer and immunofluorescence study in the rat. Brain Res.<br />

Bull. 9, 321–353.<br />

Swartz, M. S., Swanson, J. W., Hiday, V. A., Borum, R., Wagner, H. R., and Burns, B. J. (1998).<br />

Violence and severe mental illness: The effects of substance abuse and nonadherence to<br />

medication. Am. J. Psychiatry 155, 226–231.<br />

Tamarin, R. (1985). Biology of new world Microtus. Am. Soc. Mamm. Spec. Pub. 8. Shippensburg,<br />

PA.<br />

Thibonnier, M. (1992). Signal transduction of V1-vascular vasopressin receptors. Regul. Pept. 38,<br />

1–11.<br />

Thibonnier, M., Bayer, A. L., Simonson, M. S., and Douglas, J. G. (1992). Effects of amiloride<br />

analogues on AVP binding and activation of V1-receptor-expressing cells. Am. J. Physiol. 262,<br />

E76–E86.<br />

Thibonnier, M., Goraya, T., and Berti-Mattera, L. (1993). G protein coupling of human platelet V1<br />

vascular vasopressin receptors. Am. J. Physiol. 264, C1336–C1344.<br />

Thibonnier, M., Auzan, C., Madhun, Z., Wilkins, P., Berti-Mattera, L., and Clauser, E. (1994).<br />

Molecular cloning, sequencing, and functional expression of a cDNA encoding the human V1a<br />

vasopressin receptor. J. Biol. Chem. 269, 3304–3310.<br />

Thompson, R., Gupta, S., Miller, K., Mills, S., and Orr, S. (2004). The effects of vasopressin on<br />

human facial responses related to social communication. Psychoneuroendocrinology 29, 35–48.<br />

Tidey, J. W., and Miczek, K. A. (1992). Morphine withdrawal aggression: Modification with D1 and<br />

D2 receptor agonists. Psychopharmacology (Berl.) 108, 177–184.


6. Genetics of Aggression in Voles 149<br />

Tubbiola, M. L., and Wysocki, C. J. (1997). FOS immunoreactivity after exposure to conspecific or<br />

heterospecific urine: Where are chemosensory cues sorted Physiol. Behav. 62, 867–870.<br />

Vale, J. R., Ray, D., and Vale, C. A. (1972). The interaction of genotype and exogenous neonatal<br />

androgen: Agonistic behavior in female mice. Behav. Biol. 7, 321–334.<br />

Veenema, A. H., Blume, A., Niederle, D., Buwalda, B., and Neumann, I. D. (2006). Effects of early<br />

life stress on adult male aggression and hypothalamic vasopressin and serotonin. Eur. J. Neurosci.<br />

24, 1711–1720.<br />

Veenema, A. H., Beiderbeck, D. I., Lukas, M., and Neumann, I. D. (2010). Distinct correlations of<br />

vasopressin release within the lateral septum and the bed nucleus of the stria terminalis with the<br />

display of intermale aggression. Horm. Behav. 58, 273–281.<br />

Veening, J. G., Coolen, L. M., de Jong, T. R., Joosten, H. W., de Boer, S. F., Koolhaas, J. M., and<br />

Olivier, B. (2005). Do similar neural systems subserve aggressive and sexual behaviour in male<br />

rats Insights from c-Fos and pharmacological studies. Eur. J. Pharmacol. 526, 226–239.<br />

Villalba, C., Boyle, P. A., Caliguri, E. J., and De Vries, G. J. (1997). Effects of the selective serotonin<br />

reuptake inhibitor fluoxetine on social behaviors in male and female prairie voles (Microtus<br />

ochrogaster). Horm. Behav. 32, 184–191.<br />

Walsh, C., MacMillan, H. L., and Jamieson, E. (2003). The relationship between parental substance<br />

abuse and child maltreatment: Findings from the Ontario Health Supplement. Child Abuse Negl.<br />

27, 1409–1425.<br />

Walum, H., Westberg, L., Henningsson, S., Neiderhiser, J. M., Reiss, D., Igl, W., Ganiban, J. M.,<br />

Spotts, E. L., Pedersen, N. L., Eriksson, E., and Lichtenstein, P. (2008). Genetic variation in the<br />

vasopressin receptor 1a gene (AVPR1A) associates with pair-bonding behavior in humans. Proc.<br />

Natl. Acad. Sci. USA 105, 14153–14156.<br />

Wang, Z. (1995). Species differences in the vasopressin-immunoreactive pathways in the bed nucleus<br />

of the stria terminalis and medial amygdaloid nucleus in prairie voles (Microtus ochrogaster) and<br />

meadow voles (Microtus pennsylvanicus). Behav. Neurosci. 109, 305–311.<br />

Wang, Z., and Aragona, B. J. (2004). Neurochemical regulation of pair bonding in male prairie voles.<br />

Physiol. Behav. 83, 319–328.<br />

Wang, Z., Smith, W., Major, D. E., and De Vries, G. J. (1994). Sex and species differences in the<br />

effects of cohabitation on vasopressin messenger RNA expression in the bed nucleus of the stria<br />

terminalis in prairie voles (Microtus ochrogaster) and meadow voles (Microtus pennsylvanicus).<br />

Brain Res. 650, 212–218.<br />

Wang, Z., Zhou, L., Hulihan, T. J., and Insel, T. R. (1996). Immunoreactivity of central vasopressin<br />

and oxytocin pathways in microtine rodents: A quantitative comparative study. J. Comp. Neurol.<br />

366, 726–737.<br />

Wang, Z., Hulihan, T. J., and Insel, T. R. (1997a). Sexual and social experience is associated with<br />

different patterns of behavior and neural activation in male prairie voles. Brain Res. 767,<br />

321–332.<br />

Wang, Z., Liu, Y., Young, L. J., and Insel, T. R. (1997b). Developmental changes in forebrain<br />

vasopressin receptor binding in prairie voles (Microtus ochrogaster) and montane voles (Microtus<br />

montanus). Ann. N. Y. Acad. Sci. 807, 510–513.<br />

Wang, Z., Young, L. J., Liu, Y., and Insel, T. R. (1997c). Species differences in vasopressin receptor<br />

binding are evident early in development: Comparative anatomic studies in prairie and montane<br />

voles. J. Comp. Neurol. 378, 535–546.<br />

Wang, Z., Young, L. J., De Vries, G. J., and Insel, T. R. (1998). Voles and vasopressin: A review of<br />

molecular, cellular, and behavioral studies of pair bonding and paternal behaviors. Prog. Brain Res.<br />

119, 483–499.<br />

Wang, Z., Yu, G., Cascio, C., Liu, Y., Gingrich, B., and Insel, T. R. (1999). Dopamine D2 receptormediated<br />

regulation of partner preferences in female prairie voles (Microtus ochrogaster): A<br />

mechanism for pair bonding Behav. Neurosci. 113, 602–611.


150 Gobrogge and Wang<br />

White, F. J., and Kalivas, P. W. (1998). Neuroadaptations involved in amphetamine and cocaine<br />

addiction. Drug Alcohol Depend. 51, 141–153.<br />

Williams, J. R., Carter, C. S., and Insel, T. (1992). Partner preference development in female prairie<br />

voles is facilitated by mating or the central infusion of oxytocin. Ann. N. Y. Acad. Sci. 652,<br />

487–489.<br />

Williams, J. R., Insel, T. R., Harbaugh, C. R., and Carter, C. S. (1994). Oxytocin administered<br />

centrally facilitates formation of a partner preference in female prairie voles (Microtus ochrogaster).<br />

J. Neuroendocrinol. 6, 247–250.<br />

Winslow, J. T., Hastings, N., Carter, C. S., Harbaugh, C. R., and Insel, T. R. (1993). A role for<br />

central vasopressin in pair bonding in monogamous prairie voles. Nature 365, 545–548.<br />

Wise, R. A. (2002). Brain reward circuitry: Insights from unsensed incentives. Neuron 36, 229–240.<br />

Yamada, S., Uchida, S., Ohkura, T., Kimura, R., Yamaguchi, M., Suzuki, M., and Yamamoto, M.<br />

(1996). Alterations in calcium antagonist receptors and calcium content in senescent brain and<br />

attenuation by nimodipine and nicardipine. J. Pharmacol. Exp. Ther. 277, 721–727.<br />

Young, L. J. (1999). Frank A. Beach Award. Oxytocin and vasopressin receptors and species-typical<br />

social behaviors. Horm. Behav. 36, 212–221.<br />

Young, L. J., and Wang, Z. (2004). The neurobiology of pair bonding. Nat. Neurosci. 7, 1048–1054.<br />

Young, L. J., Winslow, J. T., Nilsen, R., and Insel, T. R. (1997). Species differences in V1a receptor<br />

gene expression in monogamous and nonmonogamous voles: Behavioral consequences. Behav.<br />

Neurosci. 111, 599–605.<br />

Young, L. J., Wang, Z., and Insel, T. R. (1998). Neuroendocrine bases of monogamy. Trends Neurosci.<br />

21, 71–75.<br />

Young, L. J., Nilsen, R., Waymire, K. G., MacGregor, G. R., and Insel, T. R. (1999). Increased<br />

affiliative response to vasopressin in mice expressing the V1a receptor from a monogamous vole.<br />

Nature 400, 766–768.<br />

Young, K. A., Liu, Y., and Wang, Z. (2008). The neurobiology of social attachment: A comparative<br />

approach to behavioral, neuroanatomical, and neurochemical studies. Comp. Biochem. Physiol. C<br />

Toxicol. Pharmacol. 148, 401–410.<br />

Young, K. A., Gobrogge, K. L., Liu, Y., and Wang, Z. (2011a). The neurobiology of pair bonding:<br />

Insights from a socially monogamous rodent. Front. Neuroendocrinol. 32, 53–69.<br />

Young, K. A., Gobrogge, K. L., and Wang, Z. (2011b). The role of mesocorticolimbic dopamine in<br />

regulating interactions between drugs of abuse and social behavior. Neurosci. Biobehav. Rev. 35,<br />

498–515.


7<br />

The Neurochemistry of Human<br />

Aggression<br />

Rachel Yanowitch and Emil F. Coccaro<br />

Clinical Neuroscience Research Unit, Department of Psychiatry, The <strong>University</strong><br />

of Chicago Pritzker School of Medicine, Chicago, Illinois, USA<br />

I. Introduction<br />

II. Serotonin<br />

III. Dopamine<br />

IV. Norepinephrine (Noradrenaline)<br />

V. GABA<br />

VI. Peptides<br />

VII. Conclusion<br />

References<br />

ABSTRACT<br />

Various data from scientific research studies conducted over the past three decades<br />

suggest that central neurotransmitters play a key role in the modulation of<br />

aggression in all mammalian species, including humans. Specific neurotransmitter<br />

systems involved in mammalian aggression include serotonin, dopamine, norepinephrine,<br />

GABA, and neuropeptides such as vasopressin and oxytocin. Neurotransmitters<br />

not only help to execute basic behavioral components but also serve<br />

to modulate these preexisting behavioral states by amplifying or reducing their<br />

effects. This chapter reviews the currently available data to present a contemporary<br />

view of how central neurotransmitters influence the vulnerability for aggressive<br />

behavior and/or initiation of aggressive behavior in social situations. Data<br />

reviewed in this chapter include emoiric information from neurochemical,<br />

pharmaco-challenge, molecular genetic and neuroimaging studies. ß 2011, Elsevier Inc.<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00005-8


152 Yanowitch and Coccaro<br />

I. INTRODUCTION<br />

Since the late 1970s, data from scientific research studies have suggested that<br />

endogenous brain chemicals called neurotransmitters play a key role in the<br />

modulation of aggression. Human aggression is a multidimensional behavior<br />

that is determined by an amalgamation of biological, genetic, environmental,<br />

and psychological factors. Neurotransmitters not only help to execute these basic<br />

behavioral components but also serve to modulate these preexisting behavioral<br />

states by amplifying or reducing their effects. Genetic abnormalities in a number<br />

of neurotransmitter pathways have been implicated in aggression-related disorders.<br />

Current and future research aims to understand how these neurotransmitters<br />

function both normally and abnormally to mediate aggression and other<br />

human behaviors. With the evolution of genetic testing and continued development<br />

of neuroimaging technologies such as functional magnetic resonance<br />

imaging (fMRI) and positron emission tomography (PET) scanning, the ability<br />

of the scientific researcher to investigate the brain’s constellation of synapses and<br />

neurotransmitters is growing ever more proficient. While it is clear from these<br />

studies that neurotransmitters contribute significantly to the predisposition of an<br />

individual toward aggressiveness, whether neurotransmitter dysfunction alone is<br />

sufficient to cause violent aggression remains unclear.<br />

Aggression may be impulsive or premeditated in nature. In the former<br />

case, impulsivity defines, or describes, the aggression. That is, it is the aggression<br />

that is impulsive not that the person is aggressive and at other times impulsive,<br />

though that may be true as well. Diagnoses associated with impulsive aggression<br />

include Intermittent Explosive Disorder (IED) characterized by frequent and<br />

problematic impulsive aggressive outbursts, and Borderline Personality Disorder<br />

(BPD) characterized by instability in self-image, in interpersonal relationships as<br />

well as impulsivity and affect (including anger and aggression). In the latter case,<br />

the aggression is planned and carried out in order to achieve some tangible goal.<br />

Diagnoses associated with this type of aggression include Antisocial Personality<br />

Disorder (AsPD) which is characterized by a pattern of disregard for, and violation<br />

of, the rights of others. These types of aggression are not mutually exclusive,<br />

however, and some individuals display both types of aggression at different times.<br />

II. SEROTONIN<br />

Serotonin or 5-hydroxytryptamine (5-HT) is a multipurpose monoamine neurotransmitter<br />

derived from the amino acid, L-tryptophan, and has been implicated<br />

as an important regulator of mood (Kumar et al., 2010; Kunisato et al., 2010;<br />

Ruhé et al., 2007), appetite (Curzon, 1991; Dourish, 1995; Lam et al., 2010),<br />

gastrointestinal muscle contractility (Gershon, 2004; Xu et al., 2007), self-


7. The Neurochemistry of Human Aggression 153<br />

injurious behavior (Peddeer, 1992), and sleep (Monti, 2010; Monti and Jantos,<br />

2008; Monti and Monti, 2000). With respect to aggression and other behavioral<br />

disorders, serotonin action is highly complex and varies depending upon which<br />

receptor it is bound to, how much 5-HT is available in the synapse, how much<br />

enzymatic activity is present, and whether other agonists or antagonists are<br />

available for competitive binding. Both the clinical and molecular data on<br />

central 5-HT function in the mammalian brain overwhelmingly suggests that a<br />

reduction in 5-HT activity in emotion-modulating brain regions such as the<br />

prefrontal cortex and the anterior cingulate cortex leads to a predisposition for<br />

impulsive aggressiveness (New et al., 2002; Parsey et al., 2002; Seo et al., 2008;<br />

Siever et al., 1999). Research suggests that 5-HT significantly contributes to the<br />

genetically determined differences seen in individuals and that its primary<br />

mechanism of action is via the genes encoding major components of 5-HT<br />

viability in the brain such as the enzymes tryptophan hydroxylase-1 and monoamine<br />

oxidase A (MAOA) (Popova, 2008). Most of the literature discussing 5-<br />

HT regulation of aggression focuses on 5-HT metabolite levels and the functional<br />

state of 5-HT receptors.<br />

The first literature written on serotonin and impulsive aggressive behavior<br />

in human subjects came from two independent research groups (Asberg<br />

et al., 1976; Sheard et al., 1976) in 1976. Sheard et al. reported that administration<br />

of the putative 5-HT-enhancing agent, lithium carbonate, significantly<br />

reduced impulsive aggressive behavior in a prison inmate population. Asberg<br />

et al. found that lower concentrations of lumbar CSF 5-hydroxyindoleactic acid<br />

(5-HIAA), the most stable 5-HT metabolite in the brain, were correlated with<br />

violent and suicidal behavior. In 1979, Brown et al. studied 26 males with<br />

significant personality disorder traits (Brown et al., 1979). CSF amine metabolite<br />

levels of serotonin (5-HIAA), norepinephrine (3-methoxy-4-hydroxy-phenylglycol,<br />

MHPG), and dopamine (homovanillic acid, HVA), respectively, were<br />

studied. CSF 5-HIAA was significantly negatively correlated with aggression<br />

(r¼ 0.78), and MHPG was significantly positively correlated with aggression<br />

(r¼0.64). Brown et al. replicated this finding for CSF 5-HIAA and extended<br />

these findings to include other measures of aggression such as “psychopathic<br />

deviance” (i.e., defiance of authority and impulsivity) (Brown et al., 1982;<br />

Coccaro and Siever, 2002). Despite the fact that a number of subsequent studies<br />

supported the findings of an inverse relationship between aggression and CSF<br />

5-HIAA levels (Kruesi et al., 1990; Lidberg et al., 1985; Limson et al., 1991;<br />

Linnoila et al., 1983), additional reports also suggest a direct (Castellanos et al.,<br />

1994; Moller et al., 1996; Prochazka and Agren, 2003) or no relationship<br />

(Coccaro et al., 1997c; Gardner et al., 1990) between the two. In a recent<br />

paper by Coccaro et al., the authors were able to reconcile the disputed data by<br />

reconsidering the CSF 5-HIAA levels in the con<strong>text</strong> of (1) the severity of the<br />

aggression of the individual and (2) the CSF HVA levels present concomitantly


154 Yanowitch and Coccaro<br />

(Coccaro and Lee, 2010). Under this new paradigm, the results emerged against<br />

Brown’s preliminary findings: CSF 5-HIAA concentrations varied directly with<br />

aggression and CSF HVA concentrations varied inversely. In this model, a<br />

deficiency hypothesis of 5-HT for aggressiveness is only fulfilled if presynaptic<br />

release of 5-HT is being reduced and there is compensation of postsynaptic 5-HT<br />

receptor function (Coccaro, 1998).<br />

Evidence for a model in which postsynaptic 5-HT receptor function is<br />

altered by presynaptic reduction begins with Stanley et al.’s (1982) report<br />

demonstrating a reduced number of presynaptic 5-HT transporter sites in aggressive<br />

suicide victims as compared with accident victims (Stanley et al., 1982). The<br />

following year, Stanley and Mann published additional results showing increased<br />

postsynaptic 5-HT2A receptor sites in suicide subjects (Stanley and Mann,<br />

1983), suggesting that, in addition to modified responsiveness, there may be a<br />

change in receptor number as well. In response to these data, researchers<br />

designed psychopharmacologic challenge studies in order to further assess preand<br />

postsynaptic function in premortem subjects (Coccaro, 1998). These pharmacochallenge<br />

studies involve the activation of a specific neurotransmitter<br />

system through the administration and consequent ligand–receptor interaction<br />

of a pharmacologic agent. Subsequent signaling cascades result in physiological<br />

events that trigger homeostatic, behavioral, and hormonal alterations that can<br />

be measured as an index of the responsiveness of the neurotransmitter system in<br />

question (Coccaro and Kavoussi, 1994).<br />

The first report of a correlation between aggression and pharmacochallenge<br />

studies were published by Coccaro et al., 1989. In this study, prolactin<br />

responses to 60 mg of oral D,L-fenfluramine of 45 males with major affective<br />

(n¼25) and/or personality (n¼20) disorder were compared to those of 18 healthy<br />

male controls. D,L-fenfluramine was chosen as a challenge probe because of its<br />

properties as a serotonin-releasing agent. Its mechanism of action is the release of<br />

serotonin by disrupting vesicular storage of the neurotransmitter and reversing<br />

serotonin transporter function (Welch and Lim, 2007). Since prolactin secretion<br />

is directly dependent upon 5-HT transmission, recording prolactin levels can<br />

provide an indirect but effective measurement of 5-HT activity (Coccaro et al.,<br />

1998a). Both groups of subjects demonstrated reduced prolactin responses to D,Lfenfluramine<br />

compared to controls. However, significant correlations appeared<br />

between reduced prolactin responses to D,L-fenfluramine and history of suicide<br />

attempts in all experimental subjects and impulsive aggression in males with<br />

personality disorder (Coccaro et al.,1989). These results suggest that altered 5-HT<br />

activity, specifically reduced receptor function, is apparent in subjects with aggression-related<br />

disorders. In a later study by Coccaro et al., the relationship between life<br />

history of aggression and prolactin response to D-fenfluramine and to CSF 5-HIAA<br />

concentration was evaluated (Coccaro et al., 1997a). The results were consistent<br />

with the altered postsynaptic 5-HT receptor function hypothesis: aggression was


7. The Neurochemistry of Human Aggression 155<br />

significantly and inversely correlated with prolactin responses to D-fenfluramine but<br />

not with CSF 5-HIAA levels. Notably, prolactin response to fenfluramine appears<br />

to reflect activation of 5-HT2 receptors, likely of the 5-HT2c subtype (Coccaro<br />

et al.,2010a). Additional research has revealed that prolactin responses to fenfluramine<br />

are also positively correlated with prolactin responses to m-CPP challenge,<br />

which assesses 5-HT postsynaptic receptor activation (Coccaro et al., 1997b).<br />

Thus far, seven subtypes of 5-HT receptors have been identified, ranging<br />

from 5-HTR1 to 5-HTR7. These receptors have been found to mediate both<br />

excitatory and inhibitory inputs in a number of brain regions associated with<br />

aggression (Siever, 2008), emotion regulation, and cognition. Inhibition of<br />

offensive aggression via agonists of 5-HT1a attenuates various forms of aggression<br />

in animals (Ferris et al., 1999; Joppa et al., 1996; Miczek et al., 2004;<br />

Olivier et al., 1995; Ricci et al., 2006; White et al., 1991). According to one<br />

study by Popova et al., less aggressive rats had higher 5-HT1a receptor expression<br />

in the midbrain (Popova et al., 2005), whereas in the frontal cortex, lower<br />

aggression was associated with a decrease in 5-HT1a receptor mRNA (Popova<br />

et al., 2007). In support of this hypothesis, a recent study showed that high 5-<br />

HT1a receptor density corresponded to increased aggressiveness in male Golden<br />

hamsters (Cervantes and Delville, 2009). Additional confirmation came from a<br />

study in which PET imaging of healthy subjects revealed that aggression is<br />

positively correlated to 5-HT1a receptor distribution in the dorsolateral and<br />

ventromedial prefrontal cortex, in the orbitofrontal cortex, and in the anterior<br />

cingulate cortex (Witte et al., 2009). Support of 5-HT1a’s involvement in<br />

aggression also comes from animal studies showing that offensive aggression in<br />

hamsters is inhibited by 5-HT1a receptors and facilitated by 5-HT3 receptor<br />

activation (Cervantes et al., 2010). Agonists of the 5-HT1a and 5-HT1b (5-<br />

HT1d in the human) receptors in the medial prefrontal cortex or septal area can<br />

increase aggressive behavior under specific conditions (Takahashi et al., 2011).<br />

Activation of these two receptors, as well as the 5-HT2a and 5-HT2c receptors<br />

in mesocorticolimbic areas, reduces species-typical and other aggressive behaviors.<br />

Pathological aggression is reportedly reduced by activation of 5-HT transporters,<br />

whereas dysfunction of genes that affect the 5-HT system directly such as<br />

MAOA cause an escalation in pathological aggression (Alia-Klein et al., 2008).<br />

With respect to 5-HT2a distribution, PET imaging demonstrates that<br />

individuals with IED and current physical aggression have increased receptor<br />

density in the orbitofrontal cortex when compared to individuals with IED but<br />

no current physical aggression or when compared to individuals who served as<br />

healthy controls (Rosell et al., 2010). This is similar to 5-HT1a distribution in<br />

the orbitofrontal cortex with the respect to aggression, as noted above (Witte<br />

et al., 2009). In a separate study, 5-HT2a receptor-binding activity was investigated<br />

in a nearby brain region, the dorsolateral prefrontal cortex, and a pattern<br />

different to that seen in the orbitofrontal cortex emerged. The results found that


156 Yanowitch and Coccaro<br />

5-HT2a receptor-binding potentials were lower in the dorsolateral prefrontal<br />

cortex in individuals with more severe impulsivity and aggression than in healthy<br />

subjects (Meyer et al., 2008). Lower 5-HT2a binding potentials occur at younger<br />

ages, when violent behavior is more frequent and is more prominent when<br />

impulsivity and aggression are more severe. However, this has not been causally<br />

linked; a low binding potential indicates low ligand-receptor-binding interaction<br />

and therefore the cause of these reduced binding potentials require further<br />

investigation. In a novel study led by Soloff et al., gender differences were<br />

identified in 5-HT2a availability with respect to aggression, negativism, and<br />

suspiciousness, highlighting a potential for gender biases and a need to control<br />

for them when conducting research (Soloff et al., 2010).<br />

Advances in molecular biology and neuroimaging have allowed for<br />

experimental studies in which 5-HT activity can be altered by tryptophan<br />

manipulation and subsequent brain activity and behavior monitored.<br />

These studies have long noted that 5-HT in the central nervous system (CNS),<br />

as well as in the periphery (e.g., through assessment of 5-HT transporter binding<br />

sites on the blood platelet) is reduced in aggressive behavior (Coccaro et al.,<br />

2010a). Platelet 5-HTT sites are structurally identical to corresponding sites on<br />

central 5-HT neurons (Lesch et al., 1993) and are therefore appropriate for further<br />

hypothesis testing. Preliminary studies by Stoff et al. found that lowered tryptophan<br />

levels and ingestion of alcohol were associated with increased aggression<br />

and lower 5-HTT binding (B)byH 3 -imipramine in normal adult males, suggesting<br />

that low 5-HT levels may be involved in the etiology of aggression and particularly,<br />

alcohol-induced violence (Pihl et al., 1995). Similarly, Birmaher et al., reported<br />

that a reduction in platelet H 3 -imipramine (B max ) was associated with aggression in<br />

children and adolescents (Birmaher et al.,1990). Two studies by Coccaro et al. have<br />

also demonstrated that the number of 5-HTT binding sites assessed by platelet H 3 -<br />

paroxetine is inversely related to aggression (Coccaro et al., 1996, 2010b). Individuals<br />

with IED also had fewer 5-HT transporter platelet binding sites than comparable<br />

personality disordered subjects without IED; measures of impulsivity did not<br />

correlate with 5-HTT binding in these studies.<br />

Animal and clinical studies have highlighted that impulsive aggression<br />

and its comorbid psychiatric disorders may result from a failure of the 5-HT<br />

system to communicate properly with other neurotransmitter systems, particularly<br />

that of dopamine (De Simoni et al., 1987). Specifically, failure of the<br />

dopamine and serotonin systems to success<strong>full</strong>y interact in the prefrontal cortex<br />

may underlie impulsive aggression (Seo et al., 2008). Van Erp and Miczek<br />

recently reported that increased aggressive behavior in male Long-Evans rats<br />

was related to both increased dopamine in the nucleus accumbens and reduced<br />

5-HT levels in the frontal cortex (Van Erp and Miczek, 2000). Previous studies<br />

have illustrated that serotonergic and dopaminergic systems are tightly linked<br />

(Daw et al., 2002; Kapur and Remington, 1996; Wong et al., 1995), and it is


7. The Neurochemistry of Human Aggression 157<br />

thought that subnormal serotonergic function may lead to dopaminergic hyperactivity,<br />

which in turn leads to impulsive and aggressive behavior (Seo et al.,<br />

2008).<br />

III. DOPAMINE<br />

Dopamine (DA) is a catecholamine neurotransmitter that acts both on the central<br />

and the sympathetic branch of the peripheral nervous systems. DA in the CNS has<br />

been linked to cognition (Browman et al., 2005; Heijtz et al., 2007), movement<br />

(Devos et al., 2003), sleep (Dzirasa et al., 2006; Lima et al., 2008), mood (Brown<br />

and Gershon, 1993; Diehl and Gershon, 1992), attention (Nieoullon, 2002), and<br />

learning and memory (Arias-Carrión andPöppel, 2007; Denenberg et al., 2004).<br />

Additionally, DA has developed a well-established and essential role as the<br />

neurotransmitter responsible for reward pathways involved in drug use (Pettit<br />

and Justice, 1991; Ranaldi et al., 1999; Weiss et al., 1992), eating (Hernandez<br />

and Hoebel, 1988), and sexual behavior (Hull et al., 1993; Pfaus et al., 1990). In<br />

patients with frontotemporal dementia, increased dopaminergic neurotransmission<br />

and serotonergic modulation of dopaminergic activity is, respectively, associated<br />

with agitated and aggressive behavior (Engelborghs et al.,2008), suggesting<br />

DA function contributes to the aggressive behavioral state. While much of what<br />

we know about dopamine and its biological effects remains to be determined, it is<br />

clear from the literature and data so far that dopamine is a neurotransmitter with a<br />

multitude of behavioral, physiological, and psychological capabilities.<br />

From a molecular perspective, research into DA function in aggressive<br />

individuals has revealed a spectrum of genetic variability that is linked to a<br />

number of polymorphisms in DA-specific genes. Led by Elena L. Grigorenko of<br />

Yale <strong>University</strong>’s Child Study Center, a coalition of scientists in 2010 found<br />

positive correlates between genetic polymorphisms in four genes involved in DA<br />

turnover and behavior pathology (Grigorenko et al., 2010). The four genes<br />

investigated included catechol-O-methyl-transferase (COMT), involved in catecholamine<br />

metabolism; dopamine beta hydroxylase (DbH), responsible for<br />

dopamine conversion; and MAOA and MAOB, both involved in the degradation<br />

of DA and/or other neurotransmitters. In this study, blood samples from 179<br />

adolescent offender males sentenced to a juvenile detention center in a large<br />

capital city in Northern Russia were compared for genetic analysis to those of<br />

two control groups of Russian male adolescents (n¼182; n¼60). While no<br />

single dopaminergic polymorphism revealed a definitive causal link to conduct<br />

disorder, criminality, aggression, or delinquency, combination of variants across<br />

two (COMT and DbH), three (COMT, DbH, and MAOB), or all four (COMT,<br />

DbH, MAOA, and MAOB) of the DA-specific genes investigated showed<br />

positive correlations with the behavioral traits in question. Nemoda et al.


158 Yanowitch and Coccaro<br />

found similar results among patients with borderline personality disorder in a<br />

separate study done in 2010 using young adults from low-to-moderate income<br />

households (n¼99) and major depressive or bipolar patients (n¼136) (Nemoda<br />

et al., 2010). The results of this study found that a promoter variant in the<br />

dopamine D4 receptor may be involved in the development of BPD traits,<br />

including aggression. The DA D4 receptor has been postulated as a candidate<br />

nexus for BPD because of its preferential expression in the prefrontal cortex (Oak<br />

et al., 2000), and its noted role in novelty-seeking and impulsivity (Munafò et al.,<br />

2008). Data from both experimental groups showed polymorphisms in COMT<br />

and the DA transporter (DAT1) of the dopamine D2 receptor were directly<br />

related to self-injurious and impulsive behavior, both BPD traits. Other reports<br />

have confirmed genetic abnormalities with COMT in the presence of BPD,<br />

including a recent study citing an over-representation of the low activity Met/<br />

Met genotype of the gene in BPD patients (n¼161) (Tadić et al., 2009).<br />

Interestingly, COMT and DAT1 are similarly implicated in bipolar and major<br />

depressive disorder (Joyce et al., 2006), suggesting DA dysfunction may encompass<br />

a much larger behavioral and physiological state.<br />

In 2008, Couppis and Kennedy published novel findings that found<br />

dopamine to be a reward for aggressive behavior in mice (Couppis and<br />

Kennedy, 2008). The authors had hypothesized that aggression could be linked<br />

to the dopaminergic receptors of the nucleus accumbens (NAc), citing their<br />

reputation as the most strongly implicated neurotransmitter in positive reinforcement<br />

(Wise, 2004) and reward behavior. The results of their studies showed<br />

that administration of a D1-like (D1 and D5) receptor antagonist (SCH-23390),<br />

or a D2-like (D2, D3, and D4) receptor antagonist (sulperide) into the NAc<br />

significantly reduced aggression responses when compared to administration<br />

outside of the NAc. In addition to the reductions in aggression, concomitant<br />

reductions in mobility were seen in these first studies. These results showing a<br />

simultaneous reduction in aggression and DA levels were consistent with previous<br />

reports that had suggested increased DA in the NAc led to increased<br />

aggression (Van Erp and Miczek, 2000). After Couppis and Kennedy’s initial<br />

publication, Schwartzer and Melloni reported similar findings that dopamine<br />

activity primarily mediated by D2 receptors was involved in modulating anabolic/androgenic<br />

steroid-induced offensive aggression in Syrian hamsters<br />

(Schwartzer and Melloni, 2010b). Interestingly, administration of the D2-like<br />

DA antagonist into the anterior hypothalamus (AH) rather than the NAc<br />

produced no side effects of reduced mobility: in a follow-up experiment, the<br />

authors reported that treatment of male Syrian hamsters with the D2-like<br />

receptor antagonist eticlopride in the AH results in dose-dependent suppression<br />

of aggression behaviors without causing mobility changes (Schwartzer and<br />

Melloni, 2010a). Conversely, injection of SCH-23390 into the AH reduced<br />

aggressiveness but showed simultaneous changes in sociability and mobility.


7. The Neurochemistry of Human Aggression 159<br />

Postmortem studies revealed sparse population of GAD 67 (a GABA production<br />

marker) neurons distributed within the D5 receptors of the lateral AH. Based on<br />

these findings, the authors conclude that D5 receptors in the lateral AH modulate<br />

non-GABAergic pathways that may indirectly influence aggression behavior.<br />

Future aggression studies should aim to better understand the role of<br />

dopaminergic activity in the hypothalamus and other limbic structures that are<br />

in part physiologically responsible for emotion and behavior regulation.<br />

IV. NOREPINEPHRINE (NORADRENALINE)<br />

Synthesized from tyrosine-derived dopamine via dopamine decarboxylase and<br />

b-hydroxylase (Sofuoglu and Sewell, 2009), norepinephrine (NE) is both a<br />

catecholamine neurotransmitter and a stimulant stress hormone. As a stress<br />

hormone, NE primarily targets brain regions responsible for attention such as<br />

the amygdala and works in conjunction with epinephrine (adrenaline) to produce<br />

the “fight-or-flight” response (Tanaka et al., 2000). During times of high<br />

stress, this response increases heart rate, releases glucose from energy stores, and<br />

increases blood flow to skeletal muscle in an attempt to increase the oxygen<br />

supply to the brain. When released from the locus ceruleus, NE also works to<br />

actively suppress neuroinflammation that may potentially cause damage to the<br />

brain (Heneka et al., 2010).<br />

One of the earliest reports relating aggression to norepinephrine emerged<br />

in a 1972 publication by Thoa and colleagues in Science magazine (Thoa et al.,<br />

1972). In this study, rats that received an intraventricular injection of 90 mg of<br />

6-hydroxydopamine (a neurotoxic agent used to selectively target dopaminergic or<br />

noradrenergic neurons) showed increased shock-induced aggression and reduced<br />

brain norepinephrine while dopamine levels remained unaltered. This inverse<br />

relationship between norepinephrine availability and shock-induced aggression<br />

suggests that the behavioral trait is partially modulated by noradrenergic function.<br />

In the mid-1980s, Pucilowski and colleagues launched a series of studies that<br />

confirmed NE was intimately related to aggression. The first paper, dating from<br />

1985, demonstrated that chemically induced muricide could be in part suppressed<br />

by norepinephrine (Pucilowski and Valzelli, 1985). A second study, published<br />

shortly thereafter, showed that bilateral microinjections of hydroxydopamine into<br />

the nuclei loci coerulei of male Wistar rats resulted in decreased mesencephalic<br />

and striatal norepinephrine levels as well as marked increased aggression<br />

(Pucilowski et al., 1986). In 1987, a similar study by the same group gave microinjections<br />

of the NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine<br />

(DSP-4) with or without apomorphine into the amygdala. Here, the results


160 Yanowitch and Coccaro<br />

clearly showed that NE was able to markedly reduce apomorphine-induced aggression<br />

(Pucilowski et al., 1987). The data also showed that norepinephrine significantly<br />

reduced locomotor activity and to a lesser degree, sensitivity to pain.<br />

In 1998, a novel research experiment authored by Spivak et al. reported<br />

that neuroleptic-resistant chronic schizophrenic patients maintained on clozapine<br />

for 1 year had significantly less aggression (p


7. The Neurochemistry of Human Aggression 161<br />

schizophrenia (Kantrowitz et al., 2009; Wassef et al., 1999), epilepsy (Snodgrass,<br />

1992; Treiman, 2001), and pain and nociception (Enna and McCarson, 2006;<br />

Sawynok, 1984).<br />

In 2007, researchers at the Model Organism Research Center of the<br />

Shanghai Institutes for Biological Sciences found that GABA transporter subtype<br />

1-deficient (or GAT1 / ) mice exhibit lower behavioral aggressiveness compared<br />

to wild-type mice (Coccaro et al., 1998b). Deficiency in GABA transporter function,<br />

analogous to inhibition of GABA transporters, leads to an increase in synaptic<br />

GABA. Later, Takahashi et al. found that pharmacological activation of GABA<br />

(B), but not GABA(A), receptors in the dorsal raphé nucleus significantly increased<br />

aggression (Takahashi et al., 2010). The authors theorized that since the majority of<br />

forebrain 5-HT originates from the raphé nucleus, GABAergic control of this region<br />

could provide an indirect mechanism for escalations in behavioral aggression. A<br />

similar study by the same group showed that male CFW mice, treated with the<br />

GABA(A) receptor agonist muscimol, had increased aggressive tendencies following<br />

alcohol consumption compared to mice given water. These results demonstrate<br />

that GABA(A), but not GABA(B), receptors in the dorsal raphé nucleus are one of<br />

the neurobiological targets of alcohol-induced aggression (Coid et al., 1983), and<br />

illustrate a functional role for GABA in modulating aggressive behavior.<br />

In addition to its affiliations with serotonin, it has also been demonstrated<br />

that GABA is associated with the dopaminergic systems as well and that<br />

this relationship may influence displays of aggression. GABAergic interneurons<br />

in various brain regions including the AH are commonly found to express<br />

dopamine D2 receptors (Gerfen et al., 1990; Santana et al., 2009). Based on<br />

this observation and the known presence of DA D2 receptors in the AH (Ricci<br />

et al., 2009), Schwartzer et al. postulated that DA D2 receptor activity may be<br />

modulating behavioral aggression through direct inhibition of GABA in the AH<br />

(Schwartzer et al., 2009). The authors found that adolescent male Syrian hamsters<br />

exposed to anabolic-androgenic steroids had DA-stimulated increased aggression<br />

marked by the removal of GABAergic inhibition in the lateral AH.<br />

Human studies of GABA and aggression are limited but include two<br />

studies in personality disordered subjects from the laboratory of Coccaro et al. In<br />

the first study, Lee et al. demonstrated a direct relationship between CSF GABA<br />

and measures of impulsivity and history of suicide attempt (but not aggression) in<br />

personality disordered subjects (Lee et al., 2008). In the second study, the growth<br />

hormone (GH) response to the GABA(B) receptor agonist, baclofen, was found<br />

to be inversely correlated with measures of impulsivity (but not aggression) (Lee<br />

et al., in press). Taken together, these studies suggest that elevated central<br />

GABA may lead to, or be associated with, a reduction of GABA(B) receptors<br />

and that this reduction in downstream GABA(B) mediated activity is associated<br />

with increased liability to impulsive behavior. As such, these data are consistent<br />

with the work of Takahashi et al. (2010).


162 Yanowitch and Coccaro<br />

VI. PEPTIDES<br />

Limited published data suggest relationships between human aggression and<br />

central vasopressin, oxytocin, and opiates. (Coccaro et al., 1998b) first reported<br />

a positive correlation between CSF vasopressin concentration and life history of<br />

aggression in male and female subjects with personality disorders. This relationship<br />

was confined to males and remained even after the inverse correlation<br />

between CSF vasopressin and a collateral assessment of serotonin function<br />

(i.e., PRL response to FEN) was accounted for. Later, Lee et al. (2009) reported<br />

an inverse relationship between CSF oxytocin and life history of aggression in an<br />

overlapping group of subjects. CSF vasopressin and CSF oxytocin were inversely<br />

correlated, but CSF oxytocin continued to be related to aggression even after the<br />

influence of CSF vasopressin on aggression was controlled for. This lab has also<br />

noted a positive correlation between CSF Neuropeptide Y and CSF Substance P<br />

in these same subjects. In addition, circulating levels of metenkephalins have<br />

been associated with self-injurious behaviors in one study (Coid et al., 1983).<br />

Postmortem studies of violent suicide victims have found greater number of mu<br />

receptors in the brain. In healthy volunteers, administration of codeine (Spiga<br />

et al., 1990) or morphine (Berman et al., 1993) heightened aggression on<br />

laboratory measures. These studies suggest that increased opioid activity may<br />

increase the likelihood of aggressive behavior. In fact, naltrexone, an opioid<br />

antagonist, attenuates self-injurious behavior in autistic and retarded patients<br />

(Sandman et al., 1990, 2000).<br />

VII. CONCLUSION<br />

The neurobiology of aggression is clearly complex. However, we now know more<br />

about the biological underpinnings of this behavior than ever before and this<br />

knowledge points the way to possible strategies for treatment. Many agents appear<br />

to have therapeutic efficacy but many only work on the brain 5-HT system. In the<br />

upcoming years, we look to the development of agents that work on non-5-HT<br />

systems (e.g., vasopressin, oxytocin, etc.) so that we may have a more varied<br />

toolbox with which to treat individuals with problematic aggressive behavior.<br />

References<br />

Alia-Klein, N., Goldstein, R. Z., Kriplani, A., Logan, J., Tomasi, D., Williams, B., Telang, F., Shumay, E.,<br />

Biegon, A., Craig, I. W., Henn, F., Wang, G. J., Volkow, N. D., and Fowler, J. S. (2008). Brain<br />

monoamine oxidase. A activity predicts trait aggression. J. Neurosci. 28(19), 5099–5104.<br />

Arias-Carrión, O., and Pöppel, E. (2007). Dopamine, learning and reward-seeking behavior. Act.<br />

Neurobiol. Exp. 67(4), 481–488.


7. The Neurochemistry of Human Aggression 163<br />

Asberg, M., Traksman, L., and Thoren, P. (1976). 5-HIAA in the cerebrospinal fluid: A biochemical<br />

suicide predictor. Arch. Gen. Psychiatry 33, 1193–1197.<br />

Berman, M., Taylor, S., and Marged, B. (1993). Morphine and human aggression. Addict. Behav. 18,<br />

263–268.<br />

Birmaher, B., Stanley, M., <strong>Green</strong>hill, L., Twomey, J., Gavrilescu, A., and Rabinovich, H. (1990).<br />

Platelet imipramine binding in children and adolescents with impulsive behavior. J. Am. Acad.<br />

Child Adolesc. Psychiatry 29, 914–918.<br />

Browman, K. E., Curzon, P., Pan, J. B., Molesky, A. L., Komater, V. A., Decker, M. W., Brioni, J. D.,<br />

Moreland, R. B., et al. (2005). A-412997, a selective dopamine D4 agonist, improves cognitive<br />

performance in rats. Pharmacol. Biochem. Behav. 82(1), 148–155.<br />

Brown, A. S., and Gershon, S. (1993). Dopamine and depression. J. Neural Transm. Gen. Sect.<br />

91(2–3), 75–109. Review.<br />

Brown, G. L., Goodwin, F. K., Ballenger, J. C., Goyer, P. F., and Major, L. F. (1979). Aggression in<br />

human correlates with cerebrospinal fluid amine metabolites. Psychiatry Res. 1, 131–139.<br />

Brown, G. L., Ebert, M. H., Goyer, P. F., et al. (1982). Aggression, suicide, and serotonin: Relationships<br />

to CSF amine metabolites. Am. J. Psychiatry 139, 741–746.<br />

Castellanos, F. X., Elia, J., Kreusi, M. J. P., Gulotta, C. S., Mefford, I. N., Potter, W. Z., Ritchie, G. F.,<br />

and Rapoport, J. L. (1994). Cerebrospinal fluid monoamine metabolites in boys with attention<br />

deficit hyperactivity disorder. Psychiatry Res. 52, 305–316.<br />

Cervantes, M. C., and Delville, Y. (2009). Serotonin 5-HT1A and 5-HT3 receptors in an impulsiveaggressive<br />

phenotype. Behav. Neurosci. 123(3), 589–598.<br />

Cervantes, M. C., Biggs, E. A., and Delville, Y. (2010). Differential responses to serotonin receptor<br />

ligands in an impulsive-aggressive phenotype. Behav. Neurosci. 124(4), 455–469.<br />

Coccaro, E. F. (1998). Central neurotransmitter function in human aggression and impulsivity.<br />

In “Neurobiology and Clinical Views on Aggression and Impulsivity” (M. Maes and<br />

E. F. Coccaro, eds.), pp. 143–168. Chichester, Wiley, UK.<br />

Coccaro, E. F., and Kavoussi, R. J. (1994). The neuropsychopharmacologic challenge in biological<br />

psychiatry. Clin. Chem. 40, 319–327.<br />

Coccaro, E. F., and Kavoussi, R. J. (2010). GH response to intravenous clonidine challenge: Absence<br />

of relationship with behavioral irritability, aggression, or impulsivity in human subjects. Psychiatry<br />

Res. 178, 443–445.<br />

Coccaro, E. F., and Lee, R. (2010). Cerebrospinal fluid 5-hydroxyindolacetic acid and homovanillic<br />

acid: Reciprocal relationships with impulsive aggression in human subjects. J. Neural Transm.<br />

117(2), 241–248.<br />

Coccaro, E. F., and Siever, L. J. (2002). Pathophysiology and treatment of aggression.<br />

In “Psychopharmacology: The Fifth Generation of Progress” (K. L. Davis, D. Charney,<br />

J. T. Coyle, and C. Nemeroff, eds.), pp. 1709–1723. Lippincott Williams & Wilkins, Philadelphia.<br />

Coccaro, E. F., Siever, L. J., Klar, H. M., Maurer, G., Cochrane, K., Mohs, R. C., and Davis, K. L.<br />

(1989). Serotonergic studies in affective and personality disorder: Correlates with suicidal and<br />

impulsive aggressive behavior. Arch. Gen. Psychiatry 46, 587–599.<br />

Coccaro, E. F., Lawrence, T., Trestman, R., Gabriel, S., Klar, H. M., and Siever, L. J. (1991). Growth<br />

hormone responses to intravenous clonidine challenge correlate with behavioral irritability in<br />

psychiatric patients and healthy volunteers. Psychiatry Res. 39(2), 129–139.<br />

Coccaro, E. F., Kavoussi, R. J., Sheline, Y. I., Lish, J. D., and Csernansky, J. G. (1996). Impulsive<br />

aggression in personality disorder: Correlates with 3-H-Paroxetine binding in the platelet. Arch.<br />

Gen. Psychiatry 53, 531–536.<br />

Coccaro, E. F., Kavoussi, R. J., Cooper, T. B., and Hauger, R. L. (1997a). Central serotonin activity<br />

and aggression: Inverse relationship with prolactin response to d-fenfluramine, but Not CSF<br />

5-HIAA concentration, in human subjects. Am. J. Psychiatry 154, 1430–1435.


164 Yanowitch and Coccaro<br />

Coccaro, E. F., Kavoussi, R. J., and Hauger, R. L. (1997b). Serotonin function and antiaggressive<br />

responses to fluoxetine: A pilot study. Biol. Psychiatry 42, 546–552.<br />

Coccaro, E. F., Kavoussi, R. J., Cooper, T. B., and Hauger, R. L. (1997c). Central serotonin and<br />

aggression: Inverse relationship with prolactin response to d-fenfluramine, but not with CSF<br />

5-HIAA concentration in human subjects. Am. J. Psychiatry 154, 1430–1435.<br />

Coccaro, E. F., Kavoussi, R. J., Cooper, T. B., and Hauger, R. (1998a). Acute tryptophan depletion<br />

attenuates the prolactin response to d-fenfluramine challenge in healthy human subjects. Psychopharmacology<br />

138, 9–15.<br />

Coccaro, E. F., Kavoussi, R. J., Hauger, R. L., Cooper, T. B., and Ferris, C. F. (1998b). Cerebrospinal<br />

fluid vasopressin: Correlates with aggression and serotonin function in personality disordered<br />

subjects. Arch. Gen. Psychiatry 55, 708–714.<br />

Coccaro, E. F., Lee, R., and McCloskey, M. (2003). Norepinephrine function in personality disorder:<br />

Plasma free MHPG correlates inversely with life history of aggression. CNS Spectr. 8, 731–736.<br />

Coccaro, E. F., Lee, R., and Kavoussi, R. J. (2010a). Aggression, Suicidality, and Intermittent<br />

Explosive Disorder: Serotonergic correlates in personality disorder and healthy control subjects.<br />

Neuropsychopharmacology 35, 435–444.<br />

Coccaro, E. F., Lee, R., and Kavoussi, R. J. (2010b). Inverse relationship between numbers of 5-HT<br />

transporter binding sites and life history of aggression and intermittent explosive disorder.<br />

J. Psychiatr. Res. 44(3), 137–142.<br />

Coid, J., Allolio, B., and Rees, L. H. (1983). Raised plasma metenkephalin in patients who habitually<br />

mutilate themselves. Lancet 2, 545–546.<br />

Couppis, M. H., and Kennedy, C. H. (2008). The rewarding effect of aggression is reduced by nucleus<br />

accumbens dopamine receptor antagonism in mice. Psychopharmacology (Berl.) 197(3), 449–456.<br />

Curzon, G. (1991). Effects of tryptophan and of 5-hydroxytryptamine receptor subtype agonists on<br />

feeding. Adv. Exp. Med. Biol. 294, 377–388.<br />

Daw, N. D., Kakade, S., and Dayan, P. (2002). Opponent interactions between serotonin and<br />

dopamine. Neural Netw. 15, 603–616.<br />

de Almeida, R. M., Ferrari, P. F., Parmigiani, S., and Miczek, K. A. (2005). Escalated aggressive<br />

behavior: Dopamine, serotonin and GABA. Eur. J. Pharmacol. 526, 51–64.<br />

De Simoni, M. G., Dal Toso, G., Fodritto, F., Sokola, A., and Algeri, S. (1987). Modulation of<br />

striatal dopamine metabolism by the activity of dorsal raphe serotonergic afferences. Brain Res.<br />

411, 81–88.<br />

Denenberg, V. H., Kim, D. S., and Palmiter, R. D. (2004). The role of dopamine in learning, memory,<br />

and performance of a water escape task. Behav. Brain Res. 148(1–2), 73–78.<br />

Devos, D., Labyt, E., Derambure, P., Bourriez, J. L., Cassim, F., Guieu, J. D., Destée, A., and<br />

Defebvre, L. (2003). Effect of L-dopa on the pattern of movement-related (de)synchronisation<br />

in advanced Parkinson’s disease. Neurophysiol. Clin. 33(5), 203–212.<br />

Diehl, D. J., and Gershon, S. (1992). The role of dopamine in mood disorders. Compr. Psychiatry<br />

33(2), 115–120. Review.<br />

Dourish, C. T. (1995). Multiple serotonin receptors: Opportunities for new treatments for obesity<br />

Obes. Res. 3(Suppl. 4), 449S–462S.<br />

Dzirasa, K., Ribeiro, S., Costa, R., Santos, L. M., Lin, S. C., Grosmark, A., Sotnikova, T. D.,<br />

Gainetdinov, R. R., Caron, M. G., and Nicolelis, M. A. (2006). Dopaminergic control of sleepwake<br />

states. J. Neurosci. 26(41), 10577–10589.<br />

Engelborghs, S., Vloeberghs, E., Le Bastard, N., Van Buggenhout, M., Mariën, P., Somers, N.,<br />

Nagels, G., Pickut, B. A., and De Deyn, P. P. (2008). The dopaminergic neurotransmitter system<br />

is associated with aggression and agitation in frontotemporal dementia. Neurochem. Int. 52(6),<br />

1052–1060.<br />

Enna, S. J., and McCarson, K. E. (2006). The role of GABA in the mediation and perception of pain.<br />

Adv. Pharmacol. 54, 1–27. Review.


7. The Neurochemistry of Human Aggression 165<br />

Ferris, C. F., Stolberg, T., and Delville, Y. (1999). Serotonin regulation of aggressive behavior in male<br />

golden hamsters Mesocricetus auratus. Behav. Neurosci. 113, 804–815.<br />

Gardner, D. L., Lucas, P. B., and Cowdry, R. W. (1990). CSF metabolites in borderline personality<br />

disorder compared with normal controls. Biol. Psychiatry 28, 247–254.<br />

Gerfen, C. R., Engber, T. M., Mahan, L. C., Susel, Z., Chase, T. N., Monsma, F. J., Jr., et al. (1990).<br />

D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal<br />

neurons. Science 250, 1429–1432.<br />

Gershon, M. D. (2004). Review article: Serotonin receptors and transporters—Roles in normal and<br />

abnormal gastrointestinal motility. Aliment. Pharmacol. Ther. 20(Suppl. 7), 3–14.<br />

Grigorenko, E. L., De Young, C. G., Eastman, M., Getchell, M., Haeffel, G. J., Klinteberg, B.,<br />

Koposov, R. A., Oreland, L., Pakstis, A. J., Ponomarev, O. A., Ruchkin, V. V., Singh, J. P., and<br />

Yrigollen, C. M. (2010). Aggressive behavior, related conduct problems, and variation in genes<br />

affecting dopamine turnover. Aggress. Behav. 36(3), 158–176.<br />

Heijtz, R. D., Kolb, B., and Forssberg, H. (2007). Motor inhibitory role of dopamine D1 receptors:<br />

Implications for ADHD. Physiol. Behav. 92(1–2), 155–160.<br />

Heneka, M. T., Nadrigny, F., Regen, T., Martinez-Hernandez, A., Dumitrescu-Ozimek, L.,<br />

Terwel, D., Jardanhazi-Kurutz, D., Walter, J., Kirchhoff, F., Hanisch, U. K., and Kummer, M. P.<br />

(2010). Locus ceruleus controls Alzheimer’s disease pathology by modulating microglial functions<br />

through norepinephrine. Proc. Natl. Acad. Sci. USA 17, 6058–6063.<br />

Hernandez, L., and Hoebel, B. G. (1988). Food reward and cocaine increase extracellular dopamine<br />

in the nucleus accumbens as measured by microdialysis. Life Sci. 42, 1705–1712.<br />

Hull, E. M., Eaton, R. C., Moses, J., and Lorrain, D. (1993). Copulation increases dopamine activity<br />

in the medial preoptic area of male rats. Life Sci. 52, 935–940.<br />

Joppa, M. A., Rowe, R. K., and Meisel, R. L. (1996). Effects of serotonin 1A or 1B receptor agonists<br />

on social aggression in male and female Syrian hamsters. Pharmacol. Biochem. Behav. 58, 349–353.<br />

Joyce, P. R., McHugh, P. C., McKenzie, J. M., Sullivan, P. F., Mulder, R. T., Luty, S. E., Carter, J. D.,<br />

Frampton, C. M., Robert Cloninger, C., Miller, A. M., and Kennedy, M. A. (2006). A dopamine<br />

transporter polymorphism is a risk factor for borderline personality disorder in depressed patients.<br />

Psychol. Med. 36(6), 807–813.<br />

Kantrowitz, J., Citrome, L., and Javitt, D. (2009). GABA(B) receptors, schizophrenia and sleep<br />

dysfunction: A review of the relationship and its potential clinical and therapeutic implications.<br />

CNS Drugs 23(8), 681–691.<br />

Kapur, S., and Remington, G. (1996). Serotonin-dopamine interaction and its relevance to schizophrenia.<br />

Am. J. Psychiatry 153, 436–476.<br />

Kruesi, M. J. P., Rapoport, J. L., Hamberger, S., Hibbs, E., Potter, W. Z., Lenane, M., and<br />

Brown, G. L. (1990). Cerebrospinal fluid metabolites, aggression, and impulsivity in disruptive<br />

behavior disorders of children and adolescents. Arch. Gen. Psychiatry 47, 419–462.<br />

Kumar, K. K., Tung, S., and Iqbal, J. (2010). Bone loss in anorexia nervosa: Leptin, serotonin, and the<br />

sympathetic nervous system. Ann. N. Y. Acad. Sci. 1211, 51–65.<br />

Kunisato, Y., Okamoto, Y., Okada, G., Aoyama, S., Demoto, Y., Munakata, A., Nomura, M.,<br />

Onoda, K., and Yamawaki, S. (2010). Modulation of default-mode network activity by acute<br />

tryptophan depletion is associated with mood change: A resting state functional magnetic<br />

resonance imaging study. Neurosci. Res. 69, 129–134.<br />

Lam, D. D., Garfield, A. S., Marston, O. J., Shaw, J., and Heisler, L. K. (2010). Brain serotonin system<br />

in the coordination of food intake and body weight. Pharmacol. Biochem. Behav. 97(1), 84–91.<br />

Lee, R., Petty, F., and Coccaro, E. F. (2008). Cerebrospinal fluid GABA concentration: Relationship<br />

with impulsivity and history of suicidal behavior, but not aggression, in human subjects.<br />

J. Psychiatric. Res. 43, 353–359.<br />

Lee, R., Ferris, C., Van de Kar, L. D., and Coccaro, E. F. (2009). Cerebrospinal fluid oxytocin, life<br />

history of aggression, and personality disorder. Psychoneuroendocrinology 34, 1567–1573.


166 Yanowitch and Coccaro<br />

Lee, R., Chong, B., and Coccaro, E. F. (2011). Growth hormone responses to GABA-B receptor<br />

challenge with baclofen and impulsivity in healthy control and personality disorder subjects.<br />

Psychopharmacology (Berl) 215(1), 41–48.<br />

Lesch, K. P., Wolozin, B. L., Murphy, D. L., and Reiderer, P. (1993). Primary structure of the human<br />

platelet serotonin uptake site: Identity with the brain serotonin transporter. J. Neurochem. 60,<br />

2319–2322.<br />

Lidberg, L., Tuck, J. R., Asberg, M., Scalia-Tomba, G. P., and Bertilsson, L. (1985). Homicide,<br />

suicide and CSF 5-HIAA. Acta Psych. Scand. 71, 230–236.<br />

Lima, M. M., Andersen, M. L., Reksidler, A. B., Silva, A., Zager, A., Zanata, S. M., Vital, M. A., and<br />

Tufik, S. (2008). Blockage of dopaminergic D(2) receptors produces decrease of REM but not of<br />

slow wave sleep in rats after REM sleep deprivation. Behav. Brain Res. 188(2), 406–411.<br />

Limson, R., Goldman, D., Roy, A., Lamparski, D., Ravitz, B., Adinoff, B., and Linnoila, M. (1991).<br />

Personality and cerebrospinal fluid monoamine metabolites in alcoholics and controls. Arch. Gen.<br />

Psychiatry 48, 437–441.<br />

Linnoila, M., Virkkunen, M., Scheinin, M., Nuutila, A., Rimon, R., and Goodwin, F. K. (1983).<br />

Low cerebrospinal fluid 5-hydroxylndolacetic acid concentration differentiates impulsive from<br />

nonimpulsive violent behavior. Life Sci. 33, 2609–2614.<br />

Meyer, J. H., Wilson, A. A., Rusjan, P., Clark, M., Houle, S., Woodside, S., Arrowood, J., Martin, K.,<br />

and Colleton, M. (2008). Serotonin2A receptor-binding potential in people with aggressive and<br />

violent behaviour. J. Psychiatry Neurosci. 33(6), 499–508.<br />

Miczek, K. A., Faccidomo, S., de Almeida, R. M. M., Bannai, M., Fish, E. W., and DeBold, J. F.<br />

(2004). Escalated aggressive behavior: New pharmacotherapeutic approaches and opportunities.<br />

Ann. N. Y. Acad. Sci. 1036, 336–355.<br />

Moller, S. E., Mortensen, E. L., Breum, L., Alling, C., Larsen, O. G., Boge-Rasmussen, T., Jensen, C.,<br />

and Bennicke, K. (1996). Aggression and personality: Association with amino acids and monoamine<br />

metabolites. Psychol. Med. 26, 323–331.<br />

Monti, J. M. (2010). The role of dorsal raphe nucleus serotonergic and non-serotonergic neurons, and<br />

of their receptors, in regulating waking and rapid eye movement (REM) sleep. Sleep Med. Rev.<br />

14(5), 319–327.<br />

Monti, J. M., and Jantos, H. (2008). The roles of dopamine and serotonin, and of their receptors, in<br />

regulating sleep and waking. Prog. Brain Res. 172, 625–646.<br />

Monti, J. M., and Monti, D. (2000). Role of dorsal raphe nucleus serotonin 5-HT1A receptor in the<br />

regulation of REM sleep. Life Sci. 66(21), 1999–2012.<br />

Munafò, M. R., Yalcin, B., Willis-Owen, S. A., and Flint, J. (2008). Association of the dopamine D4<br />

receptor (DRD4) gene and approach-related personality traits: Meta-analysis and new data. Biol.<br />

Psychiatry 63(2), 197–206.<br />

Naheed, M., and <strong>Green</strong>, B. (2001). Focus on clozapine. Curr. Med. Res. Opin. 17(3), 223–229.<br />

Nemoda, Z., Lyons-Ruth, K., Szekely, A., Bertha, E., Faludi, G., and Sasvari-Szekely, M. (2010).<br />

Association between dopaminergic polymorphisms and borderline personality traits among at-risk<br />

young adults and psychiatric inpatients. Behav. Brain Funct. 6, 4.<br />

New, A. S., Hazlett, E. A., Buchsbaum, M. S., Goodman, M., Reynolds, D., Mitropoulou, V.,<br />

Sprung, L., Shaw, R. B., Jr., Koenigsberg, H., Platholi, J., Silverman, J., and Siever, L. J. (2002).<br />

Blunted prefrontal cortical 18fluorodeoxyglucose positron emission tomography response to metachlorophenylpiperazine<br />

in impulsive aggression. Arch. Gen. Psychiatry 59(7), 621–629.<br />

Nieoullon, A. (2002). Dopamine and the regulation of cognition and attention. Prog. Neurobiol.<br />

67(1), 53–83. Review.<br />

Oak, J. N., Oldenhof, J., and Van Tol, H. H. (2000). The dopamine D(4) receptor: One decade of<br />

research. Eur. J. Pharmacol. 405(1–3), 303–327. Review.<br />

Olivier, B., Mos, J., van Oorschot, R., and Hen, R. (1995). Serotonin receptors and animal models of<br />

aggressive behavior. Pharmacopsychiatry 28, 80–90.


7. The Neurochemistry of Human Aggression 167<br />

Parsey, R. V., Oquendo, M. A., Simpson, N. R., Ogden, R. T., Van Heertum, R., Arango, V., and<br />

Mann, J. J. (2002). Effects of sex, age, and aggressive traits in man on brain serotonin 5-HT1A<br />

receptor-binding potential measured by PET using [C-11]WAY-100635. Brain Res. 954, 173–182.<br />

Peddeer, J. (1992). Psychoanalytic views of aggression: Some theoretical problems. Br. J. Med.<br />

Psychol. 65, 95–106.<br />

Pettit, H. O., and Justice, J. B. (1991). Effect of dose on cocaine self-administration behavior and<br />

dopamine levels in the nucleus accumbens. Brain Res. 539, 94–102.<br />

Pfaus, J. G., Damsma, G., Nomikos, G. G., Wenkstern, D. G., Blaha, C. D., Phillips, A. G., and<br />

Fibiger, H. C. (1990). Sexual behavior enhances central dopamine transmission in the male rat.<br />

Brain Res. 530, 345–348.<br />

Pihl, R.-O., Young, S., Harden, P., Plotnick, S., Chamberlain, B., and Ervin, F. R. (1995). Acute<br />

effect of altered tryptophan levels and alcohol on aggression in normal human males. Psychopharmacology<br />

119, 353–360.<br />

Popova, N. K. (2008). From gene to aggressive behavior: The role of brain serotonin. Neurosci.<br />

Behav. Physiol. 38(5), 471–475.<br />

Popova, N. K., Naumenko, V. S., Plyusnina, I. Z., and Kulikov, A. V. (2005). Reduction in 5-HT1A<br />

receptor density, 5-HT1A mRNA expression, and functional correlates for 5-HT1A receptors in<br />

genetically defined aggressive rats. J. Neurosci. Res. 80, 286–292.<br />

Popova, N. K., Naumenko, V. S., and Plyusnina, I. Z. (2007). Involvement of brain serotonin 5-<br />

HT1A receptors in genetic predisposition to aggressive behavior. Neurosci. Behav. Physiol. 37,<br />

631–635.<br />

Prochazka, H., and Agren, H. (2003). Self-rated aggression and cerebral monoaminergic turnover.<br />

Sex differences in patients with persistent depressive disorder. Eur. Arch Psychiatry Clin. Neurosci.<br />

253(4), 185–192.<br />

Pucilowski, O., and Valzelli, L. (1985). Evidence of norepinephrine-mediated suppression of parachlorophenylalanine-induced<br />

muricidal behavior. Pharmacol. Res. Commun. 17, 983–989.<br />

Pucilowski, O., Kozak, W., and Valzelli, L. (1986). Effect of 6-OHDA injected into the locus<br />

coeruleus on apomorphine-induced aggression. Pharmacol. Biochem. Behav. 24, 773–775.<br />

Pucilowski, O., Trzaskowska, E., Kostowski, W., and Valzelli, L. (1987). Norepinephrine-mediated<br />

suppression of apomorphine-induced aggression and locomotor activity in the rat amygdala.<br />

Pharmacol. Biochem. Behav. 26(2), 217–222.<br />

Ranaldi, R., Pocock, D., Zereik, R., and Wise, R. A. (1999). Dopamine fluctuations in the nucleus<br />

accumbens during maintenance, extinction, and reinstatement of intravenous d-amphetamine<br />

self-administration. J. Neurosci. 19, 4102–4109.<br />

Ricci, L. A., Rasakham, K., Grimes, J. M., and Melloni, R. H., Jr. (2006). Serotonin-1A receptor<br />

activity and expression modulate adolescent anabolic/androgenic steroid-induced aggression in<br />

hamsters. Pharmacol. Biochem. Behav. 85, 1–11.<br />

Ricci, L. A., Schwartzer, J. J., and Melloni, R. H., Jr. (2009). Alterations in the anterior hypothalamic<br />

dopamine system in aggressive adolescent AAS-treated hamsters. Horm. Behav. 55, 348–355.<br />

Rosell, D. R., Thompson, J. L., Slifstein, M., Xu, X., Frankle, W. G., New, A. S., Goodman, M.,<br />

Weinstein, S. R., Laruelle, M., Abi-Dargham, A., and Siever, L. J. (2010). Increased serotonin 2A<br />

receptor availability in the orbitofrontal cortex of physically aggressive personality disordered<br />

patients. Biol. Psychiatry 67(12), 1154–1162.<br />

Ruhé, H. G., Mason, N. S., and Schene, A. H. (2007). Mood is indirectly related to serotonin,<br />

norepinephrine and dopamine levels in humans: A meta-analysis of monoamine depletion studies.<br />

Mol. Psychiatry 12(4), 331–359.<br />

Sandman, C. A., Barron, J. L., and Colman, H. (1990). An orally administered opiate blocker,<br />

naltrexone, attenuates self-injurious behavior. Am. J. Ment. Retard. 95, 93–102.


168 Yanowitch and Coccaro<br />

Sandman, C. A., Hedrick, W., Taylor, D. V., Marion, S. D., Touchette, P., Barron, J. L.,<br />

Martinezzi, V., Steinberg, R. M., and Crinella, F. M. (2000). Long-term effects of naltrexone on<br />

self-injurious behavior. Am. J. Ment. Retard. 105, 103–117.<br />

Santana, N., Mengod, G., and Artigas, F. (2009). Quantitative analysis of the expression of<br />

dopamine D1 and D2 receptors in pyramidal and GABAergic neurons of the rat prefrontal cortex.<br />

Cereb. Cortex 19, 849–860.<br />

Sawynok, J. (1984). GABAergic mechanisms in antinociception. Prog. Neuropsychopharmacol. Biol.<br />

Psychiatry 8(4–6), 581–586.<br />

Schwartzer, J. J., and Melloni, R. H., Jr. (2010a). Dopamine activity in the lateral anterior hypothalamus<br />

modulates AAS-induced aggression through D2 but not D5 receptors. Behav. Neurosci.<br />

124(5), 645–655.<br />

Schwartzer, J. J., and Melloni, R. H., Jr. (2010b). Anterior hypothalamic dopamine D2 receptors<br />

modulate adolescent anabolic/androgenic steroid-induced offensive aggression in the Syrian<br />

hamster. Behav. Pharmacol. 21(4), 314–322.<br />

Schwartzer, J. J., Ricci, L. A., and Melloni, R. H., Jr. (2009). Interactions between the dopaminergic<br />

and GABAergic neural systems in the lateral anterior hypothalamus of aggressive AAS-treated<br />

hamsters. Behav. Brain Res. 203(1), 15–22.<br />

Seo, D., Patrick, C. J., and Kennealy, P. J. (2008). Role of serotonin and dopamine system interactions<br />

in the neurobiology of impulsive aggression and its comorbidity with other clinical disorders.<br />

Aggress Violent Behav. 13(5), 383–395.<br />

Sheard, M., Manini, J., Bridges, C., and Wapner, A. (1976). The effect of lithium on impulsive<br />

aggressive behavior in man. Am. J. Psychiatry 133, 1409–1413.<br />

Siever, L. J. (2008). Neurobiology of aggression and violence. Am. J. Psychiatry 165(4), 429–442.<br />

Siever, L. J., and Davis, K. L. (1991). A psychobiological perspective on the personality disorders.<br />

Am. J. Psychiatry 148(12), 1647–1658. Review.<br />

Siever, L. J., Buchsbaum, M. S., New, A. S., Spiegel-Cohen, J., Wei, C. T., Hazlett, E., Sevin, E.,<br />

Nunn, M., and Mitropoulou, V. (1999). D1-Fenfluramine response in impulsive personality<br />

disorder assessed with 18F-deoxyglucose positron emission tomography. Neuropsychopharmacology<br />

20, 413–423.<br />

Snodgrass, S. R. (1992). GABA and epilepsy: Their complex relationship and the evolution of our<br />

understanding. J. Child Neurol. 7(1), 77–86. Review.<br />

Sofuoglu, M., and Sewell, R. A. (2009). Norepinephrine and stimulant addiction. Addict. Biol. 14(2),<br />

119–129. Review.<br />

Soloff, P. H., Price, J. C., Mason, N. S., Becker, C., and Meltzer, C. C. (2010). Gender, personality,<br />

and serotonin-2A receptor-binding in healthy subjects. Psychiatry Res. 181(1), 77–84.<br />

Spiga, R., Cherek, D. R., Roache, J. D., and Cowan, K. A. (1990). The effects of codeine on human<br />

aggressive responding. Int. Clin. Psychopharmacol. 5, 195–204.<br />

Spivak, B., Roitman, S., Vered, Y., Mester, R., Graff, E., Talmon, Y., Guy, N., Gonen, N., and<br />

Weizman, A. (1998). Diminished suicidal and aggressive behavior, high plasma norepinephrine<br />

levels, and serum triglyceride levels in chronic neuroleptic-resistant schizophrenic patients maintained<br />

on clozapine. Clin. Neuropharmacol. 21(4), 245–250.<br />

Stanley, M., and Mann, J. J. (1983). Increased serotonin—2 binding sites in frontal cortex of suicide<br />

victims. Lancet 1, 214–216.<br />

Stanley, M. S., Viggilio, J., and Gershon, S. (1982). Tritiated imipramine binding sites are decreased<br />

in the frontal cortex of suicides. Science 216, 1337–1339.<br />

Tadić, A., Victor, A., Başkaya, O., von Cube, R., Hoch, J., Kouti, I., Anicker, N. J., Höppner, W.,<br />

Lieb, K., and Dahmen, N. (2009). Interaction between gene variants of the serotonin transporter<br />

promoter region (5-HTTLPR) and catechol O-methyltransferase (COMT) in borderline personality<br />

disorder. Am. J. Med. Genet. B. 150, 487–495.


7. The Neurochemistry of Human Aggression 169<br />

Takahashi, A., Shimamoto, A., Boyson, C. O., DeBold, J. F., and Miczek, K. A. (2010). GABA(B)<br />

receptor modulation of serotonin neurons in the dorsal raphé nucleus and escalation of aggression<br />

in mice. J. Neurosci. 30(35), 11771–11780.<br />

Takahashi, A., Quadros, I. M., de Almeida, R. M., and Miczek, K. A. (2011). Brain serotonin<br />

receptors and transporters: Initiation vs. termination of escalated aggression. Psychopharmacology<br />

(Berl.) 213(2–3), 183–212.<br />

Tanaka, M., Yoshida, M., Emoto, H., and Ishii, H. (2000). Noradrenaline systems in the hypothalamus,<br />

amygdala and locus coeruleus are involved in the provocation of anxiety: Basic studies. Eur.<br />

J. Pharmacol. 405(1–3), 397–406. Review.<br />

Thoa, N. B., Eichelman, B., Richardson, J. S., and Jacobowitz, D. (1972). 6-Hydroxydopa depletion<br />

of brain norepinephrine and the function of aggressive behavior. Science 178(56), 75–77.<br />

Treiman, D. M. (2001). GABA ergic mechanisms in epilepsy. Epilepsia 42(Suppl. 3), 8–12. Review.<br />

Van Erp, A. M. M., and Miczek, K. A. (2000). Aggressive behavior, increased accumbal dopamine,<br />

and decreased cortical serotonin in rats. J. Neurosci. 20(24), 9320–9325.<br />

Wassef, A. A., Dott, S. G., Harris, A., Brown, A., O’Boyle, M., Meyer, W. J., 3rd, and Rose, R. M.<br />

(1999). Critical review of GABA-ergic drugs in the treatment of schizophrenia. J. Clin. Psychopharmacol.<br />

19(3), 222–232. Review.<br />

Weiss, F., Paulus, M., Lorang, M. T., and Koob, G. F. (1992). Increases in extracellular dopamine in<br />

the nucleus accumbens by cocaine are inversely related to basal levels: Effects of acute and<br />

repeated administration. J. Neurosci. 12, 4372–4380.<br />

Welch, J. T., and Lim, D. S. (2007). The synthesis and biological activity of pentafluorosulfanyl<br />

analogs of fluoxetine, fenfluramine, and norfenfluramine. Bioorg. Med. Chem. 15(21), 6659–6666.<br />

White, S. M., Kucharik, R. F., and Moyer, J. A. (1991). Effects of serotonergic agents on isolationinduced<br />

aggression. Pharmacol. Biochem. Behav. 39, 729–736.<br />

Wise, R. A. (2004). Dopamine, learning, and motivation. Nat. Rev. Neurosci. 5, 483–495.<br />

Witte, A. V., Flöel, A., Stein, P., Savli, M., Mien, L. K., Wadsak, W., Spindelegger, C., Moser, U.,<br />

Fink, M., Hahn, A., Mitterhauser, M., Kletter, K., Kasper, S., and Lanzenberger, R. (2009).<br />

Aggression is related to frontal serotonin-1A receptor distribution as revealed by PET in healthy<br />

subjects. Hum. Brain Mapp. 30(8), 2558–2570. Erratum in: Hum. Brain Mapp. 2010 Feb; 31(2):<br />

339.<br />

Wong, P. T., Feng, H., and Teo, W. L. (1995). Interaction of the dopaminergic and serotonergic<br />

systems in the rat striatum: Effects of selective antagonists and uptake inhibitors. Neurosci. Res.<br />

23, 115–119.<br />

Xu, L., Yu, B. P., Chen, J. G., and Luo, H. S. (2007). Mechanisms mediating serotonin-induced<br />

contraction of colonic myocytes. Clin. Exp. Pharmacol. Physiol. 34(1–2), 120–128.


Intentionally left as blank


8<br />

Human Aggression Across the<br />

Lifespan: Genetic Propensities<br />

and Environmental Moderators<br />

Catherine Tuvblad and Laura A. Baker<br />

<strong>University</strong> of Southern California, Los Angeles, California, USA<br />

I. Heritability of Aggression: Twin and Adoption Studies<br />

A. Does heritability vary depending on sex<br />

B. Does heritability change across age<br />

C. Do heritabilities vary across methods of assessment<br />

D. Do heritabilities vary across forms of aggression<br />

E. Does heritability vary depending on study design (twins vs.<br />

adopted siblings)<br />

F. Criticisms of twin and adoption studies: Assumptions and<br />

generalizability<br />

II. G E Interaction in Aggressive Behavior<br />

A. Potential moderators of genetic influence found in adoption<br />

and twin studies<br />

III. Specific Genes for Aggressive Behavior: Findings from Molecular<br />

Genetic Studies<br />

A. G E interaction involving specific genes for aggressive<br />

behavior<br />

IV. Conclusions<br />

References<br />

ABSTRACT<br />

This chapter reviews the recent evidence of genetic and environmental influences<br />

on human aggression. Findings from a large selection of the twin and adoption<br />

studies that have investigated the genetic and environmental architecture of<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00007-1


172 Tuvblad and Baker<br />

aggressive behavior are summarized. These studies together show that about half<br />

(50%) of the variance in aggressive behavior is explained by genetic influences in<br />

both males and females, with the remaining 50% of the variance being explained<br />

by environmental factors not shared by family members. Form of aggression<br />

(reactive, proactive, direct/physical, indirect/relational), method of assessment<br />

(laboratory observation, self-report, ratings by parents and teachers), and age of<br />

the subjects—all seem to be significant moderators of the magnitude of genetic<br />

and environmental influences on aggressive behavior. Neither study design (twin<br />

vs. sibling adoption design) nor sex (male vs. female) seems to impact the magnitude<br />

of the genetic and environmental influences on aggression. There is also some<br />

evidence of gene-environment interaction (G E) from both twin/adoption<br />

studies and molecular genetic studies. Various measures of family adversity and<br />

social disadvantage have been found to moderate genetic influences on aggressive<br />

behavior. Findings from these G E studies suggest that not all individuals will be<br />

affected to the same degree by experiences and exposures, and that genetic predispositions<br />

may have different effects depending on the environment. ß 2011, Elsevier Inc.<br />

Are all humans innately and equally capable of inflicting harm on others Do we<br />

learn by our various experiences to manipulate and even harm others for our own<br />

personal gain; or conversely, to be kind and benevolent, offering help even at<br />

costs to ourselves Although these fundamental questions pertaining to the<br />

nature of human aggression have plagued scientists and laypersons for centuries,<br />

some answers can be found in research spanning the last few decades.<br />

The early experiments of Milgram (1963) made it clear that, under<br />

certain circumstances, individuals can be coaxed into aggression and violence.<br />

The presence of a strict authority and removal of personal responsibility for one’s<br />

actions can result in aggressive behaviors that inflict harm on others. The<br />

infamous Stanford prison experiment (Haney et al., 1973) also demonstrated<br />

that the propensity toward violence and aggression can be elicited—extremely<br />

and unexpectedly—in situations, where a legitimized ideology and a powerful<br />

authority can lead to impressionability and obedience.<br />

Yet, while these powerful studies revealed the importance of social<br />

factors in inducing aggressive behaviors, not all individuals responded in an<br />

equally aggressive manner. In Milgram’s (1963) first set of experiments, while<br />

65% (26 of 40) of participants complied with the instruction to administer what<br />

they believed to be a final, massive 450-volt shock, the remaining 35% did not<br />

comply. Many of those who engaged in the aggressive behavior stated they were<br />

very uncomfortable doing so, and every participant reportedly questioned the<br />

experiment at some point or refused money promised for their study participation<br />

(Milgram, 1963). Although the studies by Milgram and Zimbardo provide clear<br />

evidence for the role of environment and social situations in affecting aggressive<br />

behavior, there are, nonetheless, large individual differences in the propensity for<br />

violence and aggression, even under these extreme circumstances.


8. Human Aggression Across the Lifespan 173<br />

What factors contribute to individual differences in aggression Behavioral<br />

genetic studies of family members’ resemblance for aggressive behavior help<br />

shed light on the matter. Twin and adoption studies agree with the experimental<br />

literature on aggression, which shows that a large effect of environmental factors<br />

is evident, particularly of the nonshared variety. Yet, there is also plenty of<br />

evidence, based on a variety of definitions of aggressive behavior from children<br />

to adults, for genetic propensity toward aggression (see reviews by Burt, 2009;<br />

Miles and Carey, 1997; Rhee and Waldman, 2002). Although few behavioral<br />

genetic studies have explicitly examined the question of gene by environment<br />

(GE) interactions, we contend that such interactions are likely to exist and<br />

that the genetic propensity for aggression should exert its effects more strongly in<br />

some situations than others. Consistent with the early findings of Milgram and<br />

Zimbardo, individual genetic predispositions should moderate the extent to<br />

which aggression can be elicited, even in extreme situations such as these<br />

infamous studies. Our view is that while many, if not most, humans may have<br />

the potential for aggression and violence under the right circumstances, not all<br />

individuals will succumb to these behaviors under the same circumstances.<br />

This chapter will review recent evidence of genetic and environmental<br />

influences on human aggression, with particular attention to several key<br />

questions and issues. We first consider how estimates of the relative importance<br />

of genetic effects (i.e., heritability) may vary across forms of aggression and<br />

the way in which it is measured. As detailed in other chapters of this volume,<br />

there are numerous definitions of aggression. Some definitions distinguish between<br />

reactive and proactive forms (Dodge et al., 1997; Raine et al., 2006), and<br />

others consider direct and indirect forms of aggression (e.g., physical vs. relational;<br />

Lahey et al., 2004; Tackett et al., 2009). Some definitions may include<br />

extreme criminal violence, such as assault, rape, and murder, although these<br />

extreme behaviors are relatively rare and have not been studied extensively in<br />

genetically informative designs. Measures of aggression can include self-reporting,<br />

teacher and parent reports (particularly for young children), and official<br />

records from schools or the justice system. This review focuses on twin and<br />

sibling adoption studies of aggressive behavior measured as a trait within the<br />

wider population. We compare effect sizes (heritability) across these various<br />

definitions and ways of measuring aggression. We also consider how heritability<br />

estimates may vary across both age and gender. Given higher levels of aggression<br />

in males across the lifespan, one obvious question concerns whether genetic<br />

propensities are of greater importance in one sex and how these differences might<br />

vary across age. We consider a variety of measurable environmental factors that<br />

might moderate these genetic influences and which may thus lead to GE<br />

interactions for aggressive behavior. Although direct tests of GE interactions<br />

have been relatively rare in the behavioral genetic literature on human aggression,<br />

it is likely that such interactions exist, given their robust effects in other forms of<br />

antisocial behavior (e.g., property criminal offending; Cloninger et al., 1982).


174 Tuvblad and Baker<br />

Finally, we briefly review evidence for specific genetic influences in aggression by<br />

summarizing some of the more recent findings from molecular genetic studies.<br />

These effects are reviewed in detail elsewhere in this volume, so our focus here is on<br />

how a few specific genes may be involved in GE interactions.<br />

I. HERITABILITY OF AGGRESSION: TWIN AND ADOPTION STUDIES<br />

Behavioral genetic research relies on the different levels of genetic relatedness<br />

between family members in order to estimate the relative contribution of heritable<br />

and environmental factors to individual differences in a phenotype of interest.<br />

Major research designs include: (a) studies of twins raised together and (b) studies of<br />

adopted individuals and their biological and adoptive family members. Although<br />

designs combining both approaches are the most powerful for separating genetic<br />

and environmental effects in human behavior, such studies of twins separated at<br />

birth and raised apart are rare and have not studied aggressive behavior extensively.<br />

Nonetheless, there are a handful of adoption studies and over two dozen studies of<br />

twins raised together which have specifically examined the genetic and environmental<br />

influence in aggression in nonselected samples from Northern America and<br />

Europe that are reasonably representative of the general population.<br />

In the classical twin design, monozygotic (identical) twins share their<br />

common environment and they are assumed to share 100% of their genes.<br />

Dizygotic (fraternal) twins also share their common environment and they are<br />

assumed to share on average 50% of their genes. By comparing the resemblance<br />

for aggressive behavior between monozygotic and dizygotic twins, the total<br />

phenotypic variance of aggression can be divided into additive genetic factors<br />

(or heritability, h 2 ), shared environmental factors (c 2 ), and nonshared environmental<br />

factors (e 2 ). Shared environmental factors refer to nongenetic influences<br />

that contribute to similarity within pairs of twins. Nonshared environmental<br />

factors are those individual experiences that cause siblings to differ in their levels<br />

of aggressive behavior. Heritability is the proportion of total phenotypic variance<br />

due to genetic variation (Neale and Cardon, 1992). Genetic influences may also<br />

be divided into those that are additive (i.e., allelic effects add up across loci) and<br />

those that are nonadditive (i.e., due to dominance or epistasis). In twin studies,<br />

however, it is not possible to estimate both additive and nonadditive genetic<br />

effects (d 2 ) simultaneously with shared twin environment effects. The twin<br />

correlations summarized in Table 8.2 can be used to estimate the genetic and<br />

environmental influences to aggressive behavior. Twice the difference between<br />

the MZ and DZ correlations provides an estimate of the relative contribution of<br />

additive genetic influences to aggressive behavior [h 2 ¼ 2(r MZ r DZ )]. The<br />

contribution of the nonadditive genetic effects due to dominance or epistasis<br />

(d 2 ) is obtained by subtracting four times the DZ correlation from twice the MZ


8. Human Aggression Across the Lifespan 175<br />

correlation (d 2 ¼ 2r MZ 4r DZ ). The proportion of the variance that is due to<br />

shared environmental influence is given by subtracting the MZ correlation from<br />

twice the DZ correlation (c 2 ¼ 2r DZ r MZ ). Finally, the contribution of the nonshared<br />

environmental influences can be obtained by subtracting the MZ correlation<br />

from unit correlation (e 2 ¼ 1 r MZ )(Posthuma et al., 2003). Many twin<br />

studies do not specifically examine or test for nonadditive genetic effects and<br />

instead report heritability estimates based on additive effects only. However,<br />

some twin studies compare models with additive effects and nonadditive effects<br />

versus models with additive genetic effects and shared environment.<br />

In sibling adoption studies, the correlation between adoptive siblings is<br />

compared with the correlation between biological siblings to estimate the influence<br />

of genetic and environmental factors on aggressive behavior (Plomin et al.,<br />

2001). Resemblance between adoptive siblings for measures of aggression is<br />

indicative of shared (or common) family environment, while the extent to<br />

which biological sibling resemblance exceeds that of adoptive siblings is taken<br />

as evidence of heritable genetic influences for aggressive behavior.<br />

There have been a few meta-analyses of twin and adoption studies of<br />

aggressive behavior and the wider construct of antisocial behavior. In one early<br />

meta-analysis of 24 twin and adoption studies, heritable influences explained<br />

about half of the total variance in aggressive behavior and the nonshared environment<br />

explained the remaining 50% (Miles and Carey, 1997). Rhee and Waldman<br />

(2002) also summarized the results from 51 twin and adoption studies on criminal<br />

behavior, delinquency, psychopathy, conduct disorder, and antisocial personality<br />

disorder, as well as aggressive behavior, in children, adolescents, and adults.<br />

Genetic factors explained 41% of the variance in antisocial behavior, 16% was<br />

explained by shared environmental influences, and the remaining 43% of variance<br />

was explained by nonshared environmental factors. A more recent review focused<br />

on 19 twin and adoption studies using child and adolescent samples; studies<br />

including adult subjects were excluded. Heritability was found to explain 65%,<br />

shared environment explained 5%, and the nonshared environment explained the<br />

remaining 30% of the variance in aggressive behavior (Burt, 2009). Both Burt<br />

(2009) and Rhee and Waldman (2002) examined nonadditive genetic effects, but<br />

only Rhee and Waldman (2002) found significant nonadditive genetic effects for<br />

antisocial behavior. It is noteworthy that genetic influences are consistently found<br />

across these reviews, while shared environmental influences are comparatively<br />

small or nonexistent. Family similarity in aggressive and antisocial behavior,<br />

therefore, is primarily the result of shared genes, not environment.<br />

Tables 8.1 and 8.2, respectively, summarize a large selection of twin and<br />

sibling adoption studies which have specifically examined the genetic and<br />

environmental influences on aggressive behavior in child, adolescent, and<br />

adult samples. Several studies use prospective, longitudinal designs, and large<br />

samples, and three of the twin studies were designed, in particular, to study


Table 8.1. Effect Sizes for Aggressive Behavior from Adoption Studies<br />

Study<br />

(author, year)<br />

Aggression<br />

measure Informant Age in years Sex<br />

Biological<br />

siblings<br />

r(N)<br />

Adoptive<br />

siblings<br />

r(N) h 2 c 2<br />

Dutch adoptees<br />

(van den Oord et al., 1994)<br />

Dutch adoptees<br />

(van der Valk et al., 1998)<br />

Colorado adoptees<br />

(Deater-Deckard and<br />

Plomin, 1999)<br />

Unknown<br />

(Parker, 1989) a<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(TRF)<br />

Aggression<br />

(as reported in Rhee<br />

and Waldman,<br />

2002; Burt, 2009)<br />

Parent ratings 10–15<br />

Mean¼12.4<br />

M<br />

F<br />

MF<br />

0.40 (30)<br />

0.45 (35)<br />

0.38 (46)<br />

0.02 (44)<br />

0.21 (48)<br />

0.05 (129)<br />

0.52<br />

0.32<br />

0.00<br />

0.25<br />

Parent ratings 10–15 MþF 0.42 (111) 0.13 (221) 0.61 0.13<br />

13–18 MþF 0.36 (152) 0.26 (156) 0.52 0.12<br />

Parent ratings 7, 9, 10, 11, 12 MþF 0.39 (94) 0.26 (78) 0.24 0.27<br />

Mean¼9.5<br />

Teacher ratings MþF 0.25 (188) 0.06 (156) 0.49 0.00<br />

Parent ratings 4–7 MþF 0.44 (66) 0.47 (45)<br />

M, Male; F, Female; h 2 , heritability; c 2 , shared environment; CBCL, Child Behavior Checklist (Achenbach, 1991b); TRF, Teacher Report Form<br />

(Achenbach, 1991a).<br />

a Genetic and shared environmental estimates were not reported by the authors.


Table 8.2. Effect Sizes (Correlations) for Aggressive Behavior from Twin Studies<br />

Study sample<br />

(author, year)<br />

Aggression measure<br />

Assessment<br />

method Age in years Sex<br />

MZ<br />

r(N)<br />

DZ<br />

r(N) h 2 c 2 Sex limitation effects<br />

Boston twins<br />

Aggression<br />

Parent ratings 6–10 F 0.35 (24) 0.08 (28) 0.40 a – N/A<br />

(Scarr, 1966)<br />

(ACL)<br />

Missouri twins<br />

Aggressive reaction Lab observation 6–14 M 0.09 (10) 0.24 (11) 0.44 a – Not tested<br />

(Owen and Sines,<br />

1970)<br />

(MCPS)<br />

F 0.58 (8) 0.22 (13)<br />

California twins Aggression<br />

Self-report 42–56 M 0.31 (93) 0.21 (97) 0.56 a – N/A<br />

(Rahe et al., 1978) (ACL)<br />

Mean¼48<br />

Colorado twins<br />

Aggression/bullying Parent ratings Mean¼7.6 M þF 0.72 (52) 0.42 (32) – – Not tested<br />

(O’Connor et al., 1980) (PSR)<br />

London twins, UK Aggression<br />

Self-report 19–60 MþF 0.40 (296) 0.04 (179) 0.72 a Not tested<br />

(Rushton et al., 1986) (IBS)<br />

Mean¼30<br />

California preschool Aggression<br />

Parent ratings Mean¼5.2 M þF 0.78 (21) 0.31 (17) 0.94 a – Not tested<br />

twins<br />

(CBCL)<br />

Ghodesian-Carpey and Aggression<br />

Mothers’ Mean¼5.2 M þF 0.65 (21) 0.35 (17) 0.60 a –<br />

Baker, 1987<br />

(MOCL)<br />

observations<br />

Philadelphia twins Impatience/aggression Teacher rating 6–11 M þF 0.67 (71) 0.11 (34) 1.12 a – Not tested<br />

(Meininger et al.,<br />

1988)<br />

Competitive achievement<br />

striving<br />

0.63 (71) 0.13 (34) 1.00 a –<br />

Minnesota twins Aggression<br />

Self-report Mean¼19.8 M þF 0.61 (79) 0.09 (48) Not tested<br />

(McGue et al., 1993) c (MPQ)<br />

Aggression<br />

Self-report Mean¼29.6 M þF 0.58 (79) 0.14 (48) Not tested<br />

(MPQ)<br />

Midwest twins BDHI—assault Self-report Mean¼42.5 F 0.07 (77) 0.41 (21) 0.00 – N/A<br />

(Cates et al., 1993) BDHI—indirect Self-report Mean¼42.5 F 0.40 (77) 0.01 (21) 0.78 – N/A<br />

hostility<br />

BDHI—verbal Self-report Mean¼42.5 F 0.41 (77) 0.06 (21) 0.70 – N/A<br />

hostility<br />

Colorado twins<br />

(Schmitz et al., 1995)<br />

Aggression<br />

(CBCL)<br />

Parent rating 2–3<br />

4–11<br />

MþF 0.68 (77)<br />

0.79 (66)<br />

0.40 (183)<br />

0.41 (137)<br />

0.52<br />

0.55<br />

0.16<br />

0.19<br />

Not tested<br />

(Continues)


Table 8.2. (Continued)<br />

Study sample<br />

(author, year)<br />

Ohio twins, Western<br />

Reserve Twin Project<br />

(Edelbrock et al., 1995)<br />

Dutch twins<br />

(van den Oord et al.,<br />

1996)<br />

Minnesota twins<br />

(Finkel and McGue,<br />

1997)<br />

Aggression measure<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(MPQ)<br />

Assessment<br />

method Age in years Sex<br />

Parent rating 7–15<br />

Mean¼11.0<br />

Parent rating 3 M<br />

F<br />

MF<br />

Self-report 27–64<br />

Mean¼37.8<br />

MZ<br />

r(N)<br />

DZ<br />

r(N) h 2 c 2 Sex limitation effects<br />

MþF 0.75 (99) 0.45 (82) 0.60 0.15 Not tested<br />

M<br />

F<br />

MF<br />

0.81 (210)<br />

0.83 (265)<br />

0.37 (220)<br />

0.39 (406)<br />

0.49 (236)<br />

0.49 (238)<br />

0.45 (409)<br />

0.12 (165)<br />

0.14 (352)<br />

0.12 (114)<br />

0.69 0.12 Not tested<br />

VET twins BDHI—assault Self-report Mean¼44.1 M 0.50 (182) 0.19 (118) 0.47 0.00 N/A<br />

(Coccaro et al., 1997) BDHI—indirect Self-report M 0.42 (182) 0.02 (118) 0.40 0.00 N/A<br />

hostility<br />

BDHI—verbal Self-report M 0.28 (182) 0.07 (118) 0.28 0.00 N/A<br />

hostility<br />

Swedish twins (TCHAD;<br />

Eley et al., 1999)<br />

Aggression<br />

(CBCL)<br />

0.72 (176)<br />

0.82 (160)<br />

UK twins (sample<br />

obtained from Register<br />

of Child Twins; Eley<br />

et al., 1999)<br />

Virginia twins<br />

(Simonoff et al., 1998)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(physical)<br />

Parent rating 8–9 M<br />

F<br />

MF<br />

Parent rating 12 M<br />

F<br />

MF<br />

0.68 (99)<br />

0.77 (124)<br />

0.41 (182)<br />

0.45 (194)<br />

0.41 (310)<br />

0.45 (93)<br />

0.44 (80)<br />

0.27 (95)<br />

0.35<br />

0.39<br />

0.00<br />

0.00<br />

NS quantitative sex<br />

differences<br />

0.70 0.07 NS quantitative sex<br />

differences,<br />

NS qualitative sex<br />

differences<br />

0.69 0.04 NS quantitative sex<br />

differences,<br />

NS qualitative sex<br />

differences<br />

Parent rating 8–16 MþF 0.76 (268) 0.46 (166) 0.58 0.18 Not tested<br />

Self-report 8–16 MþF 0.31 (268) 0.22 (166) 0.21 0.11 Not tested


Missouri twins<br />

(Hudziak et al., 2000)<br />

Dutch twins<br />

(Hudziak et al., 2003)<br />

Dutch twins<br />

(van Beijsterveldt<br />

et al., 2003) c<br />

Canadian twins<br />

(Dionne et al., 2003)<br />

UK (E-risk) twins<br />

(Taylor, 2004)<br />

South Wales twins<br />

(Button et al., 2004)<br />

Finnish twins<br />

(Vierikko et al., 2004)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(TRF)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(physical)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(IAB)<br />

Aggression<br />

(MPNI)<br />

Parent rating 8–12 M<br />

F<br />

0.77 (129)<br />

0.73 (91)<br />

0.50 (156)<br />

0.40 (115)<br />

0.77<br />

0.70<br />

0.00 Not tested<br />

Teacher rating 7 M 0.72 (181) 0.33 (160)<br />

F 0.71 (214) 0.33 (151)<br />

MF<br />

0.26 (330)<br />

10 M 0.73 (153) 0.41 (140) 0.72 0.00<br />

F 0.73 (202) 0.25 (125) 0.21 0.49<br />

MF<br />

0.17 (283)<br />

Parent rating 3 M 0.81 (1055) 0.55 (1066)<br />

F 0.82 (1226) 0.53 (997)<br />

MF<br />

0.48 (2144)<br />

7 M 0.83 (927) 0.48 (1069)<br />

F 0.84 (898) 0.53 (858)<br />

MF<br />

0.51 (1723)<br />

10 M 0.84 (526) 0.50 (621)<br />

F 0.79 (471) 0.55 (458)<br />

MF<br />

0.47 (907)<br />

12 M 0.86 (289) 0.45 (317)<br />

F 0.83 (237) 0.55 (233)<br />

MF<br />

0.57 (433)<br />

Parent rating 1.5 MþF 0.59 (107) 0.28 (174) 0.58 0.00 Not tested<br />

0.69 0.00 NS quantitative sex<br />

differences<br />

Parent rating 5 MþF 0.73 (602) 0.24 (514) 0.72 0.00 Not tested<br />

Significant quantitative<br />

sex differences<br />

Significant quantitative<br />

sex differences<br />

Significant quantitative<br />

sex differences<br />

Significant quantitative<br />

sex differences<br />

Significant quantitative<br />

sex differences<br />

Self-report 11–18<br />

Mean¼13.8<br />

MþF 0.64 (115) 0.40 (143) 0.68 0.00 Not tested<br />

Parent rating 12 M 0.72 (260) 0.59 (292) 0.14 0.75 Significant quantitative<br />

F 0.78 (300) 0.53 (278) 0.54 0.25 sex differences<br />

MF<br />

0.58 (517)<br />

(Continues)


Table 8.2. (Continued)<br />

Study sample<br />

(author, year)<br />

Dutch twins<br />

(Polderman et al.,<br />

2006)<br />

Colorado twins<br />

(Haberstick et al.,<br />

2006a)<br />

Aggression measure<br />

Aggression<br />

(TRF)<br />

Aggression<br />

(CBCL)<br />

Aggression<br />

(TRF)<br />

Assessment<br />

method Age in years Sex<br />

MZ<br />

r(N)<br />

DZ<br />

r(N) h 2 c 2 Sex limitation effects<br />

Teacher rating 5 MþF 0.84 (67) 0.43 (59) 0.49 0.00 Not tested<br />

(same teacher)<br />

Teacher rating<br />

(different<br />

teachers)<br />

5 MþF 0.40 (45) 0.21 (44)<br />

Parent rating 7 b M 0.74 (69) 0.56 (76) 0.79 0.00 NS quantitative sex<br />

F 0.79 (91) 0.44 (62)<br />

differences<br />

9 M<br />

F<br />

10 M<br />

F<br />

11 M<br />

F<br />

12 M<br />

F<br />

Teacher rating 7 M<br />

F<br />

8 M<br />

F<br />

9 M<br />

F<br />

10 M<br />

F<br />

11 M<br />

F<br />

12 M<br />

F<br />

0.57 (73)<br />

0.76 (75)<br />

0.77 (58)<br />

0.70 (67)<br />

0.64 (58)<br />

0.86 (56)<br />

0.68 (69)<br />

0.83 (78)<br />

0.63 (71)<br />

0.56 (79)<br />

0.58 (66)<br />

0.70 (70)<br />

0.54 (63)<br />

0.67 (74)<br />

0.39 (63)<br />

0.49 (64)<br />

0.56 (68)<br />

0.50 (70)<br />

0.35 (55)<br />

0.48 (60)<br />

0.50 (63)<br />

0.55 (60)<br />

0.47 (52)<br />

0.56 (57)<br />

0.41 (55)<br />

0.59 (49)<br />

0.45 (61)<br />

0.45 (65)<br />

0.39 (70)<br />

0.34 (62)<br />

0.46 (62)<br />

0.41 (60)<br />

0.29 (59)<br />

0.37 (53)<br />

0.44 (56)<br />

0.18 (54)<br />

0.12 (54)<br />

0.45 (54)<br />

0.32 (39)<br />

0.24 (49)<br />

0.76 0.00 NS quantitative sex<br />

differences<br />

0.76 0.00 NS quantitative sex<br />

differences<br />

0.84 0.00 NS quantitative sex<br />

differences<br />

0.79 0.00 NS quantitative sex<br />

differences<br />

0.58 0.00 NS quantitative sex<br />

differences<br />

0.61 0.00 NS quantitative sex<br />

differences<br />

0.59 0.00 NS quantitative sex<br />

differences<br />

0.43 0.00 NS quantitative sex<br />

differences<br />

0.52 0.00 NS quantitative sex<br />

differences<br />

0.42 0.00 NS quantitative sex<br />

differences


Ad-Health<br />

(Cho et al., 2006)<br />

Colorado twins<br />

(Gelhorn et al., 2006)<br />

Finnish twins<br />

(von der Pahlen et al.,<br />

2008)<br />

Norwegian twins<br />

(Czajkowski et al.,<br />

2008)<br />

Tennessee twins<br />

(Tackett et al., 2009)<br />

California twins—RFAB<br />

cohort (Baker et al.,<br />

2008)<br />

Aggression Self-report 12–19 M<br />

F<br />

MF<br />

Aggression<br />

Self-report 11–18 MþF<br />

(DISC items)<br />

Mean¼14.5 MF<br />

Aggression<br />

(BPAQ)<br />

Passive<br />

aggression<br />

(DSM-IV)<br />

Relational aggression<br />

(CAPS)<br />

Reactive aggression<br />

(RPQ)<br />

Self-report 18–33 M<br />

F<br />

MF<br />

Self-report 19–36<br />

Mean¼28.2<br />

M<br />

F<br />

MF<br />

Self- report 9–18 M<br />

F<br />

MF<br />

Parent rating 9–18 M<br />

F<br />

MF<br />

Parent rating 9–10 M<br />

F<br />

MF<br />

Self-report 9–10 M<br />

F<br />

MF<br />

Teacher rating 9–10 M<br />

F<br />

MF<br />

0.47 (141)<br />

0.47 (141)<br />

0.29 (131)<br />

0.27 (114)<br />

0.21 (197)<br />

0.47 (531) 0.27 (569)<br />

0.28 (212)<br />

0.45 (190)<br />

0.52 (608)<br />

0.35 (221)<br />

0.30 (448)<br />

0.54 (356)<br />

0.41 (376)<br />

0.66 (356)<br />

0.65 (376)<br />

0.48 (141)<br />

0.60 (142)<br />

0.38 (138)<br />

0.37 (139)<br />

0.59 (67)<br />

0.70 (68)<br />

0.22 (167<br />

þ321 sibs)<br />

0.18 (387<br />

þ1838 sibs)<br />

0.20 (508<br />

þ1559 sibs)<br />

0.45 (116)<br />

0.19 (261)<br />

0.21 (340)<br />

0.39 (328)<br />

0.36 (332)<br />

0.16 (589)<br />

0.61 (328)<br />

0.35 (332)<br />

0.48 (589)<br />

0.35 (87)<br />

0.46 (98)<br />

0.50 (151)<br />

0.28 (83)<br />

0.38 (96)<br />

0.08 (146)<br />

0.49 (45)<br />

0.43 (45)<br />

0.60 (62)<br />

0.50<br />

0.30<br />

0.00<br />

0.00<br />

Not tested<br />

0.49 0.00 Not tested<br />

0.70<br />

0.69<br />

0.00<br />

0.00<br />

Not tested<br />

0.14 0.18 NS quantitative sex<br />

differences,<br />

NS qualitative sex<br />

differences<br />

0.49 0.00 NS quantitative sex<br />

differences<br />

0.21<br />

0.42<br />

0.46<br />

0.22<br />

Significant quantitative<br />

sex differences<br />

0.26 0.27 NS quantitative sex<br />

differences<br />

0.38<br />

0.00<br />

0.00<br />

0.36<br />

Significant quantitative<br />

sex differences<br />

0.20 0.43 NS quantitative sex<br />

differences<br />

(Continues)


Table 8.2. (Continued)<br />

Study sample<br />

(author, year)<br />

Aggression measure<br />

Assessment<br />

method Age in years Sex<br />

MZ<br />

r(N)<br />

DZ<br />

r(N) h 2 c 2 Sex limitation effects<br />

California twins—RFAB<br />

cohort (follow-up)<br />

(Tuvblad et al., 2009)<br />

Weighted average<br />

Proactive aggression<br />

(RPQ)<br />

Reactive aggression<br />

(RPQ)<br />

Proactive aggression<br />

(RPQ)<br />

Parent rating 9–10 M<br />

F<br />

MF<br />

Self-report 9–10 M<br />

F<br />

MF<br />

Teacher rating 9–10 M<br />

F<br />

MF<br />

Parent rating 11–14 M<br />

F<br />

MF<br />

Parent rating 11–14 M<br />

F<br />

MF<br />

M<br />

F<br />

MF<br />

MþF<br />

0.61 (141)<br />

0.57 (142)<br />

0.60 (138)<br />

0.12 (139)<br />

0.56 (67)<br />

0.74 (68)<br />

0.49 (102)<br />

0.58 (98)<br />

0.55 (102)<br />

0.46 (98)<br />

0.66<br />

0.63<br />

0.59<br />

0.34 (87)<br />

0.48 (98)<br />

0.55 (151)<br />

0.34 (83)<br />

0.28 (96)<br />

0.14 (146)<br />

0.42 (45)<br />

0.38 (45)<br />

0.35 (62)<br />

0.33 (55)<br />

0.38 (77)<br />

0.42 (103)<br />

0.35 (55)<br />

0.27 (77)<br />

0.40 (103)<br />

0.42<br />

0.35<br />

0.38<br />

0.28<br />

0.32 0.21 NS quantitative sex<br />

differences<br />

0.50<br />

0.00<br />

0.00<br />

0.14<br />

Significant quantitative<br />

sex differences<br />

0.45 0.14 NS quantitative sex<br />

differences<br />

0.43 0.15 NS quantitative sex<br />

differences<br />

0.48 0.08 NS quantitative sex<br />

differences<br />

MZ, monozygotic; DZ, dizygotic; M, male twin pairs; F, female twin pairs; MF, male–female twin pairs; MþF, male and female pairs combined; h 2 , heritability; c 2 , shared<br />

environment; CBCL, Child Behavior Checklist (Achenbach, 1991b) [20 items scored as 0 (not true), 1 (somewhat true), and 2 (very true), e.g., bragging and boasting, argues a<br />

lot, cruelty or meanness to other, disobedience (home and school)]; MOCL, Mothers’ Observational Checklist, including the following behaviors: rejection, destructiveness,<br />

negativism, noncompliance, teasing, physical negative, insult, verbal threat, yelling; ACL, Adjective Checklist (Gough, 1960) [consists of 300 adjectives that yields 26 scales];<br />

MCPS, Missouri Children’s Picture Series (Sines et al., 1966) [consists of 238 line drawings, each portrays the figure of a child engaged in some activity or situation, the subject is<br />

required to sort the cards into two groups, those that look like fun and those that do not look like fun]; PSR, Parent Symptom Ratings (Conners, 1970) [includes six aggression<br />

items: bullying, hits or kicks other children, mean, sassy to grown-ups, fights constantly, picks on other children]; IBS, Interpersonal Behavior Survey (Mauger, 1980) [includes


items such as: “some people think I have a violent temper” or “I try not to give people a hard time”]; The Mathews Youth Test for Health (MYTH; Mathew and Angulo, 1980)<br />

[developed to measure Type A behavior in school-aged children. The instrument consists of 17 items characterized by overt type A behavior and yields two subscales: impatience/<br />

aggression and competitive achievement striving]; BDHI, Buss-Durkee Hostility Inventory (Buss and Durkee, 1957) [contains of three subscales: the assault scale (10 items on<br />

physical aggression), the verbal hostility scale (13 items on verbal aggression), and the indirect hostility scale (nine items on indirect or undirected or displaced aggression)];<br />

MPQ, Minnesota Personality Questionnaire (Tellegen, unpublished) [physically aggressive, vindictive, likes violent scenes, higher order factor, negative emotionality]; Physical<br />

Aggression scale, (Simonoff et al., 1998) [items on physical aggression, extortion, public fight, use of weapon in a fight, cruelty to animal, thrown objects at people, carried a<br />

weapon, sworn at teacher, based on Olweus, 1989]; Physical Aggression scale (Dionne et al., 2003) is a 37 item check list on which parents reported whether the child engaged,<br />

sometimes engaged, often engaged in a behavior. Based on factor analysis, 10 of the 37 behaviors were determined as direct physical aggression, for example, is cruel toward others,<br />

bullies other children, bites others, kicks, fights, takes things away from others, pushes, threatens to hit; IAB, instrument of aggressive behavior (Olweus, 1989) [contains two<br />

subscales: aggressive and nonaggressive antisocial behavior. The aggression scale contains 11 items of direct verbal and physical aggression, e.g., swearing at a teacher, bullying];<br />

MPNI, Multidimensional Peer Nomination Inventory (Pulikkined et al., 1999) [contains of 38 items and the aggression subscale contains of six items, e.g., calls people names,<br />

may hurt other kids, bullying, goes around telling people’s secrets to others]; Ad-Health [aggression is based on four items, got into a serious physical fight, hurt someone badly<br />

enough they needed medical care, used to threaten to use a weapon to get something from someone, took part in a gang fight]; DISC, Diagnostic Interview Schedule for Children<br />

(Shaffer et al., 2000) [Gelhorn et al. (2006) only included aggression to people or animals, items 1–7]; BPAQ, Buss and Perry Aggression Questionnaire (Buss and Perry, 1992)<br />

[nine items on physical aggression and five items on verbal aggression, e.g., I cannot help getting into arguments when people disagree with me, I have threatened people I know, I<br />

get into fights a little bit more often than average people]; DSM-IV, the Norwegian version of the Structured Interview for DSM-IV personality (Pfohl et al., 1997) [the<br />

instrument is a semi-structured diagnostic interview for the assessment of all DSM axis II disorders, including passive-aggressive personality disorder]; CAPS, Child and<br />

Adolescent Psychopathology Scale (Lahey et al., 2004) [relational aggression was assessed via the CAPS, a structured interview assessing DSM-IV symptoms of common<br />

childhood disorders. Seven items measured relational aggression, for example, tried to keep kids he/she does not like outside his/her friend group, spread rumors to make others<br />

stop liking someone, stopped talking to people because he/she was mad at them, teased other people in a mean way]; RPQ, Reactive and Proactive Questionnaire (Raine et al.,<br />

2006). [The RPQ is a validated 23-item questionnaire designed to measure reactive and proactive aggression in children and adolescents from the age of 8. The RPQ includes 11<br />

reactive items (e.g., “he/she damages things when he/she is mad”; “he/she gets mad or hits others when they tease him/her”) and 12 proactive items (e.g., “he/she threatens and<br />

bullies other kids”; “he/she damages or breaks things for fun”). The items in the RPQ have a three-point response format: 0¼never, 1 ¼sometimes, 2 ¼often.]; RFAB, USC twin<br />

study of Risk Factors for Antisocial Behavior; TCHAD: Twin Study of Child and Adolescent Development.<br />

a Heritability estimate is based on either Holzingers’ H or Falconer equation and did not report shared environmental influences.<br />

b Parent reported CBCL ratings were not collected at age 8.<br />

c Genetic and shared environmental estimates were not reported by the authors.


184 Tuvblad and Baker<br />

aggressive and antisocial outcomes. All three of these studies are ongoing. One of<br />

these is the <strong>University</strong> of Southern California Twin Study of Risk Factors for<br />

Antisocial Behavior (RFAB), which is a prospective study of the interplay of<br />

genetic, environmental, social, and biological (psychophysiological) factors on<br />

the development of antisocial and aggressive behavior from childhood to emerging<br />

adulthood. The project includes more than 750 twin pairs studied on several<br />

occasions, at ages 9–10, 11–13, 14–16, and 17–18 years (Baker et al., 2006). A<br />

second major twin study is the Environmental Risk Longitudinal Twin Study (Erisk<br />

study) in the United Kingdom. The E-risk study involves data on more than<br />

1000 twin pairs at ages 5, 7, and 12 with the special focus on what factors in the<br />

home, family, school, and neighborhood (i.e., environmental risks) promote children’s<br />

aggression (Moffitt, 2002). The Minnesota Study of Twins and Families<br />

(MFTS) is a third major longitudinal twin study that specifically investigates<br />

antisocial behavior and substance use across development. MTFS was established<br />

in 1989 using same-sexed twin pairs aged 11 or 17. Five hundred additional 11-yearold<br />

twin pairs were added in 2000. All twins of those ages, who were born in<br />

Minnesota, as identified by birth registry data, were invited to participate. Participants<br />

are asked about academic ability, personality, and interests; family and social<br />

relationships; mental and physical health; and physiological measurements. Of<br />

particular interest are prevalence of psychopathology, substance abuse, divorce,<br />

leadership, and other traits and behaviors related to mental and physical health,<br />

relationships, and religiosity (Iacono et al., 2006; Keyes et al.,2009).<br />

Before reviewing the twin and sibling adoption studies on aggressive<br />

behavior presented in Tables 8.1 and 8.2, it is important to consider the ways in<br />

which the phenotype of aggressive behavior is defined and measured. The various<br />

instruments utilized in the studies reviewed in this chapter are summarized in<br />

Tables 8.1 and 8.2, to provide a clear idea of the nature of the aggressive behavior<br />

phenotype being investigated. By and large, the Child Behavior Checklist<br />

(CBCL; Achenbach, 1991b) has been used more often than any other single<br />

instrument in behavioral genetic studies of aggression. Although self-report<br />

version of the CBCL is available for older adolescents and young adults (Youth<br />

Self Report (YSR); Achenbach, 1991c), studies more commonly rely on parent<br />

or teacher (Teacher’s Report Form (TRF); Achenbach, 1991a) rating versions.<br />

The aggressive behavior subscale of the CBCL includes 20 items on which the<br />

child is rated. These include defiance, argumentativeness, physical aggression,<br />

and cruelty toward others. Although there are a handful of other instruments<br />

that also yield single aggressive behavior scores, two instruments provide multiple<br />

scales: the Reactive and Proactive Aggression Questionnaire (RPQ; Raine<br />

et al., 2006), which provides separate scales for aggressive reactions to provocation<br />

and more planned or proactive forms of aggression; and the Buss-Durkee<br />

Hostility Inventory (BDHI; Buss and Durkee, 1957), which yields several subscales<br />

of aggression, including assault, verbal, and indirect hostility.


8. Human Aggression Across the Lifespan 185<br />

The studies summarized in Tables 8.1 and 8.2 vary on how aggressive<br />

behavior was defined (i.e., physical, verbal, relational, reactive, proactive, indirect,<br />

bullying) and measured (observation, self-report, parent/caregiver, teacher). A<br />

wide range of ages were included, from preschool children to adults; however,<br />

the vast majority of studies have used childhood samples (i.e., 12 years of age or<br />

younger) which explains why the CBCL is so frequently used to assess aggressive<br />

behavior. Correlations for biological and adoptive siblings (Table 8.1), and MZ<br />

and DZ twins (Table 8.2) are shown for each study. Most studies reported correlations<br />

separately for same-sex pairs of males (M), females (F), and opposite-sex pairs<br />

(MF); however, a few studies involve correlations for samples of male and female<br />

pairs combined. We review the key questions concerning the genetic influence<br />

(heritability) of human aggression based on the effect sizes reported for these<br />

studies. We also examine various potential moderators of these effects, including<br />

sex, age, method of assessment, form of aggression, study design (twin vs. sibling<br />

adoption design), and various social factors and circumstances that may exacerbate<br />

or ameliorate the genetic risk for aggression from one person to the next.<br />

A. Does heritability vary depending on sex<br />

Since it is well documented that males are much more likely than females to<br />

engage in most forms of aggressive behavior (Moffitt et al., 2001; Rutter et al.,<br />

2003), it is also of interest to examine whether the same genetic and environmental<br />

influences are important in both sexes and whether the magnitude of<br />

these effects differs between males and females.<br />

In the classical twin design, genetic and environmental variance components<br />

for aggressive behavior can be estimated using data from same-sex MZ<br />

and DZ twins. Apart from estimating genetic and environmental effects on<br />

aggression, it is also possible to investigate whether sex-specific genetic or<br />

environmental influences are important. Such effects are referred to as sexlimitation<br />

or sex-limited effects. There are two primary questions about sex<br />

limitation in genetic research, one being whether there are qualitative differences<br />

between males and females, such that different genes and/or environmental<br />

influences operate in the two sexes, and whether quantitative differences exist<br />

in the relative magnitude of influences across sexes. To assess whether the<br />

magnitude of genetic and environmental effects in aggressive behavior differ<br />

between males and females (i.e., quantitative sex differences), only data from<br />

same-sex twin pairs are required. However, to determine whether or not it is the<br />

same set of genes or shared environmental experiences that influences aggressive<br />

behavior in males and females (i.e., qualitative sex differences), data from<br />

opposite-sex twin pairs are also needed. If qualitatively different genetic influences<br />

are important for aggressive behavior in males and females, then the<br />

opposite-sex twins will be less genetically similar for the trait than DZ twins.


186 Tuvblad and Baker<br />

Not all twin studies have examined sex-limited effects, either qualitative<br />

or quantitative, and several studies combined males and females when<br />

computing twin correlations, making it impossible to evaluate these effects<br />

based on published results shown in Table 8.2. Nonetheless, quantitative sex<br />

differences can be easily evaluated across at least 18 studies in Table 8.2, which<br />

present separate twin or sibling correlations by sex. Among these, there are a<br />

dozen studies that also include MF, which allow investigation of qualitative sex<br />

differences. The average twin correlations across these 18 studies, weighted by<br />

their respective sample sizes, shows quite similar twin correlations for both<br />

identical (r MZ Males ¼0.66; r MZ Females ¼0.63) and nonidentical same-sex pairs<br />

(r DZ Males ¼0.42; r DZ Females ¼0.35), indicating that there are no appreciable<br />

quantitative sex differences in aggressive behavior. This is consistent with the<br />

individual results across studies which formally tested for quantitative sex differences<br />

(e.g., Baker et al., 2008; Czajkowski et al., 2008; Eley et al., 1999; Finkel<br />

and McGue, 1997; Tackett et al., 2009; Tuvblad et al., 2009). As indicated in<br />

Table 8.2, only a small handful of studies have reported significant differences in<br />

heritability of aggression for males and females (and these are primarily for<br />

younger samples; e.g., Hudziak et al., 2003; van Beijsterveldt et al., 2003;<br />

Vierikko et al., 2004). The lack of quantitative sex differences is also well in<br />

line with what was reported in a recent meta-analysis summarizing 19 twin and<br />

family/adoption studies, whereby genetic influences were found to explain 54%<br />

of the variance in aggressive behavior in boys and 57% of the variance in girls<br />

(Burt, 2009).<br />

There is no evidence of qualitative sex differences either, given that the<br />

weighted twin correlation for MF (DZ Male Female ) is 0.38, which is quite similar<br />

to the same-sex DZ twin correlations (0.42 in males and 0.35 in females).<br />

In spite of the consistent sex difference in mean levels of aggression, the<br />

underlying etiologies of aggressive behavior appear to be remarkably similar for<br />

both sexes. There may still be biological and social differences between the sexes<br />

that might account for the greater mean levels of aggression observed in males,<br />

yet the same genes and the same environmental factors appear to explain<br />

individual differences in aggression within each sex to the same degree. One<br />

interesting question that has not been addressed, however, is to what extent<br />

there may be sex differences in moderators of genetic factors. In other words,<br />

there may be different circumstances or experiences in males and females that<br />

lead to greater expression of genetic predispositions for aggression. For example,<br />

sexual jealousy might trigger genetic propensity for aggression to a greater extent<br />

in males than females, while threats to resources might be a more important<br />

moderator of genetic influences in females compared to males, as discussed<br />

in Chapter 9. Other moderators are discussed later in II.A, although more<br />

research is clearly warranted to explore the degree to which they may be sex<br />

specific.


8. Human Aggression Across the Lifespan 187<br />

B. Does heritability change across age<br />

Although genetic studies of aggression have spanned from childhood to adulthood,<br />

most studies included in Tables 8.1 and 8.2 involved children 12 years of<br />

age or younger. This suggests that more studies examining the heritability of<br />

aggressive behavior in adolescents and adults are needed. Keeping this in mind, it<br />

is useful to examine the magnitude of twin correlations across age groups, which<br />

span from early childhood to middle-age adults. These correlations are summarized<br />

in Fig. 8.1, according to five age groups (early childhood, age 1.5–6 years;<br />

middle childhood, age 7–10; adolescence 11–15; late adolescence/young adulthood,<br />

age 16–26; and adulthood, age 27–48; Fig. 8.1). These results show that<br />

aggressive behavior is clearly influenced by genetic factors across the lifespan,<br />

given the fact that the MZ correlations exceed those for DZ pairs at all ages. (The<br />

Twin correlations across age groups (all studies)<br />

0.80<br />

Mean twin correlation<br />

0.60<br />

0.40<br />

0.20<br />

0.00<br />

Early<br />

childhood<br />

(1.5–6)<br />

Middle<br />

childhood<br />

(7–10)<br />

Adolescence<br />

(11–15)<br />

Late<br />

adolescence/<br />

young adult<br />

(16–26)<br />

Adult<br />

(27–48)<br />

Age group<br />

Zygosity<br />

MZ DZ same–sex DZ male–female<br />

Figure 8.1. Twin correlations across age groups (all studies).


188 Tuvblad and Baker<br />

lack of qualitative sex differences is also evident across the life span, in that the<br />

DZ correlation is comparable for same-sex and MF pairs across ages.) However,<br />

both MZ and DZ correlations decline steadily across development, suggesting the<br />

waning importance of shared environmental effects from childhood to adolescence<br />

and then adulthood. The DZ correlation exceeds half the value of the MZ<br />

correlation (taken as evidence for shared environment) only in early childhood,<br />

but not in later age groups. The pattern shown in Fig. 8.1 is evident in individual<br />

studies as well. Aggressive behavior in childhood is influenced by genetic factors<br />

in all studies, and most of these studies also report shared environmental influences<br />

(Table 8.2; e.g., Baker et al., 2008; Eley et al., 1999; Hudziak et al., 2003;<br />

Schmitz et al., 1995; Simonoff et al., 1998; Tuvblad et al., 2009; van den Oord<br />

et al., 1996; Vierikko et al., 2004; but for an exception see Dionne et al., 2003;<br />

Taylor, 2004). Studies including adolescent twins (younger than 19 years of<br />

age) do not in most cases report finding any shared environmental influences<br />

(Table 8.2; e.g., Button et al., 2004; Cho et al., 2006; Gelhorn et al., 2006;<br />

Tackett et al., 2009). Similarly, studies including adult twins do not report<br />

finding any shared environmental influences (Table 8.2; e.g., Coccaro et al.,<br />

1997; Finkel and McGue, 1997; von der Pahlen et al., 2008; but for an exception<br />

see Czajkowski et al., 2008). The overall pattern across the studies presented in<br />

Table 8.2 and Fig. 8.1 indicates that genetic influences for aggressive behavior<br />

become increasingly more important, while shared environmental effects become<br />

less so as children develop from childhood, through adolescence, and into<br />

adulthood. Similarly, findings from a recent meta-analysis reported that genetic<br />

influences increased from 55.2% at ages 1–5 years to 62.7% at ages 6–10 years<br />

and 62.9% at ages 11–18 years. At the same time, shared environmental influences<br />

were decreasing from 18.7% at ages 1–5 years to 13.9% at ages 6–10 years<br />

and 2.7% at ages 11–18 years (Burt, 2009). This pattern of decrease in shared<br />

environment, and a concomitant increase in heritability during development, is<br />

relatively common for personality traits and cognitive abilities (Bartels et al.,<br />

2002; Loehlin, 1992; Plomin et al., 2001; Scarr and McCartney, 1983), and has<br />

also been found for other phenotypes including prosocial behavior (Knafo and<br />

Plomin, 2006).<br />

It should also be kept in mind, however, that methods of assessing<br />

aggression vary across age, such that studies of children tend to rely on ratings<br />

by teachers and parents, while studies of adults (and some older adolescents) rely<br />

more heavily on self-report methods. The confound between method of assessment<br />

and age of the subjects has made it difficult in prior studies and metaanalyses<br />

to disentangle age effects on heritability from differences that arise from<br />

different methods of assessment. Increasing heritability estimates from child to<br />

adulthood could therefore also be explained by different methods of assessment<br />

as well (e.g., parental bias may lead to overestimation of shared environmental<br />

effects and thus attenuate heritability estimates in childhood).


8. Human Aggression Across the Lifespan 189<br />

C. Do heritabilities vary across methods of assessment<br />

It is important to examine the magnitude of twin correlations across methods of<br />

assessment, as heritability estimates may vary depending on who is rating the<br />

subject. This is especially important given the age trends found for heritable<br />

influences in Fig. 8.1, since different methods of assessment tend to be employed<br />

for different age groups. As previously discussed, studies of younger subjects rely<br />

on parent or teacher ratings, while self-report methods are typically used in studies<br />

of adults and often adolescents. The twin correlations are summarized in Fig. 8.2,<br />

according to laboratory observation, self-reports, teacher ratings, and parent/<br />

caregiver ratings. Indeed, twin correlations—and thus the estimates of genetic<br />

and environmental influences on aggressive behavior—do appear to vary across<br />

method of assessment. According to the twin correlations summarized in Fig. 8.2,<br />

the heritability of aggressive behavior based on laboratory observation is approximately<br />

32% [h 2 :2(r MZ r DZ )¼2(0.27 0.11)], dominant genetic effect accounts<br />

for approximately 10% [d 2 :2(r MZ r DZ )¼2(0.27)–4(0.11)], and the nonshared<br />

0.8<br />

Twin correlations across method of assessment (all studies)<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

Observation Self-report Teacher Parent<br />

MZ DZ same-sex DZ male-female<br />

Figure 8.2. Twin correlations across method of assessment (all studies).


190 Tuvblad and Baker<br />

environment accounts for the remaining 58% of the variance. The heritability of<br />

aggressive behavior based on self-reports is 40% and the nonshared environment<br />

accounts for the remaining 60% of the variance. There is no evidence of shared<br />

environmental contribution, as the DZ correlation is approximately half the MZ<br />

correlation. The heritability of aggressive behavior based on teacher ratings is<br />

54%, shared environmental influences account for 6% [c 2 :2r DZ r MZ ¼2(0.33<br />

0.60)], and the nonshared environment accounts for the remaining 40% of the<br />

variance. The heritability of aggressive behavior based on parent/caregiver ratings<br />

is 54%, shared environmental influences account for 17%, and the nonshared<br />

environment accounts for the remaining 29% of the variance. Thus, parent/<br />

caregiver ratings have the largest familial influence, explaining 71% of the<br />

variance (h 2 þc 2 ¼0.54%þ17%) in individual differences in aggressive behavior.<br />

It should be kept in mind that this is a descriptive approach and that formal<br />

modeling is required to determine how well these estimates describe the observed<br />

data. Also, this approach does not allow for actual testing of different hypotheses,<br />

for example, to test whether it is possible to set any of these effects to zero. It is<br />

difficult to discern whether the parent/caregiver rating patterns reflect true shared<br />

environment or are instead an artifact of rater bias whereby raters are less able to<br />

discriminate between the two twins’ aggressive behavior and thus inflate the<br />

similarities between them, regardless of zygosity. In fact, when the two co-twins<br />

are rated by different teachers (e.g., they are in different classrooms), twin<br />

correlations are lower for both MZ and DZ pairs for the wider construct of<br />

antisocial behavior (Baker et al., 2007).<br />

A few specific twin studies in Table 8.2, which utilize multiple raters in<br />

their design, also illustrate that genetic and environmental influences on aggressive<br />

behavior vary depending on method of assessment. For example, twin<br />

similarity for relational aggression was influenced only by genetic factors when<br />

using self-reported data, explaining 49% of the total variance. When using parent<br />

ratings (only biological mothers were used as raters) of relational aggression both<br />

genetic and shared environmental influences were important (boys: h 2 ¼21%,<br />

c 2 ¼46%, e 2 ¼33%; girls: h 2 ¼42%, c 2 ¼22%, e 2 ¼36%). However, when using<br />

youth self-reports, only genetic and nonshared environmental influences were<br />

significant (h 2 ¼49%, e 2 ¼51%; Tackett et al., 2009). A similar pattern was found<br />

for reactive and proactive aggression in 9–10-year-old boys, whereby only genetic<br />

influences were important for self-reports, but both genes and shared environment<br />

were important for teacher and parent ratings (Baker et al., 2008). Aggressive<br />

behavior in another sample of twins aged 7–12 years was found to be largely<br />

influenced by genetic (or familial) factors (76%–84%), as reported by parents.<br />

Data were collected from one or both parents; however, only mother-reported<br />

ratings were included in the analyses, as they accounted for the majority of the<br />

ratings collected (85.3%–90.1%). In contrast, when teacher ratings were used,<br />

aggressive behavior was found to be slightly less influenced by genetic (or familial)


8. Human Aggression Across the Lifespan 191<br />

factors (42%–61%; Haberstick et al., 2006a). Significant shared environmental<br />

effects were not found for either parent or teacher ratings, and were therefore<br />

dropped from the models, suggesting that any shared environmental influences<br />

are likely to be included in the genetic component. Apart from rater bias, which<br />

may result in inflated twin similarity across the board when using single raters for<br />

multiple children, the varying patterns of genetic and environmental influence<br />

across methods of assessment could be the result of different raters reporting<br />

different aspects of the child’s aggressive behavior. This could arise in part because<br />

individuals behave differently in different situations (e.g., school vs. home) or<br />

because some types of aggressive behaviors are more likely to be noticed (e.g.,<br />

overt forms such as physical aggression) than other types of aggressive behaviors<br />

which may be more subtle or covert (e.g., relational aggression). Different raters<br />

provide important and unique pieces of information regarding behaviors. Selfreporters<br />

are aware of their own motives and behaviors, which may go undetected<br />

by their caregivers, teachers, or peers. On the other hand, caregivers or teachers<br />

may be able to understand difficult and complex constructs better than children.<br />

A teacher is also more likely to compare a child’s behavior to his or her peers,<br />

whereas a parent is likely to compare a child’s behavior to his or her siblings<br />

(Bartels et al., 2003). Regardless of the source of these discrepant results across<br />

methods of assessment, it is important to keep in mind that when it comes to<br />

studies of aggression, it matters who is doing the rating.<br />

D. Do heritabilities vary across forms of aggression<br />

Different types of aggressive behavior have been investigated across twin and<br />

adoption studies, with notable distinctions between reactive and proactive forms<br />

of aggression, as well as direct/physical and indirect/relational aggression<br />

(Table 8.3). It is likely that there are different etiologies for different forms of<br />

aggression; for example, defensive reactions to threatening stimuli may be more<br />

environmentally influenced, while more planned, proactive forms may be more<br />

genetically influenced (Tuvblad et al., 2009). Comparing heritability estimates<br />

collectively across the various measures employed is a reasonable way to address<br />

Table 8.3. Forms of Aggression<br />

Form of aggression<br />

Reactive/hostile/affective<br />

Proactive/instrumental<br />

Direct/physical<br />

Indirect/relational<br />

Description<br />

Angry or frustrated responses to a real or perceived threat<br />

Planning, the motive of the act extends beyond harming the victim<br />

Intentionally causing pain or harm to the victim<br />

Relational social manipulation such as gossip and peer exclusion


192 Tuvblad and Baker<br />

this question about whether some kinds of aggression are more heritable than<br />

others. When multiple forms of aggressive behavior are measured within the<br />

same study, it is also possible to investigate the extent to which the same genes<br />

and/or environmental factors are important to different manifestations of aggressive<br />

tendencies.<br />

Reactive aggression refers to angry or frustrated responses to a real or<br />

perceived threat. This specific type of aggression has been characterized to<br />

involve both high emotional arousal, impulsivity, and an inability to regulate<br />

or control affect. In contrast, proactive aggression is conceptualized as a more<br />

regulated, instrumental form of aggression, with more positive expectancies<br />

about the outcomes of aggression (Dodge, 1991; Dodge and Coie, 1987;<br />

Schwartz et al., 1998). Although reactive and proactive aggression have each<br />

been found to be mainly influenced by genetic and nonshared environmental<br />

factors, their genetic correlation is significantly less than 1.0, indicating some<br />

genetic specificity for the two forms of aggression. Reactive and proactive aggression<br />

each exhibit different developmental patterns in these influences (see Table 8.2;<br />

Baker et al., 2008; Tuvblad et al., 2009), that is, the genetic and environmental<br />

stability in reactive and proactive aggression has been found to differ. In one of the<br />

few longitudinal analyses of these constructs, the stability in reactive aggression<br />

from childhood to adolescence could be explained by genetic (48%), shared (11%),<br />

and nonshared (41%) environmental influences, whereas the continuity in proactive<br />

aggression was primarily genetically (85%) mediated (Tuvblad et al.,2009).<br />

Relational forms of aggression, which involve social manipulation such<br />

as gossip and peer exclusion, are often more indirect compared to other forms of<br />

aggression (Crick and Grotpeter, 1996). Like reactive and proactive forms,<br />

relational aggression appears to be influenced by genetic factors, both in<br />

self-report (49%) as well as parental reports (boys, 42%; girls, 21%). However,<br />

unlike the other more direct forms of aggression, relational aggression is also<br />

influenced by shared environmental influences, but only in parental reports<br />

(boys, 22%; girls, 46%) and not in self-report measures (Tackett et al., 2009).<br />

Together these findings—that is, the less than perfect genetic correlation between<br />

reactive and proactive forms, their different developmental etiologies,<br />

and the significant shared environmental effects in relational aggression only—<br />

provide support for at least some genetic and environmental etiological distinction<br />

among different forms of aggression. It should be noted, however, that no<br />

study to date has examined the genetic and environmental overlap between<br />

relational aggression and other forms such as proactive and reactive aggression.<br />

Other studies based on multifactorial measures of aggression, such as the<br />

BDHI, suggest some variability in the heritability estimates across subscales,<br />

although the patterns are not entirely consistent across studies. For example,<br />

“indirect hostility” showed the lowest heritability (28%) in one study of adult<br />

twins (Coccaro et al., 1997), compared to modest heritability for “verbal


8. Human Aggression Across the Lifespan 193<br />

hostility” (47%) and “assault” (40%). Yet, Cates et al. (1993) found no genetic<br />

influences for assault, but strong heritability for both verbal hostility (78%) and<br />

indirect hostility (70%). In multivariate genetic analyses, both studies found<br />

some support for genetic specificity for the various subscales, similar to what has<br />

been found for reactive and proactive aggression, in that genetic correlations<br />

(r G ) among the BDHI subscales were less than unity: r G ¼0.39 between indirect<br />

hostility and assault, r G ¼0.60 between indirect hostility and verbal hostility,<br />

r G ¼0.17 between verbal hostility and assault (Coccaro et al., 1997), r G ¼0.35<br />

between indirect hostility and assault, r G ¼0.39 between indirect hostility and<br />

verbal hostility, and r G ¼0.49 between verbal hostility and assault (Cates et al.,<br />

1993). Overall, genetic influences are generally found for most, if not all forms of<br />

aggression, although somewhat different genetic factors may be operating across<br />

these different forms. The mechanisms that underlie more direct, planned,<br />

confrontational, and often physical forms of aggression may to some extent be<br />

different than those for reactive or indirect aggressive behaviors.<br />

E. Does heritability vary depending on study design (twins vs.<br />

adopted siblings)<br />

There were only a handful of studies identified examining the heritability of<br />

aggressive behavior using the sibling adoption design. Visual inspection of the<br />

results from these sibling adoption studies (see Table 8.1) compared to the results<br />

from studies including twin samples (Table 8.2) indicate that heritability estimates<br />

(i.e., h 2 ) and the shared environmental estimates (i.e., c 2 ) for aggressive behavior are<br />

very similar. This is also well in line with the results of a meta-analysis on antisocial<br />

and aggressive behavior that found no differences between twin and sibling adoption<br />

studies (h 2 ¼48%; c 2 ¼13%, e 2 ¼0.39%) (Rhee and Waldman, 2002). Thus, the<br />

heritability of aggressive behavior does not seem to vary depending on study design.<br />

F. Criticisms of twin and adoption studies: Assumptions and<br />

generalizability<br />

There are several assumptions in both twin and adoption studies that are important<br />

to consider when reviewing their findings. In adoption studies, the most important<br />

factors are (1) random placement of the adoptees into homes and (2) generalizability.<br />

Selective placement or matching (i.e., similarities between adoptive and<br />

biological parents) for certain characteristics can lead to inflated correlations<br />

between adoptive siblings (and thus overestimated effects of shared environment).<br />

Although such matching may occur for physical characteristics (including race),<br />

direct selective placement is unlikely to be made for aggressive behavior, per se.<br />

(Children with more aggressive or antisocial biological parents would not be placed<br />

into homes with more aggressive adoptive parents.) Thus, it is unlikely that the


194 Tuvblad and Baker<br />

genetic and environmental effects summarized in Table 8.1 are biased in any way as<br />

a result of selective placement. In terms of generalizability, it is often the case that<br />

adoptive parents tend to be in good health and from higher socioeconomic levels;<br />

thus, findings from adoption studies may not always be unquestionably generalized<br />

to the entire population (Rutter, 2006). Adopted children may also be at greater<br />

risk for aggression compared to nonadoptees, since birth parents giving up their<br />

children may have increased rates of disordered behaviors, including substance use,<br />

criminal offending, and aggression (Cloninger et al., 1985; Lewis et al.,2001). In the<br />

Deater-Deckard and Plomin (1999) study, the adopted children did in fact have<br />

higher aggression scores compared to the nonadopted children, consistent with the<br />

notion that adoptees may be at higher genetic risk for aggression compared to<br />

nonadopted individuals. The elevated levels of aggression in adoptees occurred in<br />

spite of the fact that background characteristics of the adoptive families were found<br />

to be representative of families with children in the larger Denver area, and that the<br />

demographic characteristics of the adoptive grandparents and the adopted children’s<br />

biological grandparents were similar, with regard to educational and occupational<br />

level. Similarly, van der Valk et al. (1998) reported mean differences<br />

between adoptees and nonadoptees, with adoptees showing higher mean levels in<br />

aggressive behavior. About 75% of the adoptees were adopted from Korea, India,<br />

Columbia, Indonesia, Bangladesh, or Lebanon and the remaining 25% were<br />

adopted from European or other non-European countries in both the van der<br />

Valk et al. (1998) and the van den Oord et al. (1994) studies, and the majority of<br />

the adoptive parents had a higher level of occupation. Given the higher aggression<br />

scores among adoptees compared to nonadoptees, as well as the somewhat greater<br />

affluence and ethnic heterogeneity in at least some of the adoption samples, the<br />

generalizability of adoption study results to the wider population could be<br />

questioned.<br />

To what extent are the twin study results generalizable to the wider<br />

population Twins and singletons have been found to experience similar rates of<br />

psychiatric disorders (e.g., attention-deficit hyperactivity disorder (ADHD),<br />

oppositional defiant disorder, conduct disorder) and behavioral and emotional<br />

problems (Gjone and Novik, 1995; Moilanen et al., 1999; Simonoff et al., 1997;<br />

van den Oord et al., 1995). Findings from the RFAB study show no differences in<br />

mean levels between MZ and DZ twins in reactive or proactive aggression (Baker<br />

et al., 2008; Tuvblad et al., 2009). It can, therefore, be assumed that twins and<br />

singletons display equal rates of aggressive behavior.<br />

There are, however, two ways in which twins differ from singletons:<br />

(i) lower birth weight, due to shorter length of gestation (Plomin et al.,2001)and<br />

(ii) delayed language development (Rutter and Redshaw, 1991). Birth weight has<br />

been found to have a minimal effect on academic performance; for twins this effect<br />

was judged relative to what is a normal birth weight for twins and not for singletons<br />

(Christensen et al., 2006). However, studies have shown that children with birth


8. Human Aggression Across the Lifespan 195<br />

complications are more likely to later develop antisocial and aggressive behavior<br />

(Raine, 2002), but birth complications may not by themselves predispose antisocial<br />

and aggressive behaviors, but will require the presence of an environmental risk<br />

factor (e.g., poor parenting, maternal rejection). In other words, the relationship<br />

between birth complications and antisocial and aggressive behavior is confounded<br />

by environmental risk factors (Hodgins et al.,2001;Raineet al.,1997).<br />

In addition to generalizability, there are several key assumptions of the<br />

classical twin design that need to be kept in mind when reviewing findings from<br />

these studies. These include (1) the equal environments assumption, (2) random<br />

mating, and (3) lack of correlation or interaction between genetic and environmental<br />

influences. We briefly review each of these assumptions here—both in<br />

general and as they pertain to aggressive behavior in particular—and consider<br />

their possible effects on the results summarized across studies.<br />

Perhaps the most important and commonly criticized assumption is the<br />

“equal environment assumption” (EEA). In the classical twin design, MZ twins,<br />

who are assumed to share 100% of their genes, are compared to DZ twins, who<br />

are assumed to on average share 50% of their genes. If MZ twins are more similar<br />

than DZ twins, it may be inferred that the difference is caused by genetic effects.<br />

To make this inference, however, it is necessary to rely on the EEA. It is assumed<br />

that environmentally caused similarity is roughly equal for both MZ and DZ<br />

twins. If this assumption is violated, higher correlations among MZ twins may be<br />

due to environmental factors, rather than genetic factors, and heritability estimate<br />

will be overestimated (Plomin et al., 2001).<br />

Several twin studies of various phenotypes have examined the EEA.<br />

One way to test the validity of the EEA is to examine whether a trait of interest is<br />

influenced by perceived versus assigned zygosity. The effect of perceived zygosity<br />

can be added as a “specified” familial environment in a univariate ACE twin<br />

model (Kendler et al., 1993) and if this parameter can be omitted without any<br />

significant loss in data fit, it can be assumed that the EEA holds for the<br />

phenotype under study. These studies generally report that the EEA assumption<br />

holds for numerous phenotypes such as physical activity, eating behavior, psychiatric<br />

disorders (e.g., major depression, generalized anxiety disorder, phobia,<br />

alcohol, and drug abuse; Eriksson et al., 2006; Hettema et al., 1995; Kendler et al.,<br />

1993; Klump et al., 2000; Xian et al., 2000), including child and adolescent<br />

psychopathology such as anxiety disorder, ADHD, oppositional defiant disorder,<br />

conduct disorder, antisocial behavior (Cronk et al., 2002; Jacobson et al., 2002;<br />

Tuvblad et al., 2011) as well as aggressive behavior (Derks et al., 2006).<br />

The assumption of random mating for aggression in the parents of the<br />

twins is also important to consider, since nonrandom mating can lead to<br />

increased resemblance for DZ but not MZ twin pairs. Assortative mating in the<br />

parent generation acts to increase the resemblance between dizygotic twins and<br />

thereby bias shared environmental estimates upward and additive genetic effects


196 Tuvblad and Baker<br />

downward. A significant correlation between spouses for a particular trait is often<br />

interpreted as assortative mating (Maes et al., 1998). This assumption is probably<br />

violated when it comes to antisocial and aggressive behavior, as significant<br />

spouse correlations have been found suggesting that assortative mating exists<br />

in this behavioral domain (Krueger et al., 1998; Maes et al., 2007; Taylor et al.,<br />

2000). Taylor et al. (2000) found that parents of twins were correlated for<br />

retrospectively reported delinquency (r¼0.23 in families of boys and r¼0.35<br />

in families of girls) and concluded that assortative mating is modest in degree.<br />

Another study using data from the Dunedin sample in New Zealand (Silva and<br />

Stanton, 1996) when the participants were 21-years-old found a correlation<br />

(r¼0.54) between couple members’ reports of antisocial behavior in their<br />

peers (i.e., participants were asked how many of their friends had aggression,<br />

personal, alcohol, or drug problems, or did things against the law), which was<br />

identical to the correlation for couple members’ reports of their own antisocial<br />

behavior as measured by a variety of offenses (e.g., theft, force, fraud, vice). They<br />

concluded that assortative mating for antisocial behavior is substantial and that<br />

antisocial individuals tend to cluster in peer groups with similar antisocial peers.<br />

As such, assortative mating should to be taken into account when modeling<br />

antisocial behavior (Krueger et al., 1998). It is interesting, however, that the<br />

shared environmental effects are fairly negligible in twin studies of aggressive<br />

behavior, both in the prior meta-analyses as well as in our summary in Table 8.2.<br />

Thus, any assortative mating for aggression does not appear to have resulted in<br />

severe overestimates of shared environment when considering these studies<br />

en masse. It is possible, on the other hand, that genetic influences themselves<br />

have been underestimated and could be larger than the 50% or so than these<br />

meta-analyses and our summary suggest.<br />

It is also assumed in the classical twin design that genetic and environmental<br />

influences combine additively (i.e., do not interact) and are uncorrelated.<br />

It is possible, however, that some genetic predispositions may be associated with<br />

certain kinds of social environments or experiences, leading to a correlation<br />

between genes and environments. (GE interactions are also discussed at<br />

length in a later section of this chapter.) Such GE correlations (r GE ) can<br />

arise in three different ways (Scarr and McCartney, 1983): (i) Passive r GE occurs<br />

when genes overlap between parents and their offspring. For example, a child<br />

with aggressive parents inherits genetic susceptibility for aggression as well as<br />

experiences an adverse rearing environment. An example of passive r GE was<br />

reported in a study comparing genetic and environmental influences on mothering.<br />

Passive r GE correlations were suggested for mother’s positivity and monitoring.<br />

For mother’s negativity and control, primarily nonpassive r GE correlations<br />

were suggested (Neiderhiser et al., 2004). (ii) Evocative/reactive r GE can arise<br />

when a specific child characteristic elicits a particular response from the environment.<br />

For example, aggressive children tend to elicit more negative affect and


8. Human Aggression Across the Lifespan 197<br />

harsh discipline from their parents (Ge et al., 1996; O’Connor et al., 1998). In a<br />

more recent study, using the classical twin design the association between<br />

parental criticism and adolescent antisocial behavior was found to be entirely<br />

genetically influenced. Approximately half of the genetic contribution to this<br />

association was explained by early adolescent aggression. Thus, child aggression<br />

seemed to elicit negative parenting followed by adolescent antisocial behavior,<br />

indicating an evocative r GE (Narusyte et al., 2006). (iii) Active r GE is defined as<br />

the process whereby an individual actively seeks out environmental situations<br />

that are more closely matched to the person’s genotype. Active r GE has been<br />

suggested in adolescent drinking behavior, specifically among girls (Loehlin,<br />

2010). If the assumption of no GE correlation is violated, heritability estimates<br />

for aggressive behavior in twin studies could include both additive genetic effects<br />

and the effects of GE correlation (i.e., heritability estimates are inflated).<br />

Apart from these specific examples cited here, few studies have examined the<br />

effects of r GE in aggressive behavior, making it difficult to know the extent of<br />

their effect on heritability estimates in twin studies.<br />

In conclusion, findings from adoption studies should probably be<br />

generalized cautiously to other populations as adoptees tend to show higher<br />

scores on aggressive behavior compared to controls. On the other hand, most<br />

of the assumptions of the classical twin design seem to hold for aggressive<br />

behavior. The EEA has been tested and found to hold for various phenotypes<br />

including aggressive behavior, and twins and singletons have been found to<br />

display similar scores on aggressive behavior, suggesting that findings from twin<br />

studies can be generalized to other populations. Most twin studies report finding<br />

little or no shared environmental influences on aggressive behavior, suggesting<br />

that random mating is of little importance for aggressive behavior. Only a few<br />

studies have examined the influence of GE correlation on aggressive behavior,<br />

suggesting that more research is needed on this topic before we can draw any firm<br />

conclusions. Last, in the classical twin design, it is assumed that genetic and<br />

environmental influences combine additively and do not interact. This assumption<br />

is probably violated to some extent when it comes to aggressive behavior,<br />

as several studies have reported finding significant interaction effects. GE<br />

interaction is discussed in detail in the next section of this chapter.<br />

II. G E INTERACTION IN AGGRESSIVE BEHAVIOR<br />

There is a general recognition that genes and environment work together—often<br />

in complex ways—to produce wide variations in behavior and psychological<br />

function. GE interaction, by definition, is a statistical term indicating that<br />

genetic effects on a given phenotype depend upon environmental factors or vice<br />

versa. Gene expression, for example, can be moderated by an individual’s


198 Tuvblad and Baker<br />

experiences or exposure to certain environments. Likewise, various individuals<br />

may respond differently to the same environmental exposure because they have<br />

different genotypes. Such genetic sensitivity to the environment has been<br />

demonstrated extensively in plant and animal species for a variety of traits. But<br />

even though the importance of GE interactions in human behavior has long<br />

been considered (Eaves, 1984; Mather and Jinks, 1982), GE interactions have<br />

been rarely reported in human traits until relatively recently. The failure to find<br />

GE interactions in studies of human characteristics may be due to a number of<br />

factors. One likely explanation is related to statistical power. In general, it is<br />

difficult to detect GE effects due to their low statistical power (Rowe, 2003).<br />

For example, behavioral genetic studies rely on genetic relatedness for groups of<br />

individuals, rather than on sharing of specific alleles between pairs of relatives.<br />

Studies relying on variance partitioning often do not find significant GE<br />

effects, or find that they explain a very small portion of the total variance, and<br />

are thus dropped from further analysis. When G E is not taken into account in<br />

behavioral genetic studies, heritability estimates will tend to be biased, although<br />

the direction of the bias depends on whether the moderating environmental<br />

influences are of the shared or nonshared variety.<br />

GE interactions can be tested or modeled in behavioral genetic studies<br />

using several different study designs (e.g., twin or adoption). The two most<br />

frequently used methods testing for GE interactions in twin and adoption studies<br />

include: (1) a mean levels approach, testing whether mean values of a phenotype<br />

differ across different combinations of genetic risk and environmental settings and<br />

(2) a moderated variance components approach, examining whether genetic and<br />

environmental variance for a trait varies across different measured environmental<br />

settings. These two different methods stem from the same conceptual idea, namely,<br />

that genetic effects vary across environments or vice versa. Their interpretations<br />

and meanings can be rather different, since one is based on means and the other is<br />

based on variances. The mean levels GE is perhaps a more traditional approach,<br />

is typically presented as a statistical interaction in an ANOVA, and indicates<br />

whether particular experiences, exposures, or other conditions may (on average)<br />

ameliorate or exacerbate specific genetic effects in groups of individuals with<br />

similar genetic and environmental risks. In comparison, the moderated effects<br />

approach does not address mean levels of risk, but instead evaluates whether<br />

variance in genes or environment differs across various measured conditions.<br />

Moderation can occur in either raw variances (V A , V C ,andV E )orinrelative<br />

effects (i.e., h 2 , c 2 , and e 2 ), and may not necessarily coincide with GE interactions<br />

found in mean levels. The latter point is important when evaluating GE<br />

interactions across studies using these different approaches, since different patterns<br />

can emerge from them. For example, it is possible that certain adverse environments<br />

(e.g., low socioeconomic status (SES)) may lead to some genes exerting<br />

stronger effects (mean levels), while overall, the relative variance explained by


8. Human Aggression Across the Lifespan 199<br />

genes (heritability) may be greater in other environments (e.g., high SES). The<br />

approach used for testing GE interactions can vary across study design, such that<br />

adoption designs or studies with measured genes are generally required for the<br />

mean levels approach, while the moderated variance components approach may be<br />

used in both twin and adoption studies in the absence of measured genes.<br />

In molecular genetic studies, both genes and environment are measured,<br />

rather than inferred from correlations among family members. GE interactions<br />

can therefore be tested in the general population, that is, without necessary<br />

reliance on a twin or adoption design. There are still advantages, nonetheless,<br />

to include measured genes and measured environments in the con<strong>text</strong> of a familybased<br />

design, including twins and other siblings as well as parents and offspring.<br />

Evidence of GE interaction in aggressive behavior has been reported<br />

in twin and adoption studies, and more recently in molecular genetic studies.<br />

Below is a summary of some of the GE interaction findings in aggressive<br />

behavior from adoption, twin and molecular genetic studies. We also discuss<br />

two potential moderators of genetic and environmental influences on aggressive<br />

behavior, exposure to media violence, and alcohol use.<br />

A. Potential moderators of genetic influence found in adoption and<br />

twin studies<br />

1. Family adversity and social disadvantage<br />

GE interaction for aggressive behavior has been found in several of the early<br />

adoption studies, using a mean levels approach. What these early adoption study<br />

findings generally showed was that early adverse environments had a greater<br />

negative impact on genetically “higher risk” children. Adopted children with<br />

criminal biological parents reared by a family where there was adversity showed<br />

higher rates of antisocial and aggressive behavior than adopted children with<br />

antisocial biological parents not raised in a home with adversity, and than<br />

adopted children raised in adversity who are not at higher genetic risk. For<br />

example, the interaction of inherited and postnatal factors was examined in<br />

about 800 Swedish men adopted at an early age. When both inherited factors<br />

and environmental risk factors were present, 40% were found to be criminal; if<br />

only genetic factors were present, 12.1% were criminal; if only environmental<br />

factors were present, 6.7% were criminal; and with neither inherited nor environmental<br />

factors being present, 2.7% were criminal (Cloninger et al., 1982).<br />

The fact that 12.1% plus 6.7% is less than 40% would thus be an indication of<br />

GE interactions. This finding was later replicated in females (Cloninger and<br />

Gottesman, 1987). It should be pointed out, however, that in the adoption<br />

design, the genetic risk factors themselves are considered in a general way,<br />

such that the exact nature of the genes is left unspecified, both in terms of


200 Tuvblad and Baker<br />

which loci or alleles may be involved and what underlying mechanisms may be<br />

involved in the path from genes to phenotype. Similarly, the environmental risk<br />

factors as indexed by certain traits in the adoptive parents or characteristics of<br />

their home do not necessarily specify the exact nature of the child’s experiences<br />

or how these lead to various outcomes.<br />

Further, maltreatment places children at risk for psychiatric morbidity,<br />

especially conduct problems. However, not all maltreated children will develop<br />

conduct problems. A recent twin study tested whether the effect of physical<br />

maltreatment on risk for conduct problems was strongest among those who were<br />

at high genetic risk for these problems using data from the E-risk study, a representative<br />

cohort of 1116 5-year-old British twin pairs and their families. Maltreatment<br />

was found to be associated with a greater increase in the probability of developing<br />

conduct problems among children who had a high genetic liability for conduct<br />

disorder compared to children who had a low genetic liability (Jaffee et al.,2005).<br />

This finding is consistent with the GE interaction found in adoption studies of<br />

antisocial and aggressive behavior, in which genetic effects were more pronounced<br />

in adverse environments. This clearly suggests that children in risky environments<br />

would benefit from interventions. However, another view of this interaction is that<br />

favorable genotypes can play a protective role on children’s risk for conduct<br />

problems, especially under circumstances of maltreatment.<br />

There are also a few studies based on twin samples that have used the<br />

moderated variance components approach to examine whether measured environmental<br />

(risk) factors moderate the genetic and environmental variances for<br />

aggressive behavior. For example, the heritability of conduct problems was found<br />

to be lower in children growing up in dysfunctional families and higher in<br />

children growing up in families where dysfunction was absent (Button et al.,<br />

2005). Another twin study used DeFries-Fulker regression analysis to examine<br />

whether genetic and environmental influences on aggressive behavior varied<br />

depending on levels of family warmth (DeFries and Fulker, 1985). Genetic<br />

influence on aggressive behavior was found to be higher in schools with higher<br />

average levels of family warmth. In contrast, environmental influences (both<br />

shared and nonshared) were more important in schools with lower average levels<br />

of family warmth (Rowe et al., 1999). These findings suggest that genetic effects<br />

are more likely to explain individual differences in aggression in more benign<br />

environments, whereas in more disadvantaged environments negative familyrelated<br />

factors and con<strong>text</strong>-dependent risks may play a greater role than genetic<br />

predispositions in aggressive and antisocial outcomes.<br />

Many early theories about the causes of delinquency and crime assumed<br />

that delinquents come from socially disadvantaged backgrounds. For example,<br />

Merton postulated that antisocial behavior resulted from the strain caused by the<br />

gap between cultural goals and the means available for their achievement<br />

(Merton, 1957). Social disadvantage and poverty constitute a reasonable robust,


8. Human Aggression Across the Lifespan 201<br />

although not always a strong, indication of an increased risk for antisocial and<br />

aggressive behavior, assessed by self-reports and official convictions (Leventhal<br />

and Brooks-Gunn, 2000; Rutter et al., 1998). SES has also been found to moderate<br />

the relative influence of genetic factors on antisocial and aggressive behavior. In a<br />

sample of Swedish 16–17-year-old twins, heritability for antisocial and aggressive<br />

behavior was higher in the more affluent neighborhoods (boys, 37%; girls, 69%)<br />

compared to the less advantaged neighborhoods (boys, 1%, girls, 61%). Conversely,<br />

the shared environment was higher in the less advantaged neighborhoods<br />

(boys, 69%; girls, 16%) compared to better-off neighborhoods (boys, 13%; girls,<br />

6%). Following the “social push hypothesis,” Raine (2002) would suggest that the<br />

genetic factors on antisocial and aggressive behavior are more expressed in a<br />

socioeconomically advantaged environment where the environmental risk factors<br />

are absent. On the contrary, genetic factors for antisocial behavior will be<br />

weaker and the shared environment more important in a socioeconomically<br />

disadvantaged environment because the environmental risk factors will “camouflage”<br />

the genetic contribution (Tuvblad et al., 2006).<br />

These studies using the moderated variance components approach (e.g.,<br />

Button et al., 2005; Rowe et al., 1999; Tuvblad et al., 2006), all examine whether<br />

an environmental (risk) factor moderates genetic and environmental variance<br />

on antisocial and aggressive behavior. Findings from these studies show that<br />

heritable influences on aggressive behavior vary depending on environmental<br />

con<strong>text</strong>, indicating the importance of the environmental risk factors in the<br />

development of aggressive behavior as well as for gene expression.<br />

2. Violent media exposure<br />

There is an ongoing debate about whether exposure to violent video games<br />

increases aggressive behavior, and it is very possible that exposure to media<br />

violence could moderate the influences of genetic and environmental influences<br />

on aggressive behavior. One line of research argues that mass media exposures<br />

contribute to a child’s socialization. A primary process in such socialization is<br />

observational learning (Bandura, 1973). Children and adolescents mimic what<br />

they see and acquire complicated scripts for behaviors, beliefs about the world,<br />

and moral precepts about how to behave in the long run from what they observe<br />

(Huesmann, 2010). In contrast, another line of research argues that there is little<br />

empirical evidence for a link between media exposure and violence. This line of<br />

research argues that media violence cannot have any important psychological<br />

effect on the risk for aggressive behavior (Ferguson and Kilburn, 2010).<br />

A recent meta-analysis that included 136 studies examined the effects<br />

of violent video games on aggressive behaviors. The evidence suggested that<br />

exposure to violent video games is a risk factor for increased aggressive behavior,


202 Tuvblad and Baker<br />

aggressive cognition, and aggressive affect, and for decreased empathy and<br />

prosocial behavior. Moderator analyses showed significant research design<br />

effects, weak evidence of cultural differences in susceptibility and type of measurement<br />

effects, and no evidence of sex differences in susceptibility. Sensitivity<br />

analyses were also carried out and they revealed these effects to be robust, with<br />

little evidence of selection (publication) bias (Anderson et al., 2010).<br />

Others studies examining the relationship between violent video games<br />

and aggressive acts have found little evidence for a relationship. A recent review<br />

included a total of 25 studies comprising 27 independent observations. The<br />

corrected overall effect size for all included studies was only r¼0.08 (Ferguson<br />

and Kilburn, 2009). The mixed findings in the literature clearly suggest that more<br />

research is needed to resolve whether there is a link between exposure to violent<br />

video games and aggressive behavior. Also, some studies have found that exposure<br />

to violent video games only explains a small fraction of the variance. An explanation<br />

for this paradox could be that exposure to violent video games moderates<br />

the influence of genetic and environmental effects on aggressive behavior, rather<br />

than exerting direct effects. No genetically informative studies have examined<br />

violent video game exposure as a possible moderator of genetic influence on<br />

aggression, however, leaving this as an important area in need of study.<br />

3. Alcohol use<br />

It has long been known that some individuals become aggressive after consuming<br />

alcohol, and the relationship of violence and aggression with alcohol is well<br />

established (Bushman and Cooper, 1990; White et al., 2001). For example, a<br />

review including 130 independent studies found that alcohol was correlated with<br />

both criminal and domestic violence (Lipsey et al., 1997). Despite this, there is so<br />

far no behavioral genetic study that had examined whether alcohol use moderates<br />

the influence of genetic and environmental factors on aggressive behavior.<br />

However, the genetic and environmental relationship among alcohol<br />

use and aggressive behavior as well as other disruptive and problem behaviors<br />

within the disinhibitory spectrum such as antisocial behavior, ADHD, conduct<br />

disorder, impulsive and sensation seeking personality traits has been examined in<br />

several large population-based twin studies. On a phenotypic level, disruptive<br />

and problem behaviors within the disinhibitory spectrum can be united by a<br />

common higher order externalizing factor (Krueger et al., 2002, 2005, 2007).<br />

This higher order externalizing factor has been found to be largely influenced by<br />

genetic factors. For example, the genetic influences on a common externalizing<br />

factor describing conduct disorder, substance use, ADHD, and novelty seeking<br />

was found to account for more than 80% of the variation in an adolescent sample<br />

(Young et al., 2000). Strong heritable influences on an externalizing factor of


8. Human Aggression Across the Lifespan 203<br />

antisocial behavior, substance abuse, and conduct disorder has also been found<br />

among adults (Kendler et al., 2003). Together these studies provide important<br />

insight into our understanding of externalizing behaviors. It seems that behaviors<br />

and disorders within the externalizing spectrum, including aggressive behavior,<br />

share a common genetic liability.<br />

III. SPECIFIC GENES FOR AGGRESSIVE BEHAVIOR: FINDINGS FROM<br />

MOLECULAR GENETIC STUDIES<br />

Increasing evidence suggests the importance of heritable factors in the development<br />

of aggressive behavior (Burt, 2009; Miles and Carey, 1997; Rhee and<br />

Waldman, 2002). The first study that showed a link between a specific genotype<br />

and aggressive behavior examined the genetic material of members of a large<br />

Dutch family. This specific family had for decades been found to be prone to<br />

violent, aggressive, and impulsive behavior, including fighting, arson, attempted<br />

rape, and exhibitionism. Some of the male family members were also intellectually<br />

disabled. The aggressive males in this large family were shown to share a<br />

mutation in the gene that codes for the enzyme MAO (monoamine oxidase A).<br />

MAO breaks down brain chemicals (neurotransmitters) such as serotonin, noradrenaline,<br />

and dopamine, which transmit messages from one nerve cell to the<br />

next. In the afflicted males, however, a mistake in the coding sequence governing<br />

proper production of MAO was detected. As a result, abnormally large quantities<br />

of these neurotransmitters were found in the blood of the affected males<br />

(Brunner et al., 1993). Although this genetic defect remains the first such link<br />

to aggressive behavior in humans, exactly how the genetic defect causes aggressive,<br />

impulsive behavior, or mental retardation is not known.<br />

Apart from MAO, only a few candidate genes have been linked to<br />

aggressive behavior to date. The candidate genes that have been found to be<br />

associated with aggressive behavior in humans have, in many cases, been replicated<br />

in animal studies. The majority of these candidate genes are genes of the<br />

dopamine, serotonin, and norepinephrine neurotransmitter systems. The dopamine<br />

system is involved in mood, motivation and reward, arousal, as well as other<br />

behaviors. The serotonin system is involved in impulse control, affect regulation,<br />

sleep, and appetite, whereas the epinephrine and norepinephrine system facilitate<br />

fight-or-flight reactions and autonomic nervous system activity (Niv and Baker,<br />

2010). For example, dopaminergic candidates, including dopamine receptor<br />

DRD4, has been found to be involved in ADHD and externalizing behavior,<br />

and DRD2 has been found to be involved in substance abuse and disinhibition<br />

(Niv and Baker, 2010). The DRD3 polymorphism has been found to be associated<br />

with impulsivity. This association was significant in violent, but not in nonviolent<br />

individuals, and there were no association between DRD3 and violence per se


204 Tuvblad and Baker<br />

(Retz et al., 2003). Dopamine transporter gene DAT1 has also been linked to<br />

ADHD (Waldman et al., 1998), as well as with violent behavior and delinquency<br />

in adolescents and young adults (Guo et al., 2007). Cateocholamine-O-methyltransferase<br />

(COMT) has been examined primarily in children and adults with<br />

ADHD, and mixed evidence emerged for its association with conduct disorder<br />

and aggression (Caspi et al., 2008). Several studies have provided evidence that<br />

the low activity VNTR alleles of 5HTTLPR show associations with aggression,<br />

violence, aggressive symptoms of conduct disorder, and other forms of externalizing<br />

behavior (Haberstick et al., 2006b; Linnoila et al., 1983). Aggressive behavior<br />

has also shown associations with SNPs of epinephrine and norepinephrine. A<br />

recent study linked two SNPs of PNMT to cognitive and aggressive impulsivity in<br />

children and adolescents (Oades et al., 2008).<br />

A. G E interaction involving specific genes for aggressive behavior<br />

Advances in the field of molecular genetics have also made it possible for<br />

researchers to identify GE interactions much more specifically. One of the<br />

most influential studies examining GE in antisocial and aggressive behavior is<br />

Caspi et al. (2002), a famous study from 2002. The relationship between a<br />

functional polymorphism in the MAO-A gene encoding the neurotransmittermetabolizing<br />

enzyme and early childhood maltreatment was examined in the<br />

development of antisocial behavior in males. A significant G E interaction<br />

was detected, in that maltreated boys with a genotype conferring low levels of<br />

MAO-A were found to be more likely to later develop antisocial problems,<br />

including conduct disorder, adult violent crime, and antisocial personality disorder,<br />

than maltreated boys who had a genotype conferring high levels of MAO-A<br />

(Caspi et al., 2002). So far, there have only been a few replications of this<br />

important finding (Foley et al., 2004; Kim-Cohen et al., 2006; Nilsson et al.,<br />

2006). For example, Kim-Cohen et al. (2006) found that the MAO-A polymorphism<br />

moderated the development of psychopathology after experiencing physical<br />

abuse in a sample of 975 seven-year-old boys. This finding was extended to<br />

the maltreatment exposure closer in time as the subjects were 7-years-old<br />

compared with previous work by Caspi et al. (2002) in which the subjects were<br />

26-years-old, and therefore the possibility of a spurious finding by accounting for<br />

passive and evocative GE correlation could be ruled out. Passive GE<br />

correlation, as discussed earlier, refers to the association between the genotype<br />

a child inherits from his/her parents and the environment in which the child is<br />

raised, and evocative GE correlation occurs when an individual’s (heritable)<br />

behavior evokes an environmental response. Further, the authors also conducted<br />

a meta-analysis including the following five studies: Caspi et al. (2002), Foley<br />

et al. (2004), Haberstick et al. (2005), Kim-Cohen et al. (2006), and Nilsson et al.<br />

(2006). The association between maltreatment and mental health problems


8. Human Aggression Across the Lifespan 205<br />

was significantly stronger in the group of males with a genotype conferring low<br />

versus high MAO-A activity. This provides strong evidence that the MAO-A<br />

gene influences vulnerability to environmental stress and that this biological<br />

process can be initiated early in life. However, there is at least one published<br />

failure to replicate (Haberstick et al., 2005), and this finding has been replicated<br />

neither in females (Sjöberg et al., 2007) nor in African Americans (Widom and<br />

Brzustowicz, 2006).<br />

AGE interaction between the DRD2 A1 allele and risk-level in family<br />

environments has been suggested in a sample of adolescents with criminal offenses,<br />

the National Longitudinal Study of Adolescent Health (Ad-Health). Polymorphisms<br />

in genes related to the neurotransmitter dopamine were associated with age of<br />

first police contact and arrests, but only for youth from low-risk family environments.<br />

More specifically, among those adolescents with a history of criminal<br />

offending, those at greatest risk for later onset were those with the A1 allelic<br />

form of the DRD2 gene, in combination with favorable home environments as<br />

defined by maternal attachment, involvement, and engagement (DeLisi et al.,<br />

2008). It is important to emphasize that this finding involves the age of onset of<br />

first police contact and not the overall risk for offending versus not offending.<br />

There is also some evidence for a GE interaction in the 5HTTLPR<br />

genotype with adult violence, whereby home violence, familial economic difficulties,<br />

and educational or home-life disruptions during childhood were found to<br />

predict violent behavior later in life only in individuals with the short promoter<br />

alleles present (Reif et al., 2007). A similar GE interaction between the short<br />

allele of 5HTTLPR and childhood adversity has also been reported for ADHD<br />

(Retz et al., 2008).<br />

The ability to detect GE interactions in molecular genetic studies is<br />

both exciting and controversial. The identification of specific genetic markers<br />

and specific experiences provides the opportunity to evaluate genetic and<br />

environmental risk factors at the individual level. This significantly increases<br />

opportunities for developing effective treatments and preventions for antisocial<br />

and aggressive behavior as well as other forms of psychopathology, which is<br />

exciting. At the same time, increased understanding of individual risks has<br />

often been considered cautiously because of the potential for bias and discrimination<br />

of those individuals who are identified as being at highest risk for being<br />

afflicted with disorders.<br />

IV. CONCLUSIONS<br />

Studies (and meta-analyses) including both twin and adoption samples show<br />

that about half (50%) of the variance in aggressive behavior is explained by<br />

genetic influences in both males and females, with the remaining 50% of the


206 Tuvblad and Baker<br />

variance being explained by nonshared environmental factors. Form of aggression<br />

(reactive, proactive, direct/physical, indirect/relational), method of assessment<br />

(observation, self, teacher, parent/caregiver), and age of the subjects—all<br />

seem to be significant moderators of the magnitude of genetic and environmental<br />

influences on aggressive behavior. Neither study design (twin vs.<br />

sibling adoption design) nor sex, on the other hand, seems to impact the<br />

nature or magnitude of these genetic and environmental influences on<br />

aggression.<br />

Although we are unaware of any twin or adoption studies of aggression<br />

induced in authoritative situations such as in the Milgram or Stanford Prison<br />

studies, the vast evidence for genetic influences in most forms of aggression that<br />

have been studied could suggest that individual differences in those early<br />

studies might have stemmed in part from different genetic propensities in<br />

their subjects. Findings from GE studies on aggressive behavior suggest that<br />

not all individuals will be affected to the same degree by these environmental<br />

exposures, and also that not all individuals will be affected to the same degree<br />

by the genetic predispositions. Adoption and twin studies rely on relationships<br />

between family members when examining GE interaction effects, whereas<br />

molecular genetic studies are using both a measured environmental (risk) factor<br />

and a measured genetic factor. To date, there have only been a few twin/<br />

adoption and molecular studies that report finding GE in aggressive behavior,<br />

either using the mean levels approach or the moderated effects approach. These<br />

studies have shown that various measures of family adversity and social disadvantage<br />

interact (or act as moderators) with genetic factors on aggressive<br />

behavior.<br />

Today, we have the potential to identify genetic risks at the level of<br />

specific genes, and identify aspects of the environment that make some individuals<br />

more vulnerable than others. Yet, there will always be groups of individuals<br />

with the same combination of genetic risk and environmental<br />

vulnerability who will not engage in aggressive behavior. So, it is still only an<br />

increased (probabilistic) risk and not a biological determinism. In spite of such<br />

strong support for a genetic basis to aggressive behavior, the importance of<br />

potential interventions which are environmentally based must not be ignored.<br />

Environmental interventions could be developed, for example, through family<br />

or school-based programs, to reduce aggressive behavior. In fact, a general view<br />

held by behavioral genetics researchers is that the best way to understand<br />

environment—and hence develop effect treatment interventions—is through<br />

genetically informative designs such as twin and family data. By using twin and<br />

family data, it is not only possible to estimate the influence of heritable factors<br />

on a trait or a phenotype, but also the influence of environmental factors.<br />

Modern methods for identifying and understanding GE interactions will<br />

provide a means for doing exactly this.


8. Human Aggression Across the Lifespan 207<br />

References<br />

Achenbach, T. M. (1991a). Manual for Teacher’s Report form and 1991 Profile. <strong>University</strong> of<br />

Vermont, Department of Psychiatry, Burlington, VT.<br />

Achenbach, T. M. (1991b). Manual for the Child Behavior Checklist/4-18 and 1991 Profile.<br />

<strong>University</strong> of Vermont, Department of Psychiatry, Burlington, VT.<br />

Achenbach, T. M. (1991c). Manual for the Youth Self-Report and 1991 Profile. <strong>University</strong> of<br />

Vermont, Department of Psychiatry, Burlington, VT.<br />

Anderson, C. A., Shibuya, A., Ihori, N., Swing, E. L., Bushman, B. J., Sakamoto, A.,<br />

Rothstein, H. R., and Saleem, M. (2010). Violent video game effects on aggression, empathy,<br />

and prosocial behavior in eastern and western countries: A meta-analytic review. Psychol. Bull.<br />

136(2), 151–173.<br />

Baker, L. A., Barton, M., Lozano, D. I., Raine, A., and Fowler, J. H. (2006). The southern California<br />

twin register at the university of southern California: II. Twin Res. Hum. Genet. 9, 933–940.<br />

Baker, L. A., Jacobson, K. C., Raine, A., Lozano, D. I., and Bezdjian, S. (2007). Genetic and<br />

environmental bases of childhood antisocial behavior: A multi-informant twin study. J. Abnorm.<br />

Psychol. 116(2), 219–235.<br />

Baker, L. A., Raine, A., Liu, J., and Jacobson, K. C. (2008). Differential genetic and environmental<br />

influences on reactive and proactive aggression in children. J. Abnorm. Child Psychol.<br />

36, 1265–1278.<br />

Bandura, A. (1973). Aggression: A Social Learning Theory Analysis. Prentice-Hall, New York.<br />

Bartels, M., Rietveld, M. J., van Baal, C. M., and Boomsma, D. (2002). Genetic and Environmental<br />

Influences on the Development of Intelligence. Behav. Genet. 32(4), 237–249.<br />

Bartels, M., Hudziak, J. J., van den Oord, E. J. C. G., van Beijsterveldt, C. E. M., Rietveld, M. J. H.,<br />

and Boomsma, D. (2003). Co-occurrence of aggressive behavior and rule-breaking behavior at age<br />

12: Multi-rater analyses. Behav. Genet. 33, 607–621.<br />

Brunner, H. G., Nelen, M., Breakefield, X. O., Ropers, H. H., and van Oost, B. A. (1993). Abnormal<br />

behavior associated with a point mutation in the structural gene for monoamine oxidase A.<br />

Science 262(5133), 578–580.<br />

Burt, A. S. (2009). Are there meaningful etiological differences within antisocial behavior Results of<br />

a meta-analysis. Clin. Psychol. Rev. 29, 163–178.<br />

Bushman, B. J., and Cooper, H. M. (1990). Effects of alcohol on human aggression: An integrative<br />

research interview. Psychol. Bull. 107(3), 341–354.<br />

Buss, A. H., and Durkee, A. (1957). An inventory for assessing different kinds of hostility. J. Consult.<br />

Clin. Psychol. 21, 343–349.<br />

Buss, A. H., and Perry, M. (1992). The aggression questionnaire. J. Pers. Soc. Psychol. 63, 452–459.<br />

Button, T. M. M., Scourfield, J., Martin, N., and McGuffin, P. (2004). Do aggressive and nonaggressive<br />

antisocial behaviors in adolescents result from the same genetic and environmental<br />

effects Am. J. Med. Genet. B Neuropsychiatr. Genet. 129 B, 59–63.<br />

Button, T. M. M., Scourfield, J., Martin, N., Purcell, S., and McGuffin, P. (2005). Family dysfunction<br />

interacts with genes in the causation of antisocial symptoms. Behav. Genet. 35, 115–120.<br />

Caspi, A., McClay, J., Moffitt, T. E., Mill, J., Martin, J., Craig, I. W., Taylor, A., and Poulton, R.<br />

(2002). Role of genotype in the cycle of violence in maltreated children. Science 297(5582),<br />

851–854.<br />

Caspi, A., Langley, K., Milne, B., Moffitt, T. E., O’Donovan, M., Owen, M. J., Polo Tomas, M.,<br />

Poulton, R., Rutter, M., Taylor, A., Williams, B., and Thapar, A. (2008). A replicated molecular<br />

genetic basis for subtyping antisocial behavior in children with attention-deficit/hyperactivity<br />

disorder. Arch. Gen. Psychiatry 65(2), 203–210.<br />

Cates, D. S., Houston, B. K., Vavak, C. R., Crawford, M. H., and Uttley, M. (1993). Heritability of<br />

hostility-related emotions, attitudes, and behaviors. J. Behav. Med. 16(3), 237–256.


208 Tuvblad and Baker<br />

Cho, H., Guo, G., Iritani, B. J., and Hallfors, D. D. (2006). Genetic contribution to suicidal behaviors<br />

and associated risk factors among adolescents in the US. Prev. Sci. 7(3), 303–311.<br />

Christensen, K., Petersen, I., Skytthe, A., Herskind, A. M., McGue, M., and Bingley, P. (2006).<br />

Comparison of academic performance of twins and singletons in adolescence: Follow-up study.<br />

Br. Med. J. 6, 1–5.<br />

Cloninger, C. R., and Gottesman, I. I. (1987). Genetic and environmental factors in antisocial<br />

behavior disorders. In “The Causes of Crime: New Biological Approaches” (S. A. Mednick,<br />

T. E. Moffitt, and S. A. Stack, eds.), pp. 92–109. Cambridge <strong>University</strong> Press, Cambridge.<br />

Cloninger, C. R., Sigvardsson, S., Bohman, M., and von Knorring, A. L. (1982). Predisposition to<br />

petty criminality in Swedish adoptees. II. Cross-fostering analysis of gene-environment interaction.<br />

Arch. Gen. Psychiatry 39, 1242–1247.<br />

Cloninger, C. R., Bohman, M., Sigvardsson, S., and von Knorring, A. L. (1985). Psychopathology in<br />

adopted-out children of alcoholics. The Stockholm Adoption Study. Recent Dev. Alcohol. 3,<br />

37–51.<br />

Coccaro, E. F., Bergeman, C. S., Kavoussi, R. J., and Seroczynski, A. D. (1997). Heritability of<br />

aggression and irritability: A twin study of the Buss-Durkee aggression scales in adult male<br />

subjects. Biol. Psychiatry 41(3), 273–284.<br />

Conners, C. K. (1970). Symptom patterns in hyperkinetic, neurotic, and normal children. Child Dev.<br />

41, 667–682.<br />

Crick, N. R., and Grotpeter, J. K. (1996). Children’s treatment by peers: Victims of relational and<br />

overt aggression. Dev. Psychopathol. 8, 267–280.<br />

Cronk, N., Slutske, W. S., Madden, P. A., Bucholz, K. K., Reich, W., and Heath, A. (2002).<br />

Emotional and behavioral problems among female twins: An evaluation of the equal environments<br />

assumption. J. Am. Acad. Child Adolesc. Psychiatry 41, 829–837.<br />

Czajkowski, N., Kendler, K. S., Jacobson, K., Tambs, K., Røysamb, E., and Reichborn-Kjennerud, T.<br />

(2008). Passive-aggressive (negativistic) personality disorder: A population-based twin study.<br />

J. Pers. Disord. 22(1), 109–122.<br />

Deater-Deckard, K., and Plomin, R. (1999). An adoption study of the etiology of teacher and parent<br />

reports of externalizing behavior problems in middle childhood. Child Dev. 70, 144–154.<br />

DeFries, J. C., and Fulker, D. F. (1985). Multiple regression analysis of twin data. Behav. Genet. 15,<br />

467–473.<br />

DeLisi, M., Beaver, K. M., Wright, J. P., and Vaughn, M. G. (2008). The etiology of criminal onset:<br />

The enduring salience of nature and nurture. J. Crim. Justice 36, 217–223.<br />

Derks, E. M., Dolan, C. V., and Boomsma, D. I. (2006). A test of the equal environment assumption<br />

(EEA) in multivariate twin studies. Twin Res. 9(3), 403–411.<br />

Dionne, G., Tremblay, R., Boivin, M., Laplante, D., and Perusse, D. (2003). Physical aggression and<br />

expressive vocabulary in 19-month-old twins. Dev. Psychol. 39(2), 261–273.<br />

Dodge, K. (1991). The structure and function of reactive and proactive aggression. In “The Development<br />

and Treatment of Childhood Aggression” (D. J. Pepler and K. H. Rubin, eds.),<br />

pp. 201–218. Erlbaum, Hillsdale, NJ.<br />

Dodge, K. A., and Coie, J. D. (1987). Social-information processing factors in reactive and proactive<br />

aggression in children’s peer groups. J. Pers. Soc. Psychol. 53, 1146–1158.<br />

Dodge, K. A., Lochman, J. E., Harnish, J. D., Bates, J. E., and Pettit, G. S. (1997). Reactive and<br />

proactive aggression in school children and psychiatrically impaired chronically assaultive youth.<br />

J. Abnorm. Psychol. 106, 37–51.<br />

Eaves, L. J. (1984). The resolution of genotype x environment interaction in segregation analysis of<br />

nuclear families. Genet. Epidemiol. 1, 215–228.<br />

Edelbrock, C., Rende, R., Plomin, R., and Thompson, L. A. (1995). A twin study of competence and<br />

problem behavior in childhood and early adolescence. J. Child Psychol. Psychiatry 36(5), 775–785.


8. Human Aggression Across the Lifespan 209<br />

Eley, T. C., Lichtenstein, P., and Stevenson, J. (1999). Sex differences in the etiology of aggressive<br />

and nonaggressive antisocial behavior: Results from two twin studies. Child Dev. 70(1), 155–168.<br />

Eriksson, M., Rasmussen, F., and Tynelius, P. (2006). Genetic factors in physical activity and the equal<br />

environment assumption—The Swedish young male twin study. Behav. Genet. 36(2), 238–247.<br />

Ferguson, C. J., and Kilburn, J. (2009). The public health risks of media violence: A meta-analytic<br />

review. J. Pediatr. 154(5), 759–763.<br />

Ferguson, C. J., and Kilburn, J. (2010). Much ado about nothing: The misestimation and overinterpretation<br />

of violent video game effects in eastern and western nations: Comment on Anderson<br />

et al. (2010). Psychol. Bull. 136(2), 174–178; discussion 82–87.<br />

Finkel, D., and McGue, M. (1997). Sex differences and nonadditivity in heritability of the Multidimensional<br />

Personality Questionnaire Scales. J. Pers. Soc. Psychol. 72(4), 929–938.<br />

Foley, D. L., Eaves, L., Wormley, B., Silberg, J., Maes, H., Kuhn, J., and Riley, B. (2004). Childhood<br />

adversity, monoamine oxidase a genotype, and risk for conduct disorder. Arch. Gen. Psychiatry 61,<br />

738–744.<br />

Ge, X., Conger, R. D., Cadoret, R. J., Neiderhiser, J. M., Yates, W. R., Troughton, E., and<br />

Stewart, M. A. (1996). The developmental interface between nature and nurture: A mutual<br />

influence model of child antisocial behavior and parent behaviors. Dev. Psychol. 32(4), 574–589.<br />

Gelhorn, H. L., Stallings, M. C., Young, S. E., Corely, R. P., Rhee, S. H., Hopfer, C., and Hewitt, J. K.<br />

(2006). Common and specific genetic influences on aggressive and nonaggressive conduct disorder<br />

domains. J. Am. Acad. Child Adolesc. Psychiatry 45(5), 570–577.<br />

Ghodesian-Carpey, J., and Baker, L. A. (1987). Genetic and environmental influences on aggression<br />

in 4- to 7-year-old twins. Aggress. Behav. 13, 173–186.<br />

Gjone, H., and Novik, T. S. (1995). Parental ratings of behaviour problems: A twin study and general<br />

population comparison. J. Child Psychol. Psychiatry 36(7), 1213–1224.<br />

Gough, H. G. (1960). The adjective check list as a personality assessment research technique.<br />

Psychol. Rep. 6, 107–122.<br />

Guo, G., Roettger, M. E., and Shih, J. C. (2007). Contributions of the DAT1 and DRD2 genes to<br />

serious and violent delinquency among adolescents and young adults. Hum. Genet. 121, 125–136.<br />

Haberstick, B. C., Lessem, J. M., Hopfer, C. J., Smolen, A., Ehringer, M. A., Timberlake, D., and<br />

Hewitt, J. K. (2005). Monoamine oxidase A (MAOA) and antisocial behaviors in the presence of<br />

childhood and adolescent maltreatment. Am. J. Med. Genet. B Neuropsychiatr. Genet. 135B(1),<br />

59–64.<br />

Haberstick, B. C., Schmitz, S., Young, S. E., and Hewitt, J. K. (2006a). Genes and developmental<br />

stability of aggressive behavior problems at home and school in a community sample of twins aged<br />

7–12. Behav. Genet. 36, 809–819.<br />

Haberstick, B. C., Smolen, A., and Hewitt, J. K. (2006b). Family-based association test of the<br />

5HTTLPR and aggressive behavior in a general population sample of children. Biol. Psychiatry<br />

59, 836–843.<br />

Haney, C., Banks, W. C., and Zimbardo, P. G. (1973). A study of prisoners and guards in a simulated<br />

prison. Naval Res. Rev. 30, 4–17.<br />

Hettema, J. M., Neale, M. C., and Kendler, K. S. (1995). Physical similarity and the equal-environment<br />

assumption in twin studies of psychiatric disorders. Behav. Genet. 25(4), 327–335.<br />

Hodgins, S., Kratzer, L., and McNeil, T. F. (2001). Obstetric complications, parenting, and risk of<br />

criminal behavior. Arch. Gen. Psychiatry 58, 746–752.<br />

Hudziak, J. J., Rudiger, L. P., Neale, M. C., Heath, A. C., and Todd, R. D. (2000). A twin study of<br />

inattentive, aggressive, and anxious/depressed behaviors. J. Am. Acad. Child Adolesc. Psychiatry<br />

39(4), 469–476.<br />

Hudziak, J. J., van Beijsterveldt, C. E. M., Bartels, M., Rietveld, M. J. H., Rettew, D. C., Derks, E. M.,<br />

and Boomsma, D. (2003). Individual differences in aggression: Genetic analyses by age, gender,<br />

and informant in 3-, 7-, and 10-year-old Dutch Twins. Behav. Genet. 33(5), 575–589.


210 Tuvblad and Baker<br />

Huesmann, L. R. (2010). Nailing the coffin shut on doubts that violent video games stimulate<br />

aggression: Comment on Anderson et al. (2010). Psychol. Bull. 136(2), 179–181.<br />

Iacono, W. G., McGue, M., and Krueger, R. F. (2006). Minnesota center for twin and family<br />

research. Twin Res. Hum. Genet. 9(6), 978–984.<br />

Jacobson, K. C., Prescott, C. A., and Kendler, K. (2002). Sex differences in the genetic and<br />

environmental influences on the development of antisocial behavior. Dev. Psychol. 14, 395–416.<br />

Jaffee, S. R., Caspi, A., Moffitt, T. E., Dodge, K. A., Rutter, M., Taylor, A., and Tully, L. A. (2005).<br />

Nature X nurture: Genetic vulnerabilities interact with physical maltreatment to promote conduct<br />

problems. Dev. Psychopathol. 17(1), 67–84.<br />

Kendler, K., Neale, M. C., Kessler, R. C., Heath, A. C., and Eaves, L. J. (1993). A test of the equalenvironment<br />

assumption in twin studies of psychiatric illness. Behav. Genet. 23(1), 21–27.<br />

Kendler, K., Prescott, C. A., Myers, J., and Neale, M. C. (2003). The structure of genetic and<br />

environmental risk factors for common psychiatric and substance use disorders in men and<br />

women. Arch. Gen. Psychiatry 60, 929–937.<br />

Keyes, M. A., Malone, S. M., Elkins, I. J., Legrand, L. N., McGue, M., and Iacono, W. G. (2009). The<br />

enrichment study of the Minnesota twin family study: Increasing the yield of twin families at high<br />

risk for externalizing psychopathology. Twin Res. Hum. Genet. 12(5), 489–501.<br />

Kim-Cohen, J., Caspi, A., Taylor, A., Williams, B., Newcombe, R., Craig, I. W., and Moffitt, T. E.<br />

(2006). MAOA, maltreatment, and gene-environment interaction predicting children´s mental<br />

health: New evidence and a meta-analysis. Mol. Psychiatry 11(10), 903–913.<br />

Klump, K. L., Holly, A., Iacono, W. G., McGue, M., and Willson, L. E. (2000). Physical similarity<br />

and twin resemblance for eating attitudes and behaviors: A test of the equal environments<br />

assumption. Behav. Genet. 30(1), 51–58.<br />

Knafo, A., and Plomin, R. (2006). Prosocial behavior from early to middle childhood: Genetic and<br />

environmental influences on stability and change. Dev. Psychol. 42(5), 771–786.<br />

Krueger, R. F., Moffitt, T. E., Caspi, A., Bleske, A., and Silva, P. A. (1998). Assortative mating for<br />

antisocial behavior: Developmental and methodological implications. Behav. Genet. 28(3),<br />

173–186.<br />

Krueger, R. F., Hicks, B. M., Patrick, C. J., Carlson, S. R., Iacono, W. G., and McGue, M. (2002).<br />

Etiologic connections among substance dependence, antisocial behavior, and personality: Modeling<br />

the externalizing spectrum. J. Abnorm. Psychol. 111, 411–424.<br />

Krueger, R. F., Markon, K. E., Patrick, C. J., and Iacono, W. G. (2005). Externalizing psychopathology<br />

in adulthood: A dimensional-spectrum conceptualization and its implications for DSM–V.<br />

J. Abnorm. Psychol. 114(4), 537–550.<br />

Krueger, R. F., Markon, K. E., Patrick, C. J., Benning, S. D., and Kramer, M. D. (2007). Linking<br />

antisocial behavior, substance use, and personality: An integrative quantitative model of the adult<br />

externalizing spectrum. J. Abnorm. Psychol. 116(4), 645–666.<br />

Lahey, B. B., Applegate, B., Waldman, I. D., Loft, J. D., Hankin, B. L., and Rick, J. (2004). The<br />

structure of child and adolescent psychopathology: Generating new hypotheses. J. Abnorm.<br />

Psychol. 113, 358–385.<br />

Leventhal, T., and Brooks-Gunn, J. (2000). The neighbourhoods they live in: The effects of<br />

neighbourhood residence on child and adolescent outcomes. Psychol. Bull. 126, 309–337.<br />

Lewis, D. O., Yeager, C. A., Gidlow, B., and Lewis, M. (2001). Six adoptees who murdered:<br />

Neuropsychiatric vulnerabilities and characteristics of biological and adoptive parents. J. Am.<br />

Acad. Psychiatry Law 29(4), 390–397.<br />

Linnoila, M., Virkkunen, M., Scheinin, M., Nuutila, A., Rimon, R., and Goodwin, F. K. (1983). Low<br />

cerebrospinal fluid 5-hydroxyindoleacetic acid concentration differentiates impulsive from nonimpulsive<br />

violent behavior. Life Sci. 33, 2609–2614.<br />

Lipsey, M. W., Wilson, D. B., Cohen, M. A., and Derzon, J. H. (1997). Is there a causal relationship<br />

between alcohol use and violence A synthesis of evidence. Recent Dev. Alcohol. 13, 245–282.


8. Human Aggression Across the Lifespan 211<br />

Loehlin, J. C. (1992). Genes and environment in personality development. Newbury Park SAGE, CA.<br />

Loehlin, J. C. (2010). Is there an active gene-environment correlation in adolescent drinking<br />

behavior Behav. Genet. 40(4), 447–451.<br />

Maes, H. H. M., Neale, M. C., Kendler, K., Hewitt, J. K., Silberg, J. L., Foley, D. L., Meyer, J. M.,<br />

Rutter, M., Simonoff, E., Pickles, A., and Eaves, L. J. (1998). Assortative mating for major<br />

psychiatric diagnosis in two population-based samples. Psychol. Med. 28, 1389–1401.<br />

Maes, H. H., Silberg, J. L., Neale, M. C., and Eaves, L. J. (2007). Genetic and cultural transmission of<br />

antisocial behavior: An extended twin parent model. Twin Res. Hum. Genet. 10(1), 136–150.<br />

Mather, K., and Jinks, J. L. (1982). Biometrical Genetics: The Study of Continuous Variation.<br />

Chapman & Hall, London.<br />

Mathew, K. A., and Angulo, J. (1980). Measurement of the type A behavior pattern in children:<br />

Assessment of children’s competitiveness, impatience-anger, and aggression. Child Dev. 51,<br />

466–475.<br />

Mauger, P. A. (1980). Interpersonal Behavior Survey (IBS) Manual. Western Psychological Services,<br />

Los Angeles.<br />

McGue, M., Bacon, S., and Lykken, D. T. (1993). Personality stability and change in early<br />

adulthood: A behavioral genetic analyses. Dev. Psychol. 29, 96–109.<br />

Meininger, J. C., Hayman, L. L., Coates, P. M., and Gallagher, P. (1988). Genetics or environment<br />

Type A behavior and cardiovascular risk factors in twin children. Nurs. Res. 37(6), 341–346.<br />

Merton, R. K. (1957). Social Theory and Social Structure. The Free Press, New York.<br />

Miles, D. R., and Carey, G. (1997). Genetic and environmental architecture of human aggression.<br />

J. Pers. Soc. Psychol. 72(1), 207–217.<br />

Milgram, S. (1963). Behavioral study of obedience. J. Abnorm. Soc. Psychol. 67, 371–378.<br />

Moffitt, T. (2002). Teen-aged mothers in contemporary Britain & the E-risk Study Team (39<br />

authors). J. Child Psychol. Psychiatry 43, 727–742.<br />

Moffitt, T. E., Caspi, A., Rutter, M., and Silva, P. (2001). Sex Differences in Antisocial Behaviour:<br />

Conduct Disorder, Delinquency and Violence in the Dunedin Longitudinal Study. Cambridge<br />

<strong>University</strong> Press, Cambridge.<br />

Moilanen, I., Linna, S.-L., Kumpulainen, K., Tamminen, K., Piha, J., and Almqvist, F. (1999). Are<br />

twins’ behavioural 7emotional problems different from singletons’ Eur. Child Adolesc. Psychiatry<br />

8, 62–67.<br />

Narusyte, J., Andershed, A. K., Neiderhiser, J. M., and Lichtenstein, P. (2006). Aggression as a<br />

mediator of genetic contributions to the association between negative parent–child relationships<br />

and adolescent antisocial behavior. Eur. Child Adolesc. Psychiatry 16(2), 128–137.<br />

Neale, M. C., and Cardon, L. R. (1992). Methodology for Genetic Studies of Twins and Families.<br />

Kluwer Academic Publications, Dordrecht, The Netherlands.<br />

Neiderhiser, J. M., Reiss, D., Pedersen, N. L., Lichtenstein, P., Spotts, E. L., Hansson, K.,<br />

Cederblad, M., and Elthammar, O. (2004). Genetic and environmental influences on mothering<br />

of adolescents: A comparison of two samples. Dev. Psychol. 40(3), 335–351.<br />

Nilsson, K. W., Sjöberg, R. L., Damberg, M., Leppert, J., Ohrvik, J., Alm, P. O., Lindström, L., and<br />

Oreland, L. (2006). Role of monoamine oxidase A genotype and psychosocial factors in male<br />

adolescent criminal activity. Mol. Psychiatry 10(11), 903–913.<br />

Niv, S., and Baker, L. A. (2010). Genetic marker for antisocial behavior. In “The Origins of<br />

Antisocial Behavior: A Developmental Perspective” (C. Thomas and K. Pope, eds.). Oxford<br />

<strong>University</strong> Press, New York.<br />

Oades, R. D., Lasky-Su, J., Christiansen, H., Faraone, S. V., Sonuga-Barke, E. J. S., Banaschewski, T.,<br />

Chem, W., Anney, R. J. L., Buitelaar, J. K., Ebstein, R. P., Franke, B., Gill, M., et al. (2008). The<br />

influence of serotonin and other genes and impulsive behavioral aggression and cognitive impulsivity<br />

in children with attention-deficit/hyperactivity disorder (ADHD): Finding from a familybased<br />

association test (FBAT) analysis. Behav. Brain Funct. 48, 4–48.


212 Tuvblad and Baker<br />

O’Connor, M., Foch, T., Sherry, T., and Plomin, R. (1980). A twin study of specific behavioral<br />

problems of socialization as viewed by parents. J. Abnorm. Child Psychol. 8(2), 189–199.<br />

O’Connor, T. G., Deater-Deckard, K., Fulker, D., Rutter, M., and Plomin, R. (1998). Genotypeenvironment<br />

correlations in late childhood and early adolescence: Antisocial behavioral problems<br />

and coercive parenting. Dev. Psychol. 34, 970–981.<br />

Olweus, D. (1989). Prevalence and incidence in the study of antisocial behavior: Definitions and<br />

measurements. In “Cross-National Research in Self-Reported Crime and Delinquency”<br />

(M. W. Klein, ed.). Kluwer Academic Publishers, Dordrecht.<br />

Owen, D. R., and Sines, J. O. (1970). Heritability of personality in children. Behav. Genet. 1(3),<br />

235–248.<br />

Parker, T. (1989). Television Viewing and Aggression in Four and Seven Year Old Children.<br />

Boulder, Boulder, CO.<br />

Pfohl, B., Blum, N., and Zimmerman, M. (1997). Structured Interview for DSM-IV Personality:<br />

SIDP-IV. American Psychiatric Press, Washington, DC.<br />

Plomin, R., DeFries, J. C., McClearn, G. E., and McGuffin, P. (2001). Behavioral Genetics. Worth<br />

Publisher, United <strong>State</strong>s of America.<br />

Polderman, T. J., Posthuma, D., De Sonneville, L. M., Verhulst, F. C., and Boomsma, D. I. (2006).<br />

Genetic analyses of teacher ratings of problem behavior in 5-year-old twins. Twin Res. Hum.<br />

Genet. 9(1), 122–130.<br />

Posthuma, D., Beem, A. L., de Geus, E. J. C., van Baal, C. M., von Hjelmborg, J. B., Iachine, I., and<br />

Boomsma, D. I. (2003). Theory and practice in quantitative genetics. Twin Res. Hum. Genet.<br />

6(5), 361–376.<br />

Pulikkined, L., Kaprio, J., and Rose, R. J. (1999). Peers, teachers, and parents as assessors of the<br />

behavioural and emotional problems of twins and their adjustment: The Multidimensional Peer<br />

Nomination Inventory. Twin Res. Hum. Genet. 2, 274–285.<br />

Rahe, R. H., Hervig, L., and Rosenman, R. H. (1978). Heritability of type A behavior. Psychosom.<br />

Med. 40(6), 478–486.<br />

Raine, A. (2002). Biosocial studies of antisocial and violent behavior in children and adults: A<br />

review. J. Abnorm. Child Psychol. 30(4), 311–326.<br />

Raine, A., Brennan, P. A., and Mednick, S. A. (1997). Interaction between birth complications and<br />

early maternal rejection in predisposing individuals to adult violence: Specificity to serious, earlyonset<br />

violence. Am. J. Psychiatry 154(9), 1265–1271.<br />

Raine, A., Dodge, K., Loeber, R., Gatzke-Kopp, L., Lynam, D. R., Reynolds, C., Stouthamer-<br />

Loeber, M., and Liu, J. (2006). The reactive-proactive aggression questionnaire: Differential<br />

correlates of reactive and proactive aggression in adolescent boys. Aggress. Behav. 32, 159–171.<br />

Reif, A., Rosler, M., Freitag, C. M., Schneider, M., Eujen, A., Kissling, C., Wensler, D., Jacob, C. P.,<br />

Retz-Junginger, P., Thome, J., Lesch, K.-P., and Retz, W. (2007). Nature and nurture predispose to<br />

violent behavior: Serotonergic genes and adverse childhood environment. Neuropsychopahrmacology<br />

32, 2375–2383.<br />

Retz, W., Rosler, M., Supprian, T., Retz-Junginger, P., and Thome, J. (2003). Dopamine D3 receptor<br />

gene polymorphism and violent behavior: Relation to impulsiveness and ADHD-related psychopathology.<br />

J. Neural Transm. 110, 561–572.<br />

Retz, W., Freitag, C. M., Retz-Junginger, P., Wenzler, D., Schneider, M., Kissling, C., et al. (2008). A<br />

functional serotonin transporter promoter gene polymorphism increases ADHD symptoms in<br />

delinquents: Interaction with adverse childhood environment. Psychiatry Res. 158, 123–131.<br />

Rhee, S. H., and Waldman, I. D. (2002). Genetic and environmental influences on antisocial<br />

behavior: A meta-analysis of twin and adoption studies. Psychol. Bull. 128, 490–529.<br />

Rowe, D. C. (2003). Assessing genotype-environment interactions and correlations in the postgenomic<br />

era. In “Behavioral Genetics in the Postgenomic Era” (R. Plomin, J. C. DeFries, I. W. Craig,<br />

and P. McGuffin, eds.), pp. 71–99. American Psychological Association, Washington, DC.


8. Human Aggression Across the Lifespan 213<br />

Rowe, D. C., Almeida, D. M., and Jacobson, K. C. (1999). School con<strong>text</strong> and genetic influences on<br />

aggression in adolescence. Psychol. Sci. 10(3), 277–280.<br />

Rushton, J. P., Fulker, D. W., Neale, M. C., Nias, D. K., and Eysenck, H. J. (1986). Altruism and<br />

aggression: The heritability of individual differences. J. Pers. Soc. Psychol. 50(6), 1192–1198.<br />

Rutter, M. (2006). Genes and behavior. Nature-nurture interplay explained. Blackwell Publishing<br />

Ltd, Oxford, UK.<br />

Rutter, M., and Redshaw, J. (1991). Annotation: Growing up as a twin: Twin-singleton differences in<br />

psychological development. J. Child Psychol. Psychiatry 32(6), 885–895.<br />

Rutter, M., Giller, H., and Hagell, A. (1998). Antisocial Behavior by Young People. Cambridge<br />

<strong>University</strong> Press, Cambridge, UK.<br />

Rutter, M., Caspi, A., and Moffitt, T. E. (2003). Using sex differences in psychopathology to study<br />

causal mechanisms: Unifying issues and research strategies. J. Child Psychol. Psychiatry 44(8),<br />

1092–1115.<br />

Scarr, S. (1966). Genetic factors in activity motivation. Child Dev. 37(3), 663–673.<br />

Scarr, S., and McCartney, K. (1983). How people make their own environments: A theory of<br />

genotype-environment effects. Child Dev. 54, 425–435.<br />

Schmitz, S., Fulker, D. W., and Mrazek, D. A. (1995). Problem behavior in early and middle<br />

childhood: An initial behavior genetic analysis. J. Child Psychol. Psychiatry 36(8), 1443–1458.<br />

Schwartz, D., Dodge, K., Coie, J. D., Hubbard, J. A., Cillessen, A. H. N., Lemerise, E. A., and<br />

Bateman, H. (1998). Social-cognitive and behavioral correlates of aggression and victimization in<br />

boys’ play groups. J. Abnorm. Child Psychol. 26(6), 431–440.<br />

Shaffer, D., Fisher, P., Lucas, C., and Comer, J. (2000). Diagnostic Interview Schedule for Children-<br />

DISC-IV Scoring Manual. Columbia <strong>University</strong>, NY.<br />

Silva, P. A., and Stanton, W. R. (1996). From child to adult: The Dunedin Multidisciplinary Health<br />

and Development Study. Oxford <strong>University</strong> Press Inc., New York, NY.<br />

Simonoff, E., Pickles, A., Meyer, J., Silberg, J., Maes, H., Loeber, M., Rutter, M., Hewitt, J. K., and<br />

Eaves, L. (1997). The Virginia twin study of adolescent behavioral development. Arch. Gen.<br />

Psychiatry 54, 801–808.<br />

Simonoff, E., Pickles, A., Meyer, J., Silberg, J., and Maes, H. (1998). Genetic and environmental<br />

influences on subtypes of conduct disorder behavior in boys. J. Abnorm. Child Psychol. 26(6),<br />

495–509.<br />

Sines, J. O., Pauker, J. D., and Sines, L. K. (1966). The Development of an Objective, Non-verbal<br />

Personality Test for Children. Midwestern Psychological Association, Chicago.<br />

Sjöberg, R. L., Nilsson, K. W., Wargelius, H.-L., Leppert, J., Lindström, L., and Oreland, L. (2007).<br />

Adolescent girls and criminal activity: Role of MAOA-LPR genotype and psychosocial factors.<br />

Am. J. Med. Genet. B Neuropsychiatr. Genet. 144B, 159–164.<br />

Tackett, J. L., Waldman, I. D., and Lahey, B. B. (2009). Etiology and measurement of relational<br />

aggression: A multi-informant behavior genetic investigation. J. Abnorm. Psychol. 118(4),<br />

722–733.<br />

Taylor, A. (2004). The consequences of selective participation on behavioral-genetic findings:<br />

evidence from simulated and real data. Twin Res. 7(5), 485–504.<br />

Taylor, J., McGue, M., and Iacono, W. G. (2000). Sex differences, assortative mating, and cultural<br />

transmission effects on adolescent delinquency: A twin family study. J. Child Psychol. Psychiatry<br />

41(4), 433–440.<br />

Tellegen, A. (2011). Manual for the Multidimensional Personality Questionnaire <strong>University</strong> of<br />

Minnesota Press, Minneapolis.<br />

Tuvblad, C., Grann, M., and Lichtenstein, P. (2006). Heritability for adolescent antisocial behavior<br />

differs with socioeconomic status: Gene-environment interaction. J. Child Psychol. Psychiatry<br />

47(7), 734–743.


214 Tuvblad and Baker<br />

Tuvblad, C., Raine, A., Zheng, M., and Baker, L. A. (2009). Genetic and environmental stability<br />

differs in reactive and proactive aggression. Aggress. Behav. 35(6), 437–452.<br />

Tuvblad, C., Narusyte, J., Grann, M., Sarnecki, J., and Lichtenstein, P. (2011). The genetic and<br />

environmental etiology of antisocial behavior, from early adolescence to emerging adulthood.<br />

Behav. Genet. 41(5), 629–640.<br />

van Beijsterveldt, C. E. M., Bartels, M., Hudziak, J. J., and Boomsma, D. (2003). Causes of stability of<br />

aggression from early childhood to adolescence: A longitudinal genetic analysis in Dutch twins.<br />

Behav. Genet. 33, 591–605.<br />

van den Oord, E. J. C. G., Boomsma, D., and Verhulst, F. C. (1994). A study of problem behaviors in<br />

10- to 15-year-old biologically related and unrelated international adoptees. Behav. Genet. 24,<br />

193–205.<br />

van den Oord, E. J. C. G., Koot, H. M., Boomsma, D. I., Verhulst, F. C., and Orlebeke, J. F. (1995).<br />

A twin-singleton comparison of problem behaviour in 2–3-year-olds. J. Child Psychol. Psychiatry<br />

36(3), 449–458.<br />

van den Oord, E. J., Verhulst, F. C., and Boomsma, D. I. (1996). A genetic study of maternal and<br />

paternal ratings of problem behaviors in 3-year-old twins. J. Abnorm. Psychol. 105(3), 349–357.<br />

van der Valk, F. C., Verhulst, F. C., Stroet, T. M., and Boomsma, D. (1998). Quantitative genetic<br />

analysis of internalizing and externalizing problems in a large sample of 3-year-old twins. Twin<br />

Res. Hum. Genet. 1, 25–33.<br />

Vierikko, E., Pulkkinen, L., Kaprio, J., and Rose, R. J. (2004). Genetic and environmental influences<br />

on the relationship between aggression and hyperactivity-impulsivity as rated by teachers and<br />

parents. Twin Res. Hum. Genet. 7(3), 261–274.<br />

von der Pahlen, B., Santtila, P., Johansson, A., Varjonen, M., Jern, P., Witting, K., and Kenneth<br />

Sandnabba, N. (2008). Do the same genetic and environmental effects underlie the covariation of<br />

alcohol dependence, smoking, and aggressive behaviour Biol. Psychol. 78(3), 269–277.<br />

Waldman, I. D., Rowe, D. C., Abramowitz, A., Kozel, S. T., Mohr, J. H., Sherman, S. L.,<br />

Cleveland, H. H., Sanders, M. L., Gard, J. M. C., and Stever, C. (1998). Association and linkage<br />

of the dopamine transporter gene and attention-deficit hyperactivity disorder in children: Heterogeneity<br />

owing to diagnostic subtype and severity. Am. J. Hum. Genet. 63, 1767–1776.<br />

White, H. R., Xie, M., Thompson, W., Loeber, R., and Stouthamer-Loeber, M. (2001). Psychopathology<br />

as a predictor of adolescent drug use trajectories. Psychol. Addict. Behav. 15(3), 210–218.<br />

Widom, C. S., and Brzustowicz, L. M. (2006). MAOA and the “cycle of violence”: Childhood abuse<br />

and neglect, MAOA genotype, and risk for violent and antisocial behavior. Biol. Psychiatry 1(1),<br />

60.<br />

Xian, H., Scherrer, J. F., Eisen, S. A., True, W. R., Heath, A. C., Goldberg, J., Lyons, M. J., and<br />

Tsuang, M. T. (2000). Self-Reported zygosity and the equal-environments assumption for psychiatric<br />

disorders in the Vietnam Era Twin Registry. Behav. Genet. 30(4), 303–310.<br />

Young, S. E., Stallings, M. C., Corley, R. P., Krauter, K. S., and Hewitt, J. K. (2000). Genetic and<br />

environmental influences on behavioral disinhibition. Am. J. Med. Genet. 96, 684–695.


9<br />

Perinatal Risk Factors in<br />

the Development of<br />

Aggression and Violence<br />

Jamie L. LaPrairie, Julia C. Schechter, Brittany A. Robinson,<br />

and Patricia A. Brennan<br />

Department of Psychology, Emory <strong>University</strong>, Atlanta, Georgia, USA<br />

I. Introduction<br />

II. The Neurobiological and Psychophysiological Systems Involved in<br />

the Regulation of Aggression and Violence<br />

A. Types of aggressive behavior<br />

B. Neurobiological bases of aggression and violence<br />

C. Neurochemical signals of aggression and violence<br />

D. Hormones<br />

E. Autonomic response measures<br />

F. Electro cortical response measures<br />

III. Perinatal Factors Related to the Development of Aggression<br />

A. Birth complications<br />

B. Preterm birth and low birth weight<br />

C. Prenatal drug and alcohol exposure<br />

D. Smoking<br />

E. Maternal psychological stress<br />

F. Environmental con<strong>text</strong><br />

IV. Genetic Contributions<br />

A. Genetic factors as explanatory<br />

B. Gene by environment (G E) interactions<br />

C. The role of epigenetics<br />

V. Conclusions<br />

References<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00004-6


216 LaPrairie et al.<br />

ABSTRACT<br />

Over the past several decades, the relative contribution of both environmental<br />

and genetic influences in the development of aggression and violence has been<br />

explored extensively. Only fairly recently, however, has it become increasingly<br />

evident that early perinatal life events may substantially increase the vulnerability<br />

toward the development of violent and aggressive behaviors in offspring<br />

across the lifespan. Early life risk factors, such as pregnancy and birth complications<br />

and intrauterine exposure to environmental toxins, appear to have a<br />

profound and enduring impact on the neuroregulatory systems mediating violence<br />

and aggression, yet the emergence of later adverse behavioral outcomes<br />

appears to be both complex and multidimensional. The present chapter reviews<br />

available experimental and clinical findings to provide a framework on perinatal<br />

risk factors that are associated with altered developmental trajectories leading to<br />

violence and aggression, and also highlights the genetic contributions in the<br />

expression of these behaviors. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

Aggressive behavior and violent offending are significant burdens not only to the<br />

individual but also to society at large; incurring costs upward of 2.3 million dollars<br />

per individual in the most severe cases in the United <strong>State</strong>s (Cohen et al., 2010).<br />

In contrast to popular belief, longitudinal epidemiological research indicates that<br />

the sudden onset of physical aggression in adolescence or adulthood is unusual<br />

(Tremblay, 2008). Rather, physically aggressive behaviors can already be detected<br />

by 12 months of age, and their frequency peaks by 2–4 years of age (Côté et al.,<br />

2006; Tremblay, 2008). Indeed 85% of children exhibit aggression and tantrums<br />

by age 2 (Potegal and Davidson, 2003). In the majority of children, the frequency<br />

of physical aggression gradually decreases toward the end of the preschool period,<br />

reflecting an increase in cognitive control of behavior, the acquisition of alternate<br />

problem-solving strategies, and the influence of socialization (Nagin, and<br />

Tremblay, 1999; Tremblay, 2008). A 60-year longitudinal study of juvenile delinquents<br />

concluded that very few show a lifespan high frequency of violent offending<br />

(Sampson and Laub, 2003), and among those who do express chronic physical<br />

aggression, impaired executive functioning is evident in adolescence and early<br />

childhood, even after controlling other cognitive deficits (Barker et al., 2007).<br />

Longitudinal follow-up studies of elementary school children with continued high<br />

levels of physical aggression demonstrate that they are at greater risk for numerous<br />

adjustment problems throughout their lifetime, including substance abuse, academic<br />

failure, antisocial behavior, suicide, depression, spouse abuse, and neglectful<br />

parenting (Broidy et al., 2003; Tremblay et al., 2004).


9. Perinatal Factors in the Development of Aggression 217<br />

The neurobiological substrates underlying the individual variability in<br />

the development of aggression and violence are many (Loeber and Pardini, 2008),<br />

and current research suggests that some of the most relevant risk factors for a<br />

trajectory of consistently high aggression are predicted by perinatal life events<br />

including pregnancy and birth complications and intrauterine exposure to tobacco,<br />

drugs, and alcohol (Brennan and Mednick, 1997; Cannon et al., 2002).<br />

Genetic studies of aggressive behavior also indicate that childhood aggression is<br />

highly heritable (Brendgen et al., 2005; Hicks et al., 2004). Together, these<br />

congenital factors appear to produce neuropsychological deficits in the nervous<br />

system of offspring, manifesting as subtle neurocognitive difficulties, altered temperament,<br />

delayed motor development, and hyperactivity (Moffitt and Caspi,<br />

2001). The two most commonly characterized developmental trajectories for<br />

aggression and violence are: (1) an early-onset persistent offender and (2) a lateonset<br />

adolescent-limited offender (Moffitt, 1993; Patterson et al., 1989). In the<br />

early-onset offender, behavioral problems typically manifest early in life, develop<br />

into serious juvenile delinquency during adolescence, and ultimately evolve into a<br />

long-term adult history of criminal behavior. Alternatively, late-onset adolescentlimited<br />

individuals typically do not begin offending until middle to late adolescence<br />

and cease violent and criminal behavior by their mid-1920s (Moffitt, 1993).<br />

These two distinct trajectories are associated with substantial variation in severity,<br />

chronicity, etiology, and prognosis. In fact, adolescent-limited offending can be<br />

considered normative and functional, given the developmental demands on adolescents<br />

in modern society (Moffitt, 1993). Adolescent-limited offending may also<br />

be explained by normative changes in brain development in postpubertal children.<br />

Life-course-persistent offending, in contrast, is not socially sanctioned, functional,<br />

or reflective of normative changes in brain development. Instead, this type of<br />

offending is likely caused by genetic and perinatal factors which lead to deficits in<br />

neurocognitive and neurophysiological functioning throughout the life course.<br />

Prior to reviewing the current research on perinatal factors and violence, it is<br />

important to briefly describe the neurophysiological processes that are involved in<br />

the regulation of aggression and violence. Then we will describe how these factors<br />

may mediate that relationship between perinatal risk and aggressive outcomes.<br />

Finally, we will attempt to highlight what is known about the genetic contributions<br />

to this developmental process.<br />

II. THE NEUROBIOLOGICAL AND PSYCHOPHYSIOLOGICAL SYSTEMS<br />

INVOLVED IN THE REGULATION OF AGGRESSION AND VIOLENCE<br />

The central nervous system (CNS) and autonomic nervous system (ANS)<br />

maintain homeostasis and monitor physiological responses in humans from the<br />

perinatal period throughout the lifespan. These systems are also involved in the


218 LaPrairie et al.<br />

mediation of aggressive, violent, and antisocial behavior. Although neurophysiological<br />

systems have been the focal point of studies on aggression for decades<br />

(Scarpa and Raine, 1997), significant technological advances over the past<br />

20 years have allowed researchers to advance their investigation of the biological<br />

basis underlying violent and aggressive behaviors. Indeed, researchers are now<br />

able to ask questions concerning the neural underpinnings of aggression and how<br />

the interactions between genes and environment may result in later violence;<br />

ultimately leading to a more complete picture of the development of aggression<br />

and violence throughout the lifespan.<br />

A. Types of aggressive behavior<br />

Two forms of aggression are present in both experimental animal and human<br />

models. Throughout the animal literature, predatory aggression has been differentiated<br />

from defensive aggression (Scarpa and Raine, 1997). Similarly, human<br />

studies often discriminate between instrumental/proactive aggression and hostile/reactive<br />

or impulsive aggression (Crick and Dodge, 1996; Nelson and<br />

Trainor, 2007). Reactive aggression is characterized as aggressive behavior occurring<br />

in the con<strong>text</strong> of anger and high emotionality. It is often more impulsive,<br />

less controlled, and occurring in reaction to a provocation or frustration (Scarpa<br />

and Raine, 1997). Instrumental aggression is qualified as being more goal-oriented<br />

and relatively nonemotional. Studies suggest that this latter type of aggression<br />

is regulated by higher cortical systems rather than brain regions (i.e., limbic<br />

systems) associated with impulsiveness (Nelson and Trainor, 2007).<br />

Of note, instrumental aggression is characteristic of psychopathy, a<br />

subtype of aggression often associated with particularly low levels of physiological<br />

arousal (Raine, 2002a). While psychopathological disorders are outside the<br />

scope of this chapter, understanding that individuals may display different types<br />

of aggressive behavior is essential to conceptualizing physiological research<br />

findings and the development of violence, in general.<br />

B. Neurobiological bases of aggression and violence<br />

Recent technological advances have allowed scientists to observe parallel neurochemical<br />

and anatomical correlates that are activated during aggression in<br />

both human and nonhuman animals (Nelson and Trainor, 2007). Neurobiological<br />

literature regarding aggression appears to focus on substrates that are implicated<br />

in either the expression of aggressive behavior or inhibitory control of<br />

aggression (Siegel and Victoroff, 2009). Below is a brief synthesis of the animal<br />

and human literature that pertains to the neurobiological systems that have been<br />

implicated in the development and modulation of aggression and violence.


1. Amygdala<br />

9. Perinatal Factors in the Development of Aggression 219<br />

The amygdala is part of the limbic system and is considered to play a central role in<br />

both emotional regulation (Joseph, 1999) and fear conditioning (Susman, 2006).<br />

Lesion studies in animals have been critical to the understanding of the relationship<br />

between the amygdala and aggression. Lesioning of the medial amygdala reduces<br />

aggression in rats (Kruk, 1992). Male rhesus monkeys with amygdalar lesions display<br />

significant increases in aggressive behavior in a group setting (Machado and<br />

Bachevalier, 2006), while decreases in aggressive behavior are observed when<br />

animals are tested within a dyad (Emery et al., 2001). An explanation for these<br />

divergent findings may be that reintroduction into a group is a more fearful situation,<br />

thus leading to increases in amygdalar responsiveness (Nelson and Trainor, 2007).<br />

Functional abnormalities in the amygdala have also been noted in human<br />

studies in childhood, adolescence (Marsh et al., 2008; Sterzer et al., 2005), and<br />

adulthood (Veit et al., 2002); however, these studies have provided mixed results.<br />

The posteroventral medial amygdala appears to be involved in the regulation of<br />

reactive aggression, as in defensive situations, while the posterodorsal medial amygdala<br />

appears to be associated with instrumental or offensive situations (Swanson,<br />

2000). Coccaro et al. (2007) found that amygdalar activation is positively associated<br />

with scores on the Lifetime History of Aggression scale for adults with intermittent<br />

explosive disorder and healthy controls. Notably, only individuals with intermittent<br />

explosive disorder display increased activation of the amygdala in response to angry<br />

faces (Coccaro et al., 2007). Conversely, Sterzer et al. (2005) found a negative<br />

correlation between aggression and the left amygdala in response to negative affective<br />

images in a group of boys diagnosed with conduct disorder (CD). However, the<br />

inverse relationship (i.e., positive correlation between amygdala activation and<br />

aggression) was found in youth with comorbid anxiety and depression, symptoms<br />

often found to be associated with CD (Loeber et al., 2000). Reduced responsiveness of<br />

the amygdala in response to fearful faces has been suggested to reflect impairment in<br />

the processing of distress cues (Marsh et al., 2008), which may result in a lack of<br />

empathy and increases in instrumental aggressive behavior (Blair, 2001). One<br />

explanation for these discrepant findings across samples is that aggressive individuals<br />

may appear hyporesponsive when faced with detecting threat or distress, but hyperresponsive<br />

to distressful situations that can lead to aggression (Decety et al., 2009).<br />

Further, these mixed findings highlight that the amygdala’s role in the mediation of<br />

aggression may be based largely on the type of aggression expressed.<br />

2. Anterior cingulate cortex<br />

The anterior cingulate cortex (ACC) is also a part of the brain’s limbic system<br />

and has been implicated in emotional and cognitive processing (Bush et al.,<br />

2000). Lesions of the ACC have led to a range of outcomes, including


220 LaPrairie et al.<br />

inattention, dysregulation of autonomic functions, and emotional instability<br />

(Kennard, 1954; Tow and Whitty, 1953). ACC lesions as a treatment for<br />

affective disorders in humans have produced decreased distress and emotional<br />

liability (Corkin, 1979). More recently, deactivation in the dorsal ACC, an area<br />

associated with cognitive monitoring and behavioral regulation (Bush et al.,<br />

2000), has been noted in aggressive youth compared to controls (Sterzer et al.,<br />

2005). Reduced activation in the ACC is also associated with “novelty seeking”<br />

(Cloninger, 1987), a dimension of temperament encompassing a quick-tempered<br />

personality and high impulsivity (Stadler et al., 2007). Thus, reduced activation<br />

of the ACC may be a connection between temperament, behavior, and emotion<br />

processing (Sterzer et al., 2005).<br />

3. Prefrontal cortex<br />

The prefrontal cortex (PFC) modulates subcortical behavior (Siever, 2008),<br />

specifically by inhibiting connections between the amygdala and hypothalamus,<br />

thereby resulting in increased aggression (Davidson, 2000). Prefrontal lesions in<br />

humans as a result of tumors, trauma, or metabolic disturbances have been<br />

instrumental in illustrating the role of the PFC in aggressive behavior (Siever,<br />

2008). The well-known case of Phineas Gage, a stable and dependable railroad<br />

worker who became irritable, angry, and showed poor judgment following an<br />

accident in which a rod entered his skull at the frontal cortex, is a prime example<br />

of the critical role of the PFC in monitoring aggression.<br />

Imaging studies exploring brain functioning have further elucidated the<br />

role of the frontal cortices in modulation of aggression. Individuals with lowerthan-average<br />

baseline activity in the frontal cortex demonstrate higher levels of<br />

reactive or impulsive aggression (Coccaro and Kavoussi, 1997; Soloff et al., 2003;<br />

Volkow et al., 1995). Moreover, the ventromedial prefrontal cortex (vmPFC) has<br />

been specifically implicated in the calculation of reward expectation (Elliott and<br />

Deakin, 2005), and increased activation is observed in the vmPFC when errors<br />

are made during a reversal learning task (Finger et al., 2008). Reversal learning<br />

tasks are designed to frustrate participants and measure their ability to adjust<br />

behavior in response to changing reinforcement (i.e., avoid frustration; Rolls<br />

et al., 1994), a deficit observed in individuals with psychopathic traits (Blair<br />

et al., 2001; Budhani and Blair, 2005). Violations of reward expectations (i.e.,<br />

expecting but not receiving reinforcement) have been linked to frustration and<br />

reactive aggression (Berkowitz, 1989), which may be a result of not achieving an<br />

expected reward (Sterzer et al., 2005). Thus, increased vmPFC responses to<br />

violations of expectations may indeed be associated with an increased risk of<br />

frustration and subsequent aggressive or violent behaviors (Blair, 2010).


9. Perinatal Factors in the Development of Aggression 221<br />

Experimental animal studies indicate a connection between the orbitofronal<br />

cortex (OFC) and aggressive behavior, particularly with regard to the<br />

interpretation of social cues and behavioral responses in social con<strong>text</strong>s (Nelson<br />

and Trainor, 2007). OFC lesions in male rhesus monkeys increase aggression in<br />

dominant but not in subordinate animals (Machado and Bachevalier, 2006).<br />

Butter and Snyder (1972) observed similar results, but these effects diminished<br />

over several months. In humans, structural abnormalities in the OFC, such as<br />

reduced levels of gray-matter volume in youth with CD (Huebner et al., 2008)<br />

and early brain damage in this region have also been associated with conduct<br />

problems (Anderson et al., 1999). Overall, dysfunction in the prefrontal regions<br />

of the brain appears to underlie impaired regulation of affective responses and<br />

reduced inhibition of aggression (Davidson, 2000).<br />

4. Hypothalamus<br />

The hypothalamus is another critical brain structure involved in the development<br />

of aggression. Research with nonhuman primates suggests that electrical<br />

stimulation of the ventromedial hypothalamus is linked to vocal threats and<br />

aggressive behaviors in marmosets (Lipp and Hunsperger, 1978), while lesions of<br />

the anterior hypothalamus reduce vocal threats toward a male intruder (Lloyd<br />

and Dixson, 1988). Electrical stimulation of the anterior hypothalamus increases<br />

vocalizations in rhesus monkeys (Robinson, 1967) and aggression toward insubordinate<br />

male rhesus monkeys (Alexander and Perachio, 1973). Similar findings<br />

have been found with electrical stimulation of the hypothalamus in male rats<br />

(Kruk, 1992) and cats (Siegel and Victoroff, 2009). In humans, the frontal cortex<br />

inhibits circuits in the hypothalamus that increase aggression (Davidson, 2000).<br />

During a period in the mid-twentieth century, electrolytic lesions of the hypothalamus<br />

were used as a treatment for “excessive aggression” (Heimburger et al.,<br />

1966). Although conclusions from such studies should be interpreted cautiously<br />

for both methodological and ethical reasons (Scarpa and Raine, 1997), these<br />

lesions were found to inhibit aggression in humans.<br />

C. Neurochemical signals of aggression and violence<br />

Specific signaling molecules have provided additional information about neural<br />

circuits underlying aggression. As experimental research has turned to the brain<br />

for answers regarding the development of aggression and violence, the neurochemistry<br />

of aggressive behavior has also come to the forefront. In general,<br />

neurotransmitters in the brain either increase or inhibit aggressive behavior.<br />

Below is a brief review; a more extensive review of this area of research can be<br />

found in a separate chapter in this volume.


222 LaPrairie et al.<br />

1. Neurotransmitters-serotonin<br />

Given the connection between emotion, cognition, and aggression, it seems<br />

fitting that many studies have focused their investigation on serotonergic neurotransmission,<br />

a system predominantly involved in the regulation of emotional<br />

states. Serotonin (5-HT) receptors in specific regions of the brain, such as the<br />

OFC and ACC, are involved in the modulation and suppression of aggressive<br />

behavior (Siever, 2008). Moreover, low levels of 5-HT are associated with<br />

increased aggression in humans (Chiavegatto et al., 2001) and nonhuman primates<br />

(Higley et al., 1992). Many studies have investigated the role of 5-HT in<br />

neuronal functioning and aggression. For example, Frankle et al. (2005) reported<br />

reduced 5-HT transporter distribution in the ACC of patients with aggressive<br />

personality disorder compared to healthy controls. Reduced prefrontal activation<br />

was observed in response to a serotonergic releasing agent (d, 1-fenfluramine) in<br />

individuals with impulsive aggression, such as those diagnosed with borderline<br />

personality disorder (Soloff et al., 2003), and in depressed patients with a history<br />

of suicidal behavior (Mann et al., 1992). Using positron emission tomography<br />

(PET) technology, Parsey et al. (2002) found a negative association between<br />

scores on the Lifetime History of Aggression scale and 5-HT receptor-binding in<br />

the PFC and amygdala. Further, individuals with high levels of impulsive aggression<br />

display reduced activation in the PFC (Coccaro and Kavoussi, 1997).<br />

Further, New et al. (2004) found that individuals with borderline personality<br />

disorder, who underwent 12 weeks of selective serotonin reuptake inhibitor<br />

(SSRI) treatment, increased baseline PFC activation, which was negatively<br />

correlated with ratings of aggression.<br />

2. Neurotransmitters-dopamine<br />

Dopamine (DA) has been implicated in the initiation and exhibition of aggression<br />

(de Almeida et al., 2005), yet its precise role remains unclear. Ferrari et al.<br />

(2003) found that rats can be conditioned to increase dopamine secretion and<br />

decrease levels of 5-HT in anticipation of aggressive interactions, whereby DA<br />

and 5-HT levels in the nucleus accumbens were measured during and following a<br />

confrontation with another rat using microdialysis. Heart rate (HR) also<br />

increased 1 h prior to the regularly scheduled interactions. Antagonists of both<br />

D 1 and D 2 receptors appear to reduce aggression in male mice (de Almeida et al.,<br />

2005). Further, animal studies suggest that the activation of catecholaminergic<br />

brainstem neurons (e.g., ventral tegmental area) project to dopaminergic structures<br />

in the forebrain, such as the hypothalamus and the limbic system (e.g.,<br />

amygdala, hippocampus, PFC, and ACC). In humans, both proactive and reactive<br />

aggressions are associated with dopamine (Siegel and Victoroff, 2009). In<br />

clinical populations, decreased D 1 receptors have been observed in depressed


9. Perinatal Factors in the Development of Aggression 223<br />

individuals suffering from anger attacks (Dougherty et al., 2006). Psychopharmacological<br />

agents most frequently employ compounds that act on dopaminergic<br />

systems in the brain (McDougle et al., 1998). Risperdone, a D 2 antagonist,<br />

effectively reduces aggression in humans (Nelson and Trainor, 2007). Moreover,<br />

haloperidol, another D 2 antagonist, is used in the treatment of aggression in<br />

psychotic patients (Fitzgerald, 1999), of violent outbursts in adults with borderline<br />

personality disorder and dementia, and of aggression in children and adolescents<br />

(Beauchaine et al., 2000).<br />

3. Neurotransmitters-norepinephrine<br />

Norepinephrine (also known as noradrenaline) is a monoamine found in the<br />

ANS. It is associated with arousing situations and has been specifically cited in<br />

the development of both proactive and reactive aggression (Siegel and Victoroff,<br />

2009). Interestingly, a meta-analysis of central (cerebrospinal fluid) measures of<br />

norepinephrine found a negative association between norepinephrine and antisocial<br />

behavior (Raine, 1993). Plasma levels of norepinephrine are also associated<br />

with induced hostile behavior during experiments with healthy controls<br />

(Gerra et al., 1997). Pharmacological manipulation studies of noradrenaline<br />

levels and noradrenergic receptors suggest that this catecholamine facilitates<br />

the development of aggression (Miczek et al., 2002). Further evidence from the<br />

animal literature demonstrates that DA beta-hydroxylase knockout mice are<br />

unable to produce noradrenaline and display reduced levels of aggression, but<br />

normal levels of anxiety (Marino et al., 2005).<br />

D. Hormones<br />

Hormonal factors have long been studied in relation to aggressive behavior,<br />

particularly as scientists sought an explanation for the notable gender differences<br />

in the rates of violence. Androgens, and in particular testosterone, have received<br />

the most attention in this regard. Research on hormones and aggression has not<br />

demonstrated a one to one relationship between these factors. Instead, empirical<br />

findings suggest that the type of aggressive behavior and the structure and quality<br />

of the social environment are likely important moderators in the association<br />

between hormones and aggression.<br />

1. Testosterone<br />

A positive relationship between testosterone and aggression is well established in<br />

the animal literature, but less support for this association has emerged in humans<br />

(Archer, 1991). Injections of testosterone increase aggression in a variety of


224 LaPrairie et al.<br />

animals (Monoghan and Glickman, 1992), and aggression has been positively<br />

associated with territorial behavior in birds (Wingfield and Hahn, 1994). Moreover,<br />

testosterone increases the display of dominance behaviors in rhesus<br />

monkeys (Rose et al., 1971). Further, the castration of lizards (<strong>Green</strong>berg and<br />

Crews, 1983) and male mice (Vom Saal, 1983) leads to reduced aggression.<br />

While some research with humans indicates that testosterone is associated with<br />

anger and aggression (Olweus, 1986), evidence from the broader literature is<br />

mixed (Archer, 1991). Some studies have revealed positive relationships between<br />

testosterone and aggression, some report negative relationships, and no<br />

association is found in others (van Bokhoven et al., 2006). Specifically, testosterone<br />

measured in cerebrospinal fluid, serum, and saliva have been linked to<br />

chronic aggression (Ehrenkranz et al., 1974), violent crimes (Dabbs et al., 1987),<br />

antisocial personality disorder (Dabbs and Morris, 1990), and peer ratings of<br />

“toughness” (Dabbs et al., 1987). However, many studies have not replicated<br />

these results (Bain et al., 1987) and a metanalytic study of community and<br />

selected samples found only modest correlations (i.e., between 0.08 and 0.14)<br />

between testosterone and aggression (Archer et al., 2005). Mixed findings have<br />

similarly been observed in adolescents (Olweus et al., 1988; Susman et al., 1987).<br />

These inconsistent results may be partially attributable to social and developmental<br />

factors (Rowe et al., 2004), as well as other hormones, such as cortisol<br />

(Popma et al., 2007b).<br />

2. Cortisol<br />

Cortisol is the end product of the hypothalamic-pituitary-adrenal (HPA) axis,<br />

the human body’s stress response system, and is often used as a marker of stress<br />

responsiveness. Literature regarding the relationship between cortisol and aggression<br />

is mixed. Although numerous studies suggest that lower basal cortisol<br />

levels are associated with increased disruptive behaviors in males (Hawes et al.,<br />

2009; Popma et al., 2007a), other studies report the opposite, with CD and<br />

aggression being associated with higher levels of cortisol (Alink et al., 2008;<br />

Van Bokhoven et al., 2005). Still other studies have not established any link<br />

between cortisol and aggressive behavior (Alink et al., 2008; Scerbo and Kolko,<br />

1994; van Goozen et al., 2000). A significant amount of research has focused on<br />

an association between an underreactive HPA axis and aggressive behaviors.<br />

Theoretically, this underreactivity may be associated with reduced comprehension<br />

of distress cues, which has been related to reduced empathy and behavioral<br />

inhibition (Marsh et al., 2008). Lower levels of cortisol are also linked with<br />

reductions in fear responsiveness (Cima et al., 2008), which may lead to persistent<br />

aggressive behavior (McBurnett et al., 2000). In reviewing the divergent<br />

findings linking cortisol levels and aggression, Hawes et al. (2009) proposed two


9. Perinatal Factors in the Development of Aggression 225<br />

hormonal pathways to antisocial behavior—one which links stress exposure to<br />

high cortisol and aggression and the other which links low cortisol to aggression<br />

through callous-unemotional traits. In support of this proposal, a recent empirical<br />

study found that changes in cortisol levels in response to a stressor were<br />

positively associated with reactive (but not proactive) aggression in boys (Lopez-<br />

Duran et al., 2009).<br />

3. Oxytocin<br />

Recently, the hormone oxytocin has been investigated as a possible link in the<br />

development of aggression. Oxytocin is a hormone that is involved in trust and<br />

affiliative behaviors (Insel and Winslow, 1998; Young et al., 2001), thereby<br />

reduced oxytocin is thought to be associated with increased aggression. This<br />

hypothesis has been supported in the animal literature as oxytocin knockout<br />

mice exhibit increased aggressive behavior (Ragnauth et al., 2005). In humans,<br />

adults administered oxytocin intranasally are significantly better at identifying<br />

happy facial expressions compared to other expressions (Marsh et al., 2010).<br />

Oxytocin has also been found to reduce activation in the amygdala (Kirsch,<br />

2005). Taken together, data suggest that deficits in oxytocin may<br />

influence mistrust and hostility which can contribute to aggression and violence<br />

(Siever, 2008).<br />

E. Autonomic response measures<br />

Autonomic arousal is most commonly measured via HR and electrodermal<br />

activity (EDA). In general, research regarding autonomic arousal and aggressive<br />

and violent behavior has yielded findings suggesting a pattern of lower baseline<br />

levels of autonomic arousal and higher autonomic reactivity in children and<br />

adolescents (Patrick, 2008). Although less consistent, research on aggressive<br />

adults indicates an increase in autonomic activity in response to a stressor<br />

(Patrick, 2008). A met analysis by Lorber (2004) found a reliable but modest<br />

association between the autonomic measures of HR and EDA and aggression and<br />

conduct problems. Below is a brief review of the specific associations between<br />

HR and EDA and aggression and violence.<br />

1. Heart rate and electrodermal activity<br />

Low resting HR is a common phenomenon among aggressive children (Scarpa<br />

and Raine, 1997) and is associated with conduct problems throughout childhood,<br />

adolescence, and adulthood (Lorber, 2004). Evidence for HR reactivity is<br />

less consistent. It appears that aggressive children have increased HR in response


226 LaPrairie et al.<br />

to a stressor (Lorber, 2004), and this finding is particularly robust in children<br />

displaying reactive aggression rather than proactive aggression (Hubbard et al.,<br />

2002). HR reactivity and its association with aggression in adults are also mixed.<br />

EDA refers to small changes in electrical activity of the skin, usually<br />

occurring 1–3 s following the onset of a stimulus (Scarpa and Raine, 1997).<br />

Lorber (2004) found that conduct problems in childhood were associated with<br />

reduced EDA in the absence of stimulation, and reduced EDA during a task, but<br />

only in the presence of nonnegative stimuli. This met analysis also revealed a<br />

positive association between adult aggression and EDA reactivity. Scarpa and<br />

Raine (1997) suggested that EDA under arousal may be associated with specific<br />

forms of crime, such as crimes of evasion. Skin conductance levels have also been<br />

shown to interact with markers of the HPA axis to predict later externalizing<br />

behaviors in children (El-Sheikh et al., 2008). Results indicate that children with<br />

higher levels of EDA and higher levels of cortisol display increased levels of<br />

externalizing behaviors.<br />

F. Electro cortical response measures<br />

Electroencephalography (EEG) uses electrodes placed around the scalp at specified<br />

points to measure electrical activity of the brain. EEG evidence suggests that<br />

slow-wave activity during adolescence may predict later antisocial behavior<br />

(Raine et al., 1990). These findings have been interpreted to suggest that cortical<br />

immaturity leads to reduced inhibition (Volavka, 1999) and increased impulsive<br />

behavior. EEG abnormalities, specifically under arousal and cortical immaturity,<br />

have been reported in violent recidivistic offenders (Raine et al., 1990).<br />

Event-related potential (ERP) refers to the averaged changes in electrical<br />

brain activity in response to a stimulus. Literature on ERP and aggressive<br />

behavior is mixed. One consistent finding has been the association between<br />

reduced amplitude of the P300 wave response, an ERP elicited by infrequent<br />

stimuli, among aggressive and impulsive individuals (Gerstle et al., 1998). The<br />

P300 is thought to reflect online updating of cognitive representations (Donchin<br />

and Coles, 1988), thus the reduced amplitude may suggest impairment in higher<br />

cognitive functioning (e.g., working memory) in these individuals (Patrick,<br />

2008). Reduced P300 has been reported in antisocial personality disorder<br />

(Bauer et al., 1994), as well as other disorders that involve impaired impulsivity<br />

such as child CD and ADHD, drug dependence, and nicotine dependence<br />

(Iacono et al., 2002). Therefore, reduced P300 amplitude may be a reflection of<br />

impulse control, a characteristic of reactive aggression (Patrick, 2008).<br />

The physiological findings presented above illustrate the apparent<br />

biological underpinnings involved in the development of aggression and violence.<br />

Research from physiological and neurobiological studies has provided<br />

significant evidence for the biological basis of violent and aggressive behavior.


9. Perinatal Factors in the Development of Aggression 227<br />

Taken together, the neurobiological and psychophysiological findings suggest a<br />

model in which aggression arises due to dysfunction in brain areas responsible for<br />

emotion processing, inhibition, and reactivity (Davidson, 2000). Repeated aggression<br />

may contribute to deficits in recognizing and processing of emotion, as<br />

well as the regulation and modulation of aggressive behavior in response to<br />

threat. Reduced activation and distribution of neurotransmitters in areas of the<br />

brain implicated in the development of violent behavior, in combination with<br />

dysregulation of the body’s hormonal responsiveness, may add to difficulties in<br />

controlling aggressive outbursts. Furthermore, reduced arousal to distressing<br />

stimuli, as evidenced by a diminished physiological response and electrical<br />

activity in the brain may contribute to further deficiencies in appropriate<br />

responses to increased arousal. Further research is necessary to provide additional<br />

evidence for this suggested model.<br />

As the field progresses, particular attention should be paid to the<br />

subtypes of aggressive behavior that may be characterized by unique physiological<br />

patterns. Future studies should focus on parsing the different types of aggression<br />

to elucidate the specific pathways underlying violent behaviors. A better<br />

understanding of these pathways allows for earlier intervention that may be able<br />

to reduce or prevent severe aggressive behavior later in life. With the knowledge<br />

of the psychophysiology associated with the development of violence throughout<br />

the lifetime, we now turn specifically to the perinatal period to better understand<br />

how these biological pathways can be altered to prevent or lead to later violence.<br />

III. PERINATAL FACTORS RELATED TO THE DEVELOPMENT<br />

OF AGGRESSION<br />

The notion of a mother’s health affecting that of her unborn infant is one that<br />

has pervaded common knowledge for centuries. Only recently, however, have<br />

scientists begun to empirically study the specific factors influencing fetal development.<br />

Preterm birth, delivery complications, maternal mental illness, gender,<br />

and exposure to drugs, alcohol, and tobacco are all topics that have been<br />

explored in relation to their impact on fetal development. These factors have<br />

been shown to have maladaptive effects on fetal brain development following<br />

prenatal exposure—a finding that makes pregnancy a critical window for the<br />

prevention of unfavorable outcomes throughout the lifespan of the offspring.<br />

One such risk that has been identified in relation to these perinatal factors is the<br />

development of aggression. It has been suggested that through disruptions in<br />

neural development, the fetus incurs neuropsychological deficits—namely neural<br />

impairments in executive and verbal functioning that may result in an irritable<br />

disposition, poor behavioral regulation, or aggression (Brennan et al., 2003). This<br />

tendency toward aggression tends to persist throughout adolescence and into


228 LaPrairie et al.<br />

adulthood, manifesting itself through externalizing behaviors, internalizing problems<br />

(depression, loneliness, etc.), poor peer relationships, psychological disorders<br />

(CD, oppositional defiant disorder, etc.), recurring criminal behaviors,<br />

and violence. Because of the extensiveness of the risks associated with aggression,<br />

it has many potentially negative outcomes both in the developing infant, as<br />

well as in the significant economic and social burdens it places on society. In this<br />

section, the potential influences of the aforementioned perinatal factors will be<br />

explored in their relation to the development of aggression and violence. It is<br />

important to note that these risks do not necessitate the development of aggression<br />

in offspring but rather are important in understanding certain conditions in<br />

which aggressive traits may be more likely to arise.<br />

A. Birth complications<br />

Many studies have found associations between birth complications and negative<br />

behavioral outcomes in offspring. Specifically, irritable temperament in childhood,<br />

violent offending in adolescence and adulthood, and aggressive behaviors<br />

throughout the lifespan are among the offspring outcomes that have been<br />

associated with higher rates of birth complications. Birth complications typically<br />

refer to the following three factors: (1) prenatal complications, such as hypertension,<br />

mental illness, stress, drug and alcohol exposure, and viral infections<br />

experienced by the mother; (2) perinatal complications, which include difficult<br />

fetal delivery (e.g., breech birth), premature breaking of the membrane, assisted<br />

delivery (forceps and cesarean), fetal distress (i.e., difficulty breathing), preeclampsia,<br />

and umbilical cord prolapsed; and (3) postnatal complications as<br />

indicated by either cyanosis or treatment with oxygen (Liu et al., 2009).<br />

The mechanisms through which birth complications may influence the<br />

development of aggression are unknown, but they are hypothesized to involve<br />

damage to the PFC, hippocampus, and dopamine systems (Brennan et al., 1997;<br />

Cannon et al., 2002; Mednick and Kandel, 1988; Raine, 2002b). More specifically,<br />

preeclampsia, maternal bleeding, and maternal infection may cause an<br />

inadequate supply of blood to the placenta, fetal hypoxia or anoxia (lack of<br />

oxygen), and disrupted development of the hippocampus, dopamine systems, and<br />

other parts of the brain (Cannon et al., 2002; Liu, 2004; Mednick and Kandel,<br />

1988). Animal research has supported these findings by suggesting that perinatal<br />

complications surrounding anoxia in rats may reduce central dopamine transmission<br />

(Brake et al., 2000). Dopaminergic neurotransmission appears to be<br />

involved in impulse control, aggression, and violence (Chen et al., 2005; Retz<br />

et al., 2003). Animal research also suggests that perinatal complications may<br />

limit neurotransmitter functioning in the left PFC—an effect that has been one<br />

of the most frequently replicated indicators of violent offending in the brain<br />

imaging literature (Henry and Moffitt, 1997). It is important to note, however,


9. Perinatal Factors in the Development of Aggression 229<br />

that birth complications alone are unlikely to predispose infants to externalizing<br />

behavior and aggression. Instead, these complications interact with various<br />

psychosocial risk factors (i.e., poverty, poor parenting, parental rejection, negative<br />

peer relationships, bad neighborhoods, etc.), and likely genetic factors, to<br />

influence aggressive tendencies (Raine et al., 1994).<br />

Prenatal, perinatal, and postnatal health care interventions aimed at<br />

reducing birth complications may help to decrease risks of later development of<br />

aggression and violence. If women who may be at risk for birth complications are<br />

identified and educated, these mothers may be in a better position to take steps<br />

toward keeping their pregnancies and their babies healthy. If, however, birth<br />

complications do occur, early enrichment programs, that improve cognitive<br />

ability or enhance the parent–child relationship, may be effective in preventing<br />

the emergence of aggressive and violent behavior in adolescence and adulthood.<br />

B. Preterm birth and low birth weight<br />

Research supporting the role of neuropsychological deficits in mediating birth<br />

complications and adverse outcomes is consistent with preterm and low birth<br />

weight literature. White et al. (1994) have shown that medical and congenital<br />

risk factors, such as low birth weight and preterm birth, may lead to neuropsychological<br />

deficits and CNS damage that result in an increased likelihood for<br />

criminal offending. Evidence also suggests that preterm birth may be involved in<br />

the development of externalizing behaviors and aggression, and that these<br />

negative behavioral outcomes worsen with age (Bhutta et al., 2002).<br />

Low birth weight and preterm birth serve as strong and consistent<br />

predictors of neuropsychological deficits that may result in subsequent aggression<br />

and antisocial behavior (McCormick, 1985). Low birth weight infants were three<br />

times more likely to experience neurological deficits than controls (McCormick,<br />

1985). Moreover, such CNS deficits (Moffitt, 1993) may manifest themselves in<br />

a variety of ways, including temperamental difficulties, cognitive deficits, inattention,<br />

antisocial behavior, subnormal growth, learning difficulties, hyperactivity,<br />

behavioral problems, poor academic achievement, CNS damage, and<br />

psychiatric disorders (Piquero and Tibbetts, 1999).<br />

Preterm birth and low birth weight have been found to be correlated<br />

with maternal tobacco use, lack of prenatal care, drug and alcohol use by the<br />

mother during pregnancy, low socioeconomic status, poor diet, psychotropic drug<br />

use during pregnancy, and low parental educational level (Piquero and Tibbetts,<br />

1999). Because of the range of factors that might cause preterm birth and low<br />

birth weight in an infant, preventative efforts are critical.<br />

It is important to note that the effects of preterm birth and low birth<br />

weight on psychological functioning and aggression may vary depending on the<br />

sex of the child. For example, low birth weight boys exhibit a significantly more


230 LaPrairie et al.<br />

aggressive and delinquent acts in comparison to their female counterparts (Ross<br />

et al., 1990). There is also considerable evidence suggesting that disadvantaged<br />

home environments and maternal interactive style may moderate the relationship<br />

between these risks and aggressive behaviors (Piquero and Tibbetts, 1999).<br />

Therefore, a number of factors, such as sex, home environment, and maternal<br />

interactive style, are involved in the phenotypic expression of aggression in<br />

premature and low birth weight infants.<br />

Despite the decrease in infant mortality over the past 30 years, the<br />

prevalence of preterm birth and low birth weight has actually increased to<br />

approximately 12.5% of births in the United <strong>State</strong>s. Given the prevalence of<br />

preterm birth and low birth weight in the United <strong>State</strong>s, it is understandable why<br />

risk factors associated with this population are such a major public health<br />

concern (Berman and Butler, 2006). Preventative measures and public awareness<br />

campaigns focusing on the risks involved in preterm birth and low birth weight<br />

are a necessary next step in addressing these issues.<br />

C. Prenatal drug and alcohol exposure<br />

1. Alcohol<br />

Sixteen percent of children born in the United <strong>State</strong>s are exposed prenatally to<br />

alcohol, making alcohol the most common neurobehavioral teratogen affecting<br />

fetal development (Sood et al., 2001). Overall, children who are prenatally<br />

exposed to alcohol are 3.2 times more likely to develop aggression and delinquent<br />

behavior than nonexposed children. Further, children exposed to low<br />

levels of alcohol prenatally show higher scores for aggressive and externalizing<br />

behaviors on the Child Behavior Checklist (CBCL) and children exposed to<br />

moderate levels have higher scores on delinquent and total problem behavior on<br />

the CBCL (Sood et al., 2001). This suggests a higher threshold for the development<br />

of delinquency in children, as opposed to aggressive and externalizing<br />

behaviors. However, it also negates claims that low levels of alcohol consumption<br />

during pregnancy are tolerable with evidence that suggests that even a small<br />

dose may have adverse effects on fetal development. More generally speaking,<br />

the literature supports a dose–response continuum where a more heavily exposed<br />

fetus shows a greater magnitude of these adverse effects (Driscoll et al., 1990).<br />

Fetal alcohol spectrum disorder (FASD) and fetal alcohol syndrome<br />

(FAS) are conditions that may arise when children are prenatally exposed to<br />

alcohol. These disorders are characterized by physical and mental birth defects<br />

that may result in impaired interpersonal skills and social deficits. Some of the<br />

behaviors that are commonly observed among populations of FAS individuals<br />

are tendencies to demand attention, interrupt others, lie, show impaired moral<br />

judgment (especially with regard to social relationships), overreact to situations


9. Perinatal Factors in the Development of Aggression 231<br />

with excessively strong emotional responses, monopolize conversations, and<br />

demonstrate unawareness of the consequences of one’s actions. This population<br />

may also suffer major language and social communication deficits, which further<br />

hamper their social competence (Kelly et al., 2009).<br />

Animal studies have helped in linking prenatal alcohol exposure to the<br />

development of aggressive behaviors. Specifically, evidence suggests that alcohol<br />

exposure during prenatal development causes CNS damage. Rats provide an excellent<br />

model for understanding the development of aggression, because their social<br />

behavior has been shown to follow similar patterns to that of humans. Their social<br />

behavior results from a combination of influences including genetic makeup, teratogenic<br />

influences, early maternal–infant interactions, and later social learning.<br />

Primates, too, offer a suitable model for studying the effects of prenatal alcohol<br />

exposure, as their gestation characteristics and early developmental stages are<br />

similar to that of humans. In both animals and humans, prenatal alcohol exposure<br />

is not considered a singular cause of social deficits, but rather a probabilistic<br />

contributor serving as a risk factor for the developing child (Kelly et al.,2000).<br />

Because there are social–familial influences associated with prenatal<br />

exposure to alcohol, one might ask how it can be determined that alcohol<br />

exposure has any actual teratological effect. Animal models provide a means<br />

through which alcohol- and environment-related factors can be separated in an<br />

experimental fashion. For example, removing a newborn pup from its “alcoholusing”<br />

mother and transferring it to a foster-parent environment results in rates<br />

of aggression that are analogous to those remaining in the care of their “alcoholusing”<br />

mothers, suggesting that changes in aggressive behavior are initiated by<br />

the pup’s prenatal exposure to alcohol, rather than by its environment. This type<br />

of experiment, for obvious reasons, would be ethically impossible to conduct in<br />

human populations (Kelly et al., 2000).<br />

Animal models have contributed essential components to our understanding<br />

of the specific mechanisms through which prenatal ethanol (alcohol)<br />

exposure and aggression are linked. Rodent studies suggest that the behavioral<br />

deficits that result from ethanol exposure in utero are linked to ethanol-induced<br />

changes in the CNS. These changes in the CNS, however, are not uniform, and<br />

some brain regions (i.e., neocortex, hippocampus, cerebellum) are more affected<br />

than others. Notably, the HPA axis and beta-endorphin (b-EP) systems become<br />

dysregulated and hyperresponsive to social situations, which is demonstrated by<br />

heightened and prolonged concentrations of hormones, such as corticosterone<br />

(CORT) and adrenocorticotropin (ACTH), as well as elevated plasma levels and<br />

reduced pituitary content of b-EP compared to controls. Further, ethanol-exposed<br />

neonates show heightened sensitivity to stressors, significantly increased corticotrophin<br />

release factor (CRF) biosynthesis and expression, and more prolonged<br />

CORT and ACTH elevations during and after stress. These effects, which persist<br />

throughout the neonate’s lifespan, indicate deficits in pituitary–adrenal response


232 LaPrairie et al.<br />

inhibition and in recovery from stress. It is through these mechanisms that<br />

prenatal alcohol exposure may manifest in aggressive tendencies and externalizing<br />

behaviors in the lives of effected offspring (Weinberg et al., 1996).<br />

2. Drugs<br />

Drug use by pregnant women has increased steadily. Despite general awareness of<br />

detrimental effects, drug use during pregnancy continues its upward trend with<br />

prevalence estimates ranging from 0.3% to 46%. Prenatal drug exposure is associated<br />

with behavioral abnormalities, such as excessive irritability, poor socialattachment<br />

behavior, and aggression (Johns et al., 1994). The neurobiological<br />

processes through which these deficits emerge primarily involve the effects of<br />

drugs on fetal organogenesis, especially fetal brain development (Mayes, 1994).<br />

Evidence suggests that an increase in aggressive or violent behaviors associated with<br />

prenatal drug use may arise from alterations in the CNS. More specifically, aggressive<br />

behaviors can be linked to changes in fetal neurotransmitter systems, particularly<br />

within the limbic system (Miller et al., 1991). Cocaine (including crack<br />

cocaine) is one of the most commonly studied CNS stimulants in the literature on<br />

prenatal drug exposure. Cocaine affects monoaminergic neurotransmitter (dopamine,<br />

norepinephrine, and 5-HT) systems in the CNS by blocking the reuptake of<br />

dopamine, norepinephrine, and 5-HT leaving more of these neurotransmitters<br />

within the synaptic space (and therefore the peripheral blood). An excess in the<br />

amount of these neurotransmitters results in psychological effects, such as pleasure<br />

and euphoria, as well as specific behaviors and physiological reactions. Physiologically,<br />

chronic cocaine use may lead to tolerance whereby increasing amounts of the<br />

drug are necessary to achieve a desired effect.<br />

In the developing fetus, monoaminergic neurotransmitters play a critical<br />

role in fetal brain development by influencing cell proliferation, neural<br />

outgrowth, and synaptogenesis (Lauder, 1988; Mattson, 1988). Cocaine and<br />

other drugs may affect these neural processes throughout gestation through<br />

their effects on the release and metabolism of monoamines. The importance of<br />

monoamine neurons in fetal brain development has been demonstrated in both<br />

human and animal models. For example, in the rats’ second week of gestation,<br />

norepinephrine neurons appear in the locus coeruleus, 5-HT neurons are found<br />

in the raphe nuclei, and dopaminergic neurons in the substantia nigra are<br />

functional (Lauder and Bloom, 1974). By the end of the second month of<br />

human gestation, 5-HT and norepinephrine neurons can be found. In both<br />

animals and humans, these monoamine neurons are rapidly generating axonal<br />

connections with the forebrain and actively influencing the production and<br />

differentiation of cell structure in these regions (Lidov and Molliver, 1982a,b;<br />

Wallace and Lauder, 1983).


9. Perinatal Factors in the Development of Aggression 233<br />

Further evidence of these effects can be demonstrated by administering<br />

cocaine to rats during the early postnatal period when synaptogenesis begins in<br />

the forebrain. This early postnatal period in rats is functionally equivalent to the<br />

third trimester in human gestation during which axonal and dendritic growth<br />

take place. When brain glucose metabolism is used as an indicator of activity,<br />

animal models exhibit the greatest percentage change in brain regions with high<br />

dopaminergic activity in comparison to untreated controls (Dow-Edwards et al.,<br />

1988, 1989). Several brain structures associated with the mesocortical and<br />

mesolimbic systems, including the cingulate cortex, PFC, nucleus accumbens,<br />

amygdala, septum, ventral tegmental area, and ventral thalamic nucleus, appear<br />

highly affected by dopamine activity (Goeders and Smith, 1983; Shepard, 1988).<br />

Each of these areas is thought to be involved in an organism’s arousal, attention,<br />

and ability to regulate anxiety and emotional responses (Mayes, 1994).<br />

The neural processes mentioned above may lead to the development of<br />

aggression when abnormalities in fetal brain development later manifest themselves<br />

in social and behavioral ways. Basic processes, like the regulation of<br />

attention, response to sensory stimuli, and the modulation of mood states may<br />

all be linked to prenatal drug exposure through the drug’s alterations of neurotransmitter<br />

activity. Several studies have found that infants exposed to drugs<br />

prenatally are often easily irritable and difficult to engage. Evidence also suggests<br />

that prenatal drug exposure results in crying patterns that indicate a general<br />

“excitable” tendency within affected infants. Human infants exposed prenatally<br />

to cocaine have also shown elevated HRs and norepinephrine levels at 2 months<br />

of age; lending further support to the influence cocaine has on monoaminergic<br />

systems (Mayes, 1994; Mirochnick et al., 1991). Rodent models have demonstrated<br />

an increased susceptibility to stressors, higher vulnerability to the environment,<br />

and increased rates of aggressive behaviors in response to social<br />

competition among offspring prenatally exposed to drugs (Spear et al., 1998).<br />

The influence of prenatal exposure to drugs and alcohol on social and<br />

aggressive behavior has serious implications for crime prevention. Early identification<br />

and intervention among infants and children who may be affected by<br />

prenatal drug or alcohol exposure is necessary to prevent delinquency and poor<br />

social relationships within these populations (Johns et al., 1994). Such steps are<br />

also necessary for gaining a better understanding of the behavioral differences<br />

that may exist within educational, occupational, and social settings due to<br />

prenatal exposure to drugs and alcohol.<br />

D. Smoking<br />

Smoking during pregnancy remains a critical public health concern. Nearly half of<br />

all women who smoke continue to do so even while pregnant, despite some women’s<br />

intentions to refrain from doing so. Despite common knowledge of the adverse


234 LaPrairie et al.<br />

effects of maternal smoking during pregnancy among the American public, more<br />

than half a million infants per year in the United <strong>State</strong>s are prenatally exposed to<br />

maternal smoking (Wakschlag et al., 2002). This is of even greater concern when<br />

one considers the failure of public health smoking cessation campaigns for the<br />

10.2% of women in the United <strong>State</strong>s who continue to smoke through their<br />

pregnancies. Adverse outcomes, which include low birth weight, premature delivery,<br />

spontaneous abortion, and infant mortality, have been the primary focus of<br />

these campaigns (Weaver et al.,2007). In comparison, relatively little attention has<br />

been paid, from a public health standpoint, to the relationship between prenatal<br />

smoking and the development of aggression and violence in offspring.<br />

Prenatal smoking predicts children’s likelihood of displaying high aggression<br />

from as early as 1.5 years and throughout adulthood (Huijbregts et al.,<br />

2008). Several externalizing behaviors, including impulsivity, truancy, hyperactivity,<br />

attentional difficulties, and delinquency, have all been found to be<br />

associated with maternal prenatal smoking through the fetus’ exposure in<br />

uterine.<br />

Potential neurobiological mechanisms through which prenatal nicotine<br />

exposure may increase the offspring’s risk for aggressive behaviors include the<br />

HPA axis and the CNS (Brennan et al., 1997). Substantial evidence suggests<br />

that nicotine crosses the placental barrier and causes neurotoxicity in the fetus.<br />

Neurotoxicity occurs via hypoxic effects on the fetal-placental unit (e.g., reduction<br />

of fetal blood flow) and teratological effects on the developing fetal brain.<br />

Two recent human studies support this contention, noting associations between<br />

maternal prenatal smoking and decreased frontal lobe volumes in infants (Ekblad<br />

et al., 2010), and a thinning of the cerebral cortex in adolescents (Toro et al.,<br />

2008). Within the HPA axis, nicotine produces a heightened ACTH response to<br />

stress in adult rats (Poland et al., 1994). Other studies have found that elevated<br />

levels of ACTH increase aggressive and defensive behaviors in both rats and<br />

nonhuman primates, suggesting that this hormone may be related to the development<br />

of aggression (Higley et al., 1992; Veenema et al., 2007). However, lower<br />

levels of ACTH have also been found within human criminal and antisocial<br />

populations in comparison to controls, so these results are mixed and should be<br />

interpreted with caution (Coccaro and Siever, 2002; Virkkunen et al., 1994).<br />

Nicotinic acetylcholine receptors (nAChRs) are responsible for the<br />

regulation of many vital phases of brain maturation. These receptors are present<br />

in the brain early in gestation and develop throughout prenatal, postnatal, and<br />

adolescent periods, suggesting that nicotinic signaling plays a crucial role in neural<br />

development. During these developmental periods, NAChRs are particularly<br />

sensitive to environmental stimuli and, as specific nicotine-sensitive receptors,<br />

are especially vulnerable to exogenous nicotine. Nicotine affects fetal development<br />

primarily through its effect on nicotinic-binding sites in the cerebral cortex.<br />

More specifically, nicotine has been found to alter the neocortex, hippocampus,


9. Perinatal Factors in the Development of Aggression 235<br />

and cerebellum during the early postnatal period within rats (the equivalent of the<br />

third trimester in humans; Dwyer et al., 2009). Evidence suggests that prenatal<br />

nicotine-induced defects within these particular brain regions may increase the<br />

likelihood of dopamine-mediated disorders like attention deficit hyperactivity<br />

disorder and substance abuse. Patterns of continuous maternal smoking (i.e., the<br />

tendency to smoke in a way that maintains plasma nicotine levels at a steady state)<br />

cause more negative effects than more periodic patterns of use, which allow the<br />

CNS to recover between episodes. The stimulation to nicotinic receptors interacts<br />

with the genes that influence differentiation of cells, causing permanent changes<br />

in cell functioning. It has been suggested that these processes disrupt the maturation<br />

of the fetal brain and produce adverse effects in fetal development that can<br />

later manifest themselves in aggression or violence (Wakschlag et al., 2002).<br />

Animal models demonstrate many of the biological effects of prenatal<br />

smoking on neonatal behavior. Rats exposed to nicotine prenatally show deficits<br />

in learning and memory, as well as in social behavior. Benowitz (1998) found<br />

that nicotine infusion in rats causes interference with neural cell replication and<br />

abnormal synaptic activity. These, in turn, produce neuroendocrine and behavioral<br />

abnormalities that could potentially lead to aggression. Rodent models have<br />

also shown similar adverse effects linked to second hand smoke, as well as<br />

maternal use of nicotine replacement therapy (NRT), a pharmacotherapy of<br />

smoking cessation thought to be less detrimental than smoking cigarettes during<br />

pregnancy (Dwyer et al., 2009). Findings of adverse effects related to maternal<br />

use of NRT are particularly disturbing, since (1) NRT does not seem to increase<br />

the likelihood of successful smoking cessation during pregnancy and (2) NRT<br />

has actually been recommended by a number of public health authorities,<br />

including the Food and Drug Administration (Bruin et al., 2010). NRT (as<br />

well as cognitive-behavioral therapy (CBT)) has been shown to be effective<br />

among nonpregnant smokers, so prevention rather than smoking cessation<br />

during pregnancy should be the aim for reducing adverse outcomes attributed<br />

to prenatal nicotine exposure. Further, interventionists should keep in mind that<br />

any prenatal nicotine exposure, even through modes of transmission not related<br />

to smoking, can be detrimental to fetal development.<br />

As with most toxins, the effects of prenatal smoking exposure are dosedependent<br />

and thus strongest among offspring of heavy smoking mothers (10<br />

cigarettes/day). Further, the effects of prenatal smoking are exacerbated when<br />

accompanied by low socioeconomic status, poor parenting, family dysfunction,<br />

paternal absence, and parental history of antisocial behavior. However, the<br />

relationship still exists even when these variables are controlled for (Huijbregts<br />

et al., 2008). Evidence suggests that gender might moderate the relationship<br />

between maternal prenatal smoking and externalizing behaviors in that the<br />

relationship is stronger among male offspring in predicting CD and stronger<br />

among female offspring when predicting substance abuse (Brennan et al., 2002).


236 LaPrairie et al.<br />

E. Maternal psychological stress<br />

Prenatal stress is so common an occurrence that it seems unlikely that it could<br />

have any significant effects or unfavorable life-long outcomes on child development.<br />

However, it is, in fact, associated with low birth weight, preterm birth,<br />

preeclampsia, spontaneous abortion, growth-retardation (specifically reduced<br />

head circumference), developmental delays, heightened emotionality, externalizing<br />

behaviors, irritability, psychopathology, and deficits in attention, cognition,<br />

and neurodevelopment (Clarke et al., 1994, 1996; Gutteling et al., 2005;<br />

Mulder et al., 2002). Effects involving birth outcomes are relevant to the<br />

development of aggression in the ways previously described. However, prenatal<br />

stress, more broadly speaking, also affects fetal neurodevelopment in a different<br />

way. Prenatal stress may stem from a variety of sources including, but not limited<br />

to inadequate social support, low socioeconomic status, unwanted pregnancy,<br />

and sexual, physical, or verbal abuse. These stressors may take the form of one<br />

traumatic event, several recurrent ones, or more chronically on a daily basis<br />

(Mulder et al., 2002). When the stressor is experienced, the HPA axis and the<br />

sympathetic nervous system are activated, as the body’s response to a particular<br />

threat the individual perceives in her environment. This physiological response<br />

has evolutionary value in that it places us in “fight or flight” mode, increasing our<br />

awareness of problems that may exist and preparing us to find ways of solving<br />

them. This process becomes maladaptive, however, when our perception of a<br />

threat or stressor is inconsistent with its actual magnitude and relevance to our<br />

lives, and alternatively, when the physiological response following a stressor is<br />

prolonged (Clarke et al., 1994, 1996; Gutteling et al., 2005; Mulder et al., 2002).<br />

The connection between prenatal stress and HPA axis activity has been<br />

demonstrated in rodent, animal, and human studies, and in all of these, both<br />

prenatal stress and heightened HPA axis response have been found to be<br />

predictive of the development of aggression in offspring (Clarke et al., 1994,<br />

1996; Gutteling et al., 2005; Mulder et al., 2002). A number of rodent studies<br />

have introduced stress to a pregnant mother prenatally using electrical shock,<br />

immobilization, or randomly administered bursts of noise. These studies have<br />

demonstrated a dysregulation of the HPA axis in both mother and pup that<br />

eventually leads to heightened emotionality, hostility, and aggression in the<br />

offspring (Clarke and Schneider, 1993; Mulder et al., 2002; Sobrian et al.,<br />

1997; Takahashi et al., 1990; Ward and Weisz, 2011). Similarly, studies of the<br />

offspring of rhesus monkey mothers exposed to stress from mid- to late-gestation<br />

demonstrated low birth weight, impaired neuromotor development, attention<br />

deficits, and disturbed behavior (Clarke et al., 1994, 1996; Schneider, 1992a,b).<br />

These effects were long-term and persisted even into the adolescent period of<br />

development (Clarke et al., 1996). In humans, similar effects of maternal prenatal<br />

stress have been reported (Gutteling et al., 2005; Mulder et al., 2002).


9. Perinatal Factors in the Development of Aggression 237<br />

HPA axis regulation involves several hormones, including corticoreleasing<br />

hormone (CRH), cortisol, (nor)adrenaline, and ACTH, which are<br />

released into the bloodstream when a stressor is experienced. Small increases<br />

in these hormones within the pregnant mother may lead to disproportionately<br />

large increases in fetal hormonal levels. Excessive levels of these hormones may<br />

potentially inhibit the growth and development of the nervous system, cause<br />

damage to the brain, and produce programming effects on the fetal neuroendocrine<br />

system that lead to the developmental deficits mentioned above. This may<br />

be especially true when HPA axis activity is characterized by an exceptionally<br />

strong, sustained response to the stressor. Animal models have supported this<br />

explanation by demonstrating experimentally that levels of these hormones are<br />

higher in neonates prenatally exposed to stress in comparison to controls (Clarke<br />

et al., 1994, 1996; Gutteling et al., 2005; Mulder et al., 2002).<br />

It is important to note that there have been gender differences in the<br />

findings within this area, namely that hostility and aggressive behaviors appeared<br />

to be more prevalent in male offspring than in females. These findings, which are<br />

typically found in rodent studies, suggest that males may be more vulnerable to<br />

the effects of prenatal stress than their female counterparts (Clarke et al., 1996).<br />

However, in general, it seems that prenatal stress may be an important predisposing<br />

factor for a number of behavioral deficits among both male and female<br />

offspring, even if to different degrees.<br />

Because stress can be so pervasive in the life of a pregnant mother, it<br />

often manifests itself in a variety of ways. For example, stress might lead a mother<br />

to engage in smoking or alcohol and substance abuse, which, in turn, can<br />

produce fetal neurobehavioral deficits of the kind that have been described in<br />

previous sections. To prevent these types of detriments from occurring, it may be<br />

necessary to assess women’s stress levels in early pregnancy, identify those who<br />

are at risk, and provide stress reduction programs throughout their pregnancies.<br />

Educating women about the risks involved with prenatal stress, as well as training<br />

them in relaxation methods, may be helpful in alleviating these effects. Ensuring<br />

that women have the appropriate buffers needed to prevent stress is also essential.<br />

Adequate social support and financial resources are just a few factors that<br />

may be necessary to ensure a mother’s psychological well-being.<br />

F. Environmental con<strong>text</strong><br />

It is important to note that the prenatal factors discussed above may not<br />

necessarily operate in a unidirectional manner. It may often be the case that<br />

these factors result in child aggression in the form of coercion or manipulation of<br />

the parent in order to obtain something that is wanted. This coercion is likely to<br />

elicit a negative response from the parent in the form of either negative reinforcement<br />

(giving in to the child) or positive reinforcement (giving increased


238 LaPrairie et al.<br />

attention to the child by chastising or yelling). This reinforcement indirectly<br />

encourages and exacerbates the child’s behavior, producing an ongoing cycle of<br />

reinforced aggressive behaviors throughout the child’s lifetime. Further, this<br />

cycle may be maintained by preexisting neuropsychological deficits and unintentionally<br />

harmful reinforcements from other figures in the child’s life, such as<br />

teachers, grandparents, and peers.<br />

Throughout this section, it has been continually noted that neither<br />

social nor biological elements operate alone in contributing to the development<br />

of aggression in offspring. One or the other may predispose a child to developing<br />

aggressive behaviors, but it is the “double hazard” of perinatal risk and social<br />

disadvantage that places a child at maximal risk for later aggression and externalizing<br />

behaviors (Brennan and Mednick, 1997). Understanding the interaction<br />

of biological and social risk factors in generating aggression is critical for<br />

preventing both personal costs (few positive social relationships, poor job performance,<br />

etc.), as well as social costs (crime rate, prison costs, etc.). Thus,<br />

researchers and policy makers should make it a goal to identify populations<br />

potentially affected by perinatal risk factors in order to more accurately predict<br />

who might benefit from interventions aimed at preventing later aggression,<br />

violence, and criminal offending.<br />

IV. GENETIC CONTRIBUTIONS<br />

As described in detail elsewhere in this volume, aggressive and violent behavior<br />

can in part be accounted for by genetic factors. Because prenatal stress and<br />

teratogenic exposures may be linked to genetic risk, it is important to consider<br />

potential genetic contributions to the association between perinatal factors and<br />

aggression.<br />

A. Genetic factors as explanatory<br />

One prenatal risk factor, that has received recent attention in terms of the<br />

potentially confound role of genetic factors, is maternal smoking during pregnancy.<br />

For example, twin studies have been utilized to assess whether the<br />

relationship between maternal prenatal smoking and offspring externalizing<br />

behavior remains significant when controlling for genetic influences. In one<br />

such study, the association between maternal smoking and offspring ADHD<br />

was found to persist after controlling for genetic influences (Thapar et al.,<br />

2003). In a separate twin study, researchers found that genetic effects explained<br />

about half of the association between maternal prenatal smoking and child


9. Perinatal Factors in the Development of Aggression 239<br />

conduct problems; in this study, controls for both genetic influences and parent<br />

psychopathology accounted for the initial association in its entirety (Maughan<br />

et al., 2004).<br />

Recent advances in infertility treatment (i.e., in vitro fertilization using<br />

donor eggs) have also allowed for the use of a prenatal cross fostering design to<br />

assess the moderating impact of genetic influences on the maternal prenatal<br />

smoking/child externalizing disorder association (Rice et al., 2009). In this novel<br />

study, maternal smoking during pregnancy was only associated with child antisocial<br />

outcomes in cases where the mother was implanted with her own egg, as<br />

opposed to an unrelated donor’s egg. These results suggest that genetic factors are<br />

a necessary component in the noted relationship between maternal smoking and<br />

child antisocial outcomes.<br />

Another novel design strategy has recently been used to evaluate outcomes<br />

for siblings discordant for maternal prenatal smoking (D’Onofrio et al.,<br />

2010; Lindblad and Hjern, 2010). Results from studies using this design suggest<br />

that familial background factors, rather than environmental exposure effects,<br />

explain associations between maternal prenatal smoking and externalizing problems.<br />

However, as acknowledged by their authors, these sibling discordant<br />

design studies did not test for gene by environment interactions, leaving open<br />

the possibility that prenatal exposure to maternal smoking may result in externalizing<br />

behavior outcomes for offspring at particular genetic risk.<br />

B. Gene by environment (G E) interactions<br />

Gene by environment interactions have recently been examined in terms of<br />

their relevance to perinatal risks and child behavioral outcomes. These studies<br />

have primarily focused on polymorphisms linked to the neurotransmitter systems<br />

of dopamine, norepinephrine, and 5-HT, which have been described previously<br />

in terms of their relevance to both perinatal factors and aggressive outcomes.<br />

1. Monoamine oxidase genotype<br />

Monoamine oxidase (MAO) is a critical enzyme involved in the degradation of<br />

neurotransmitters, including norepinephrine, dopamine, and 5-HT. MAO exists<br />

in two forms, MAOa and MAOb. The gene that codes MAOa has functional<br />

variations that influence the level of MAOa (Sabol et al., 1998), which in turn<br />

affects central levels of dopamine and 5-HT and thus, directly regulates behavior.<br />

Therefore, this gene has been the focus of molecular genetics studies of<br />

aggressive behavior in both humans (Manuck et al., 2000) and rodents (Cases<br />

et al., 1995) and is the most well-established susceptibility variant for aggression<br />

in several species. MAOa genotype appears to influence the development of


240 LaPrairie et al.<br />

violent behaviors by altering vulnerability to the effects of early adverse environments<br />

(Caspi et al., 2002; Kim-Cohen et al., 2006). Specifically, there is robust<br />

evidence that the interaction between MAOa and childhood maltreatment<br />

predicts child CD and adult antisocial behavior such that males with low<br />

expression of MAOa (L allele), but not males with high expression of MAOa<br />

(H allele) are at increased risk (Kim-Cohen et al., 2006; Nilsson et al., 2007).<br />

Recent evidence also links MAOa-L and low brain MAOa with trait aggression<br />

and neural hypersensitivity to social cues (Alia-Klein et al., 2008; Eisenberger<br />

et al., 2007). Interestingly, MAOa is an X-linked gene (with males carrying only<br />

one allele and females carrying two), which suggests the possibility of sex<br />

differences in genetic and epigenetic regulation and may explain the increased<br />

average aggressiveness in males in comparison to females.<br />

One recent study has noted an interaction between the MAOa uVNTR<br />

(untranslated region variable number of tandem repeats) genotype, gender, and<br />

maternal prenatal smoking in the prediction of CD symptoms (Wakschlag et al.,<br />

2010). Specifically, boys with the low activity MAOa genotype whose mothers<br />

smoked during pregnancy were at an increased risk of CD symptoms, whereas<br />

girls with the high activity MAOa genotype whose mothers smoked during<br />

pregnancy were at increased risk for hostile attribution bias (a characteristic<br />

common to aggressive children) as well as CD symptoms.<br />

2. Genes related to dopaminergic function<br />

Kahn et al. (2003) noted an interaction between a DAT1 genotype and maternal<br />

prenatal smoking in the prediction of oppositional and hyperactive symptoms in<br />

young children. This finding was replicated in an adolescent sample; however,<br />

the GE effect was specific to hyperactive-impulsive symptoms in males<br />

(Becker et al., 2008). Other studies have failed to replicate this effect for<br />

DAT1 and other dopamine-related genotypes (e.g., Brookes et al., 2006;<br />

Langley et al., 2008); however, relatively few studies have been completed in<br />

this area.<br />

3. Catechol O-methyltransferase<br />

Catechol O-methyltransferase (COMT) is a key modulator of extracellular dopamine<br />

levels in the PFC. A common G/A polymorphism produces a valine-tomethionine<br />

amino acid substitution at codons 108 and 158 (Val108/158Met;<br />

rs4680), which results in a three- to fourfold variation in COMT activity,<br />

whereby the Val and Met alleles confer high and low activity, respectively<br />

(Lachman et al., 1996). This well-characterized, functional polymorphism has<br />

been associated with atypical neural processing and connectivity in healthy


9. Perinatal Factors in the Development of Aggression 241<br />

individuals (Dennis et al., 2010), deficits in executive functioning abilities<br />

(Tunbridge et al., 2006), and with aggression and serious antisocial behavior in<br />

individuals with ADHD (Caspi et al., 2008).<br />

Prenatal exposure to nicotine also leads to persistent abnormalities in<br />

neurotransmitter functioning in the cerebrocortical areas of the rat brain<br />

(Slotkin et al., 2007). Further, both maternal prenatal smoking and COMT<br />

associations with CD appear to be specific to aggressive behavior, rather than<br />

covert antisocial behavior (Monuteaux et al., 2006, 2009). Taken together, these<br />

findings suggest that the combination of the Val/Val genotype and prenatal<br />

exposure to maternal smoking may lead to neural processing deficits that increase<br />

vulnerability for aggression.<br />

One study examining the interaction of prenatal risk and COMT<br />

variation in the prediction of antisocial outcomes found that birth weight<br />

interacted with Val108/158Met to predict antisocial behavior in an ADHD<br />

sample (Thapar et al., 2005); however, this finding has had at least one failure<br />

to replicate (Sengupta et al., 2006). Another recent study (Brennan et al., 2011)<br />

found that individuals with the COMT Val/Val genotype whose mothers also<br />

smoked during pregnancy were at an increased risk for aggressive behavior outcomes<br />

in adolescence and young adulthood. These GE interaction findings are<br />

preliminary but do suggest a potentially important role for genetics in noted<br />

relationships between perinatal risk factors and aggression.<br />

C. The role of epigenetics<br />

The analysis of a GE interaction is still focused on the “fixed” nature of the<br />

genome—the DNA sequence itself. The DNA sequence is the same in every cell<br />

of the body and does not change across the lifespan. But there are other<br />

characteristics of genetic makeup that are not as fixed, and that have been<br />

found to change in response to environmental influences over time. These are<br />

known as epigenetic phenomena. Several epigenetic processes have been discovered,<br />

but the most commonly studied today is the phenomenon of methylation, a<br />

measurable chemical modification to DNA that can directly alter the expression<br />

of genes.<br />

Methylation patterns change not only in response to toxins and stress<br />

encountered in the environment but also in response to nutritional supplements<br />

and parenting sensitivity. In a series of seminal studies in this area, Meaney and<br />

colleagues discovered that the quality of the parenting (licking and grooming)<br />

that a mother rat provided to her pups during early postnatal development<br />

changed the methylation patterns in the hippocampus of her offspring<br />

(Kappeler and Meaney, 2010). Specifically, higher levels of licking and grooming<br />

made the genes (and the offspring) less responsive to stress in the environment.<br />

In contrast, low levels of licking and grooming resulted in offspring whose


242 LaPrairie et al.<br />

genetic profile enhanced the release of cortisol in response to stress. Importantly,<br />

either strategy might be considered “ideal” parenting, depending upon the<br />

environment in which the offspring will have to survive.<br />

Recent research suggests that prenatal factors may also influence DNA<br />

methylation patterns in offspring (Radtke et al., 2011). Specifically, maternal<br />

reports of abuse during pregnancy were found to be correlated with methylation<br />

of the glucocorticoid receptor (GR) gene in offspring ages 10–19. In contrast,<br />

maternal experiences of abuse prior to or after pregnancy were not associated<br />

with offspring DNA methylation patterns. Importantly, methylation of the GR<br />

gene directly impacts the functioning of the HPA axis, which (as noted previously)<br />

may, in turn, impact levels of aggression and antisocial behavior.<br />

In summary, preliminary evidence suggests that high versus low risk<br />

genotypes may moderate the effects of perinatal exposures by influencing an<br />

individual’s susceptibility or resistance to these environmental experiences. In<br />

addition, perinatal risk factors are associated with epigenetic changes that are<br />

evident in and may influence later development. Complex behaviors, like aggression,<br />

are likely based on interactions of numerous genes and numerous<br />

environmental factors. Future molecular genetics and epigenetic programming<br />

studies should attempt to unravel the interplay between genes and environment.<br />

Such knowledge will provide a clearer understanding of the role of early risk<br />

factors in the development of aggression and how they can be used as intervention<br />

targets to alter developmental trajectories that lead to a lifetime of violence.<br />

V. CONCLUSIONS<br />

It is highly evident based on experimental and clinical studies that deleterious<br />

perinatal exposures can have a profound and enduring impact on the neuroregulatory<br />

systems that mediate violence and aggression. Early adverse perinatal<br />

experiences, in combination with predisposing genetic factors, combine with<br />

unstable family environments to substantially increase the vulnerability for a<br />

trajectory of delinquent and aggressive behavior throughout the lifespan; however,<br />

these outcomes are both complex and multidimensional. Future studies<br />

should focus on genetic risk factors, as well as novel interventions that may<br />

mitigate or prevent the deleterious effects of an adverse perinatal environment<br />

on the development of aggression. Effective interventions should target prenatal<br />

maternal mental and physical health-related behaviors, address parenting behaviors<br />

during critical stages of child development (i.e., infancy, early childhood,<br />

and adolescence), as well as focus on child cognitive and social enrichment<br />

during pre- and elementary-school years. As we are just beginning to understand<br />

the complexity of the intergenerational transmission of these problems during


9. Perinatal Factors in the Development of Aggression 243<br />

pregnancy and early childhood, it is important as a field to focus on the origin,<br />

early development, and prevention of aggression and violence to prevent vulnerable<br />

families and at-risk children from a lifetime of adversity.<br />

References<br />

Alexander, M., and Perachio, A. A. (1973). The influence of target sex and dominance on evoked<br />

attack in Rhesus monkeys. Am. J. Phys. Anthropol. 38(2), 543–547.<br />

Alia-Klein, N., Goldstein, R. Z., Kriplani, A., Logan, J., Tomasi, D., Williams, B., Telang, F.,<br />

Shumay, E., Biegon, A., Craig, I. W., Henn, F., and Wang, G. (2008). Brain monoamine oxidase<br />

A activity predicts trait aggression. J. Neurosci. 28(19), 5099–5104.<br />

Alink, L. R., van Ijzendoorn, M. H., Bakermans-Kranenburg, M. J., Mesman, J., Juffer, F., and<br />

Koot, H. M. (2008). Cortisol and externalizing behavior in children and adolescents: Mixed<br />

meta-analytic evidence for the inverse relation of basal cortisol and cortisol reactivity with<br />

externalizing behavior. Dev. Psychobiol. 50(5), 427–450.<br />

Anderson, S. W., Bechara, A., Damasio, H., Tranel, D., and Damasio, A. R. (1999). Impairment of<br />

social and moral behavior related to early damage in human prefrontal cortex. Nat. Neurosci. 2<br />

(11), 1032–1037.<br />

Archer, J. (1991). The influence of testosterone on human aggression. Br. J. Psychol. 82(1), 1–28.<br />

Archer, J., Graham-Kevan, N., and Davies, M. (2005). Testosterone and aggression: A reanalysis of<br />

Book, Starzyk, and Quinsey’s (2001) study. Aggress. Violent Behav. 10(2), 241–261.<br />

Bain, J., Langevin, R., Dickey, R., and Ben-Aron, M. (1987). Sex hormones in murderers and<br />

assaulters. Behav. Sci. Law 5, 95–101.<br />

Barker, E. D., Séguin, J. R., White, H. R., Bates, M. E., Lacourse, E., Carbonneau, R., and<br />

Tremblay, R. E. (2007). Developmental trajectories of male physical violence and theft: Relations<br />

to neurocognitive performance. Arch. Gen. Psychiatry 64(5), 592–599.<br />

Bauer, L. O., O’Connor, S., and Hesselbrock, V. M. (1994). Frontal p300 decrements in antisocial<br />

personality disorder. Alcohol. Clin. Exp. Res. 18, 1300–1305.<br />

Beauchaine, T. P., Gartner, J., and Hagen, B. (2000). Comorbid depression and heart rate variability<br />

as predictors of aggressive and hyperactive symptom responsiveness during inpatient treatment of<br />

conduct-disordered, ADHD boys. Aggress. Behav. 26(6), 425–441.<br />

Becker, K., El-Faddagh, M., Schmidt, M. H., Esser, G., and Laucht, M. (2008). Interaction of<br />

dopamine transporter genotype with prenatal smoke exposure on ADHD symptoms. J. Pediatr.<br />

152, 263–269.<br />

Benowitz, N. L. (1998). Pharmacology of nicotine. In “Handbook of Substance Abuse: Neurobehavioral<br />

Pharmacology” (R. E. Tarter, R. T. Ammerman, and P. J. Ott, eds.), pp. 283–297. Plenum<br />

Press, New York, NY.<br />

Berkowitz, L. (1989). Frustration-aggression hypothesis: Examination and reformulation. Psychol.<br />

Bull. 106(1), 59–73.<br />

Berman, R. E., and Butler, A. S. (2006). Preterm Birth: Causes, Consequences, and Prevention.<br />

National Academics, Washington, DC.<br />

Bhutta, A. T., Cleves, M. A., Casey, P. H., Cradock, M. M., and Anand, K. (2002). Cognitive and<br />

behavioral outcomes of school-aged children who were born preterm: A meta-analysis. JAMA 288<br />

(6), 728–737.<br />

Blair, R. J. R. (2001). Neurocognitive models of aggression, the antisocial personality disorders, and<br />

psychopathy. J. Neurol. Neurosurg. Psychiatry 71(6), 727.<br />

Blair, R. J. R. (2010). Psychopathy, frustration, and reactive aggression: The role of ventromedial<br />

prefrontal cortex. Br. J. Psychol. 101(Pt 3), 383–399.


244 LaPrairie et al.<br />

Blair, R. J. R., Colledge, E., Murray, L., and Mitchell, D. G. (2001). A selective impairment in the<br />

processing of sad and fearful expressions in children with psychopathic tendencies. J. Abnorm.<br />

Child Psychol. 29(6), 491–498.<br />

Brake, W. G., Sullivan, R. M., and Gratton, A. (2000). Perinatal distress leads to lateralized medial<br />

prefrontal cortical dopamine hypofunction in adult rats. J. Neurosci. 20(14), 5538–5543.<br />

Brendgen, M., Dionne, G., Girard, A., Boivin, M., Vitaro, F., and Perusse, D. (2005). Examining<br />

genetic and environmental effects on social aggression: A study of 6-year-old twins. Child Dev. 76,<br />

930–946.<br />

Brennan, P. A., and Mednick, S. A. (1997). Medical histories of antisocial individuals. In “Handbook<br />

of Antisocial Behavior” (D. M. Stoff, J. Breiling, and J. D. Maser, eds.), pp. 269–279. Wiley,<br />

New York.<br />

Brennan, P. A., Mednick, S. A., and Raine, A. (1997). In “Biosocial Interactions and Violence:<br />

A Focus on Perinatal Factors” (A. Raine, ed.), pp. 163–174.<br />

Brennan, P. A., Grekin, E. R., Mortensen, E. L., and Mednick, S. A. (2002). Relationship of maternal<br />

smoking during pregnancy with criminal arrest and hospitalization for substance abuse in male and<br />

female adult offspring. Am. J. Psychiatry 159(1), 48–54.<br />

Brennan, P. A., Hall, J., Bor, W., Najman, J. M., and Williams, G. (2003). Integrating biological and<br />

social processes in relation to early-onset persistent aggression in boys and girls. Dev. Psychol. 39<br />

(2), 309–323.<br />

Brennan, P. A., Hammen, C., Sylvers, P., Bor, W., Najman, J., Lind, P., Montgomery, G., and<br />

Smith, A. K. (2011). Interactions between the COMT Val108/158Met polymorphism and<br />

maternal prenatal smoking predict aggressive behavior outcomes. Biol. Psychol. 87, 99–105.<br />

Broidy, L. M., Nagin, D. S., Tremblay, R. E., Brame, B., Dodge, K., Fergusson, D., Horwood, J.,<br />

Loeber, R., Laird, R., Lynam, D., Moffitt, T., Bates, J. E., et al. (2003). Developmental trajectories<br />

of childhood disruptive behaviors and adolescent delinquency: A six-site, cross-national study.<br />

Dev. Psychol. 39(2), 222–245.<br />

Brookes, K. J., Mill, J., Guindalini, C., Curran, S., Xu, X., Knight, J., Chen, C., Huang, Y., Sethna, V.,<br />

Taylor, E., Chen, W., Breen, G., et al. (2006). A common haplotype of the dopamine transporter<br />

gene associated with attention-deficit/hyperactivity disorder and interacting with maternal use of<br />

alcohol during pregnancy. Arch. Gen. Psychiatry 63, 74–81.<br />

Bruin, J. E., Gerstein, H. C., and Holloway, A. C. (2010). Long-term consequences of fetal and<br />

neonatal nicotine exposure: A critical review. Toxicol. Sci. 116(2), 364–374.<br />

Budhani, S., and Blair, R. J. R. (2005). Response reversal and children with psychopathic tendencies.<br />

J. Child Psychol. Psychiatry 46(9), 972–981.<br />

Bush, G., Luu, P., and Posner, M. I. (2000). Cognitive and emotional influences in anterior cingulate<br />

cortex. Trends Cogn. Sci. 4(6), 215–222.<br />

Butter, C. M., and Snyder, D. R. (1972). Alterations in aversive and aggressive behaviors following<br />

orbital frontal lesions in rhesus monkeys. Acta Neurobiol. Exp. 32, 525–565.<br />

Cannon, M., Huttunen, M. O., Tanskanen, A. J., Arseneault, L., Jones, P. B., and Murray, R. M.<br />

(2002). Perinatal and childhood risk factors for later criminality and violence in schizophrenia:<br />

Longitudinal, population-based study. Br. J. Psychiatry 180(6), 496–501.<br />

Cases, O., Seif, I., Grimsby, J., Gaspar, P., Chen, K., Pournin, S., Müller, U., Aguet, M., Babinet, C.,<br />

Shih, J. C., and De Maeyer, E. (1995). Aggressive behavior and altered amounts of brain serotonin<br />

and norepinephrine in mice lacking MAOA. Science 268, 1763–1766.<br />

Caspi, A., McClay, J., Moffitt, T. E., Mill, J., Martin, J., Craig, I. W., Taylor, A., and Poulton, R.<br />

(2002). Role of genotype in the cycle of violence in maltreated children. Science 297, 851–854.<br />

Caspi, A., Langley, K., Milne, B., Moffitt, T. E., O’Donovan, M., Owen, M. J., Polo Tomas, M.,<br />

Poulton, R., Rutter, M., Taylor, A., Williams, B., and Thapar, A. (2008). A replicated molecular<br />

genetic basis for subtyping antisocial behavior in children with attention-deficit/hyperactivity<br />

disorder. Arch. Gen. Psychiatry 65, 203–210.


9. Perinatal Factors in the Development of Aggression 245<br />

Chen, Y.-C. I., Choi, J.-K., Andersen, S. L., Rosen, B. R., and Jenkins, B. G. (2005). Mapping<br />

dopamine D2/D3 receptor function using pharmacological magnetic resonance imaging. Psychopharmacology<br />

180(4), 705–715.<br />

Chiavegatto, S., Dawson, V. L., Mamounas, L. A., Koliatsos, V. E., Dawson, T. M., and Nelson, R. J.<br />

(2001). Brain serotonin dysfunction accounts for aggression in male mice lacking neuronal nitric<br />

oxide synthase. Proc. Natl. Acad. Sci. USA 98(3), 1277.<br />

Cima, M., Smeets, T., and Jelicic, M. (2008). Self-reported trauma, cortisol levels, and aggression in<br />

psychopathic and non-psychopathic prison inmates. Biol. Psychiatry 78(1), 75–86.<br />

Clarke, A., and Schneider, M. (1993). Prenatal stress has long-term effects on behavioral responses to<br />

stress in juvenile rhesus monkeys. Dev. Psychobiol. 26(5), 293–304.<br />

Clarke, A., Wittwer, D., Abbott, D., and Schneider, M. (1994). Long-term effects of prenatal stress<br />

on HPA axis activity in juvenile rhesus monkeys. Dev. Psychobiol. 27(5), 257–269.<br />

Clarke, A., Soto, A., Bergholz, T., and Schneider, M. L. (1996). Maternal gestational stress alters<br />

adaptive and social behavior in adolescent rhesus monkey offspring. Infant Behav. Dev. 19(4),<br />

451–461.<br />

Cloninger, C. R. (1987). A systematic method for clinical description and classification of personality<br />

variants: A proposal. Arch. Gen. Psychiatry 44(6), 573.<br />

Coccaro, E. F., and Kavoussi, R. J. (1997). Fluoxetine and impulsive aggressive behavior in personality-disordered<br />

subjects. Arch. Gen. Psychiatry 54(12), 1081.<br />

Coccaro, E. F., and Siever, L. J. (2002). Pathophysiology and treatment of aggression.<br />

In “Neuropsychopharmacology: The Fifth Generation of Progress” (K. Davis, D. Charney,<br />

J. Coyle, and C. Nemeroff, eds.), pp. 1709–1723. Lippincott Williams & Wilkins, Philadephia.<br />

Coccaro, E. F., McCloskey, M. S., Fitzgerald, D. A., and Phan, K. L. (2007). Amygdala and<br />

orbitofrontal reactivity to social threat in individuals with impulsive aggression. Biol. Psychiatry<br />

62(2), 168–178.<br />

Cohen, M. A., Piquero, A. R., and Jennings, W. G. (2010). Studying the costs of crime across<br />

offender trajectories. Criminol. Public Policy 9(2), 279–305.<br />

Corkin, S. (1979). Hidden-figures-test performance: Lasting effects of unilateral penetrating head<br />

injury and transient effects of bilateral cingulotomy. Neuropsychologia 17(6), 585–605.<br />

Côté, S., Vaillancourt, T., Leblanc, J. C., Nagin, D. S., and Tremblay, R. E. (2006). The development<br />

of physical aggression from toddlerhood to pre-adolescence: A nation wide longitudinal study of<br />

Canadian children. J. Abnorm. Child Psychol. 34(1), 68–82.<br />

Crick, N. R., and Dodge, K. A. (1996). Social information-processing mechanisms in reactive and<br />

proactive aggression. Child Dev. 67(3), 993–1002.<br />

Dabbs, J. M., and Morris, R. (1990). Testosterone, social class, and antisocial behavior in a sample of<br />

4,462 men. Psychol. Sci. 1(3), 209.<br />

Dabbs, J. M., Frady, R. L., Carr, T. S., and Besch, N. F. (1987). Saliva testosterone and criminal<br />

violence in young adult prison inmates. Psychosom. Med. 49(2), 174.<br />

Davidson, R. J. (2000). Dysfunction in the neural circuitry of emotion regulation—A possible<br />

prelude to violence. Science 289(5479), 591–594.<br />

de Almeida, R. M. M., Ferrari, P. F., Parmigiani, S., and Miczek, K. A. (2005). Escalated aggressive<br />

behavior: Dopamine, serotonin and GABA. Eur. J. Pharmacol. 526(1–3), 51–64.<br />

Decety, J., Michalska, K. J., Akitsuki, Y., and Lahey, B. B. (2009). Atypical empathic responses in<br />

adolescents with aggressive conduct disorder: A functional MRI investigation. Biol. Psychol. 80<br />

(2), 203–211.<br />

Dennis, N. A., Need, A. C., Labar, K. S., Waters-Metenier, S., Cirulli, E. T., Kragel, J.,<br />

Goldstein, D. B., and Cabeza, R. (2010). COMT Val108/158 Met genotype affects neural but<br />

not cognitive processing in healthy individuals. Cereb. Cortex 20, 672–683.<br />

Donchin, E., and Coles, M. G. H. (1988). Is the p300 component a manifestation of con<strong>text</strong><br />

updating Behav. Brain Sci. 11, 355–372.


246 LaPrairie et al.<br />

D’Onofrio, B. M., Singh, A. L., Iliadou, A., Lambe, M., Hultman, C. M., Grann, M.,<br />

Neiderhiser, J. M., Långström, N., and Lichtenstein, P. (2010). Familial confounding of the<br />

association between maternal smoking during pregnancy and offspring criminality: A population-based<br />

study in Sweden. Arch. Gen. Psychiatry 67, 529–538.<br />

Dougherty, D. D., Bonab, A. A., Ottowitz, W. E., Livni, E., Alpert, N. M., Rauch, S. L., et al. (2006).<br />

Decreased striatal D1 binding as measured using PET and [11C]SCH 23,390 in patients with<br />

major depression with anger attacks. Depress Anxiety 23(3), 175–177.<br />

Dow-Edwards, D. L. (1989). Long-term neurochemical and neurobehavioral consequences of cocaine<br />

use during pregnancy. Ann. N. Y. Acad. Sci. 562, 280–289.<br />

Dow-Edwards, D., Freed, L. A., and Milhorat, T. H. (1988). Stimulation of brain metabolism by<br />

perinatal cocaine exposure. Brain Res. 470, 137–141.<br />

Driscoll, C. D., Streissguth, A. P., and Riley, E. P. (1990). Prenatal alcohol exposure: Comparability<br />

of effects in humans and animal models. Neurotoxicol. Teratol. 12(3), 231–237.<br />

Dwyer, J. B., McQuown, S. C., and Leslie, F. M. (2009). The dynamic effects of nicotine on the<br />

developing brain. Pharmacol. Ther. 122(2), 125–139.<br />

Ehrenkranz, J., Bliss, E., and Sheard, M. H. (1974). Plasma testosterone: Correlation with aggressive<br />

behavior and social dominance in man. Psychosom. Med. 36, 469–475.<br />

Eisenberger, N. I., Way, B. M., Taylor, S. E., Welch, W. T., and Lieberman, M. D. (2007).<br />

Understanding genetic risk for aggression: Clues from the brain’s response to social exclusion.<br />

Biol. Psychiatry 61, 1100–1108.<br />

Ekblad, M., Korkeila, J., Parkkola, R., Lapinleimu, H., Haataja, L., and Lehtonen, L. PIPARI Study<br />

Group (2010). Maternal smoking during pregnancy and regional brain volumes in preterm infants.<br />

J. Pediatr. 156(2), 185–190.<br />

Elliott, R., and Deakin, B. (2005). Role of the orbitofrontal cortex in reinforcement processing and<br />

inhibitory control: Evidence from functional magnetic resonance imaging studies in healthy<br />

human subjects. Int. Rev. Neurobiol. 65, 89–116.<br />

El-Sheikh, M., Erath, S. A., Buckhalt, J. A., Granger, D. A., and Mize, J. (2008). Cortisol and<br />

children’s adjustment: The moderating role of sympathetic nervous system activity. J. Abnorm.<br />

Child Psychol. 36, 601–611.<br />

Emery, N. J., Capitanio, J. P., Mason, W. A., Machado, C. J., Mendoza, S. P., and Amaral, D. G.<br />

(2001). The effects of bilateral lesions of the amygdala on dyadic social interactions in rhesus<br />

monkeys (Macaca mulatta). Behav. Neurosci. 115(3), 515–544.<br />

Ferrari, P. F., Van Erp, A. M. M., Tornatzky, W., and Miczek, K. A. (2003). Accumbal dopamine and<br />

serotonin in anticipation of the next aggressive episode in rats. Eur. J. Neurosci. 17(2), 371–378.<br />

Finger, E. C., Marsh, A. A., Mitchell, D. G., Reid, M. E., Sims, C., Budhani, S., et al. (2008).<br />

Abnormal ventromedial prefrontal cortex function in children with psychopathic traits during<br />

reversal learning. Arch. Gen. Psychiatry 65(5), 586.<br />

Fitzgerald, P. (1999). Long-acting antipsychotic medication, restraint and treatment in the management<br />

of acute psychosis. Australas. Psychiatry 33(5), 660–666.<br />

Frankle, W. G., Lombardo, I., New, A. S., Goodman, M., Talbot, P. S., Huang, Y., et al. (2005). Brain<br />

serotonin transporter distribution in subjects with impulsive aggressivity: A positron emission<br />

study with [11C] McN 5652. Am. J. Psychiatry 162(5), 915.<br />

Gerra, G., Zaimovic, A., Avanzini, P., Chittolini, B., Giucastro, G., Caccavari, R., et al. (1997).<br />

Neurotransmitter-neuroendocrine responses to experimentally induced aggression in humans:<br />

Influence of personality variable. Psychiatry Res. 66(1), 33–43.<br />

Gerstle, J. E., Mathias, C. W., and Stanford, M. S. (1998). Auditory P300 and self-reported impulsive<br />

aggression. Prog. Neuropsychopharmacol. Biol. Psychiatry 22(4), 575–583.<br />

Goeders, N. E., and Smith, J. E. (1983). Cortical dopaminergic involvement in cocaine reinforcement.<br />

Science 221, 773–775.


9. Perinatal Factors in the Development of Aggression 247<br />

<strong>Green</strong>berg, N., and Crews, D. (1983). Physiological ethology of aggression in amphibians and<br />

reptiles. In “Hormones and Aggressive Behavior” (B. Svare, ed.), pp. 469–506. Plenum, New York.<br />

Gutteling, B. M., de Weerth, C., Willemsen-Swinkels, S. H., Huizink, A. C., Mulder, E. J.,<br />

Visser, G. H., et al. (2005). The effects of prenatal stress on temperament and problem behavior<br />

of 27-month-old toddlers. Eur. Child Adolesc. Psychiatry 14(1), 41–51.<br />

Hawes, D. J., Brennan, J., and Dadds, M. R. (2009). Cortisol, callous-unemotional traits, and<br />

pathways to antisocial behavior. Curr. Opin. Psychiatry 22(4), 357–362.<br />

Heimburger, R. F., Whitlock, C. C., and Kalsbeck, J. E. (1966). Stereotaxic amygdalotomy for<br />

epilepsy with aggressive behavior. JAMA 198(7), 741–745.<br />

Henry, B., and Moffitt, T. E. (1997). Neuropsychological and neuroimaging studies of juvenile<br />

delinquency and adult criminal behavior. In “Handbook of Antisocial Behavior” (D. M. Stoff,<br />

J. Breiling, and J. D. Maser, eds.), pp. 280–288. Wiley, New York.<br />

Hicks, B., Krueger, R., Iacono, W., McGrue, M., and Patrick, C. (2004). Family transmission and<br />

heritability of externalizing disorders. Arch. Gen. Psychiatry 61, 922–928.<br />

Higley, J. D., Mehlman, P., Taub, D., Higley, S., Suomi, S. J., Linnoila, M., et al. (1992). Cerebrospinal<br />

fluid monoamine and adrenal correlates of aggression in free-ranging rhesus monkeys. Arch.<br />

Gen. Psychiatry 49(6), 436.<br />

Hubbard, J. A., Smithmyer, C. M., Ramsden, S. R., Parker, E. H., Flanagan, K. D., Dearing, K. F.,<br />

et al. (2002). Observational, physiological, and self-report measures of children’s anger: Relations<br />

to reactive versus proactive aggression. Child Dev. 73(4), 1101–1118.<br />

Huebner, T., Vloet, T. D., Marx, I., Konrad, K., Fink, G. R., Herpertz, S. C., et al. (2008).<br />

Morphometric brain abnormalities in boys with conduct disorder. J. Am. Acad. Child Adolesc.<br />

Psychiatry 47(5), 540–547.<br />

Huijbregts, S. C., Seguin, J. R., Zoccolillo, M., Boivin, M., and Tremblay, R. E. (2008). Maternal<br />

prenatal smoking, parental antisocial behavior, and early childhood physical aggression. Dev.<br />

Psychopathol. 20(2), 437–453.<br />

Iacono, W. G., Carlson, S. R., Malone, S. M., and McGue, M. (2002). P3 event-related potential<br />

amplitude and risk for disinhibitory disorders in adolescent boys. Arch. Gen. Psychiatry 59,<br />

750–757.<br />

Insel, T. R., and Winslow, J. T. (1998). Serotonin and neuropeptides in affiliative behaviors. Biol.<br />

Psychiatry 44, 207–219.<br />

Johns, J. M., Means, M. J., Bass, E., Means, L. W., Zimmerman, L. I., and McMillen, B. A. (1994).<br />

Prenatal exposure to cocaine: Effects on aggression in Sprague–Dawley rats. Dev. Psychobiol. 27<br />

(4), 227–239.<br />

Joseph, R. (1999). Environmental influences on neural plasticity, the limbic system, emotional<br />

development and attachment: A review. Child Psychiatry Hum. Dev. 29(3), 189–208.<br />

Kahn, R. S., Khoury, J., Nichols, W. C., and Lanphear, B. P. (2003). Role of dopamine transporter<br />

genotype and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and<br />

oppositional behaviors. J. Pediatr. 143, 104–110.<br />

Kappeler, L., and Meaney, M. J. (2010). Epigenetics and parental effects. Bioessays 32, 818–827.<br />

Kelly, S. J., Day, N., and Streissguth, A. P. (2000). Effects of prenatal alcohol exposure on social<br />

behavior in humans and other species. Neurotoxicol. Teratol. 22, 143–149.<br />

Kelly, S. J., Goodlett, C. R., and Hannigan, J. H. (2009). Animal models of fetal alcohol spectrum<br />

disorders: Impact of the social environment. Dev. Disabil. Res. Rev. 15(3), 200–208.<br />

Kennard, M. A. (1954). Effect of bilateral ablation of cingulate area on behaviour of cats.<br />

J. Neurophysiol. 18, 159–169.<br />

Kim-Cohen, J., Caspi, A., Taylor, A., Williams, B., Newcombe, R., Craig, I. W., and Moffitt, T. E.<br />

(2006). MAOA, maltreatment, and gene–environment interaction predicting children’s mental<br />

health: New evidence and a meta-analysis. Mol. Psychiatry 11, 903–913.


248 LaPrairie et al.<br />

Kirsch, P. (2005). Oxytocin Modulates Neural Circuitry for Social Cognition and Fear in Humans.<br />

J. Neurosci. 25(49), 11489–11493.<br />

Kruk, M. (1992). Ethology and pharmacology of hypothalamic aggression in the rat. Neurosci.<br />

Biobehav. Rev. 15, 527–538.<br />

Lachman, H. M., Papolos, D. F., Saito, T., Yu, Y. M., Szumlanski, C. L., and Weinshilboum, R. M.<br />

(1996). Human catechol-O-methyltransferase pharmacogenetics: Description of a functional<br />

polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 6,<br />

243–250.<br />

Langley, K., Turic, D., Rice, F., Holmans, P., van den Bree, M. B. M., Craddock, N., Kent, L.,<br />

Owen, M. J., O’Donovan, M. C., and Thapar, A. (2008). Testing for gene-environment interaction<br />

effects in attention deficit hyperactivity disorder and associated antisocial behavior. Am. J.<br />

Med. Genet. Part B 147, 49–53.<br />

Lauder, J. M. (1988). Neurotransmitters as morphogens. Prog. Brain Res. 74, 365–376.<br />

Lauder, J. M., and Bloom, F. E. (1974). Ontogeny of monoamine neurons in the locus coeruleus,<br />

raphe, nuclei, and substantia nigra of the rat. J. Comp. Neurol. 155, 469–481.<br />

Lidov, H. G. W., and Molliver, M. E. (1982a). An immunohistochemical study of serotonin neuron<br />

development in the rat: Ascending pathways and terminal fields. Brain Res. Bull. 8, 389–430.<br />

Lidov, H. G. W., and Molliver, M. E. (1982b). Immunohistochemical study of the development of<br />

serotonergic neurons in the rat CNS. Brain Res. Bull. 9, 559–604.<br />

Lindblad, F., and Hjern, A. (2010). ADHD after fetal exposure to maternal smoking. Nicotine Tob.<br />

Res. 12, 408–415.<br />

Lipp, H. P., and Hunsperger, R. W. (1978). Threat, attack and flight elicited by electrical stimulation<br />

of the ventromedial hypothalamus of the marmoset monkey callithrix jacchus. Brain Behav. Evol.<br />

15(4), 260–293.<br />

Liu, J. (2004). Prenatal and perinatal complications as predispositions to externalizing behavior.<br />

J. Prenat. Perinat. Psychol. Health 18(4), 301–311.<br />

Liu, J., Raine, A., Wuerker, A., Venables, P. H., and Mednick, S. (2009). The association of birth<br />

complications and externalizing behavior in early adolescents: Direct and mediating effects.<br />

J. Res. Adolesc. 19(1), 93–111.<br />

Lloyd, S. A., and Dixson, A. F. (1988). Effects of hypothalamic lesions upon the sexual and social<br />

behaviour of the male common marmoset (Callithrix jacchus). Brain Res. 463(2), 317–329.<br />

Loeber, R., and Pardini, D. (2008). Neurobiology and the development of violence: Common<br />

assumptions and controversies. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, 2491–2503.<br />

Loeber, R., Burke, J. D., Lahey, B. B., Winters, A., and Zera, M. (2000). Oppositional defiant and<br />

conduct disorder: A review of the past 10 years, part I. J. Am. Acad. Child Adolesc. Psychiatry 39<br />

(12), 1468–1484.<br />

Lopez-Duran, N. L., Hajal, N. J., Olson, S. L., Felt, B., and Vazquez, D. M. (2009). Hypothalamic<br />

pituitary adrenal axis functioning in reactive and proactive aggression in children. J. Abnorm.<br />

Child Psychol. 37(2), 169–182.<br />

Lorber, M. F. (2004). Psychophysiology of aggression, psychopathy, and conduct problems: A metaanalysis.<br />

Psychol. Bull. 130(4), 531–552.<br />

Machado, C. J., and Bachevalier, J. (2006). The impact of selective amygdala, orbital frontal cortex,<br />

or hippocampal formation lesions on established social relationships in rhesus monkeys (Macaca<br />

mulatta). Behav. Neurosci. 120(4), 761–786.<br />

Mann, J. J., McBride, P. A., Brown, R. P., Linnoila, M., Leon, A. C., DeMeo, M., et al. (1992).<br />

Relationship between central and peripheral serotonin indexes in depressed and suicidal psychiatric<br />

inpatients. Arch. Gen. Psychiatry 49(6), 442–446.<br />

Manuck, S. B., Flory, J. D., Ferrell, R. E., Mann, J. J., and Muldoon, M. F. (2000). A regulatory<br />

polymorphism of the monoamine oxidase-A gene may be associated with variability in aggression,<br />

impulsivity, and central nervous system serotonergic responsivity. Psychiatry Res. 95, 9–23.


9. Perinatal Factors in the Development of Aggression 249<br />

Marino, M. D., Bourdelat-Parks, B. N., Cameron Liles, L., and Weinshenker, D. (2005). Genetic<br />

reduction of noradrenergic function alters social memory and reduces aggression in mice. Behav.<br />

Brain Res. 161(2), 197–203.<br />

Marsh, A. A., Finger, E. C., Mitchell, D. G. V., Reid, M. E., Sims, C., Kosson, D. S., et al. (2008).<br />

Reduced amygdala response to fearful expressions in children and adolescents with callousunemotional<br />

traits and disruptive behavior disorders. Am. J. Psychiatry 165(6), 712–720.<br />

Marsh, A. A., Yu, H. H., Pine, D. S., and Blair, R. J. R. (2010). Oxytocin improves specific<br />

recognition of positive facial expressions. Psychopharmacology 209(3), 225–232.<br />

Mattson, M. P. (1988). Neurotransmitters in the regulation of neuronal cytoarchitecture. Brain Res.<br />

Rev. 13, 179–212.<br />

Maughan, B., Taylor, A., Caspi, A., and Moffitt, T. E. (2004). Prenatal smoking and early childhood<br />

conduct problems: Testing genetic and environmental explanations of the association. Arch. Gen.<br />

Psychiatry 61, 836–843.<br />

Mayes, L. C. (1994). Neurobiology of prenatal cocaine exposure effect on developing monoamine<br />

systems. Infant Ment. Health J. 15(2), 121–133.<br />

McBurnett, K., Lahey, B. B., Rathouz, P. J., and Loeber, R. (2000). Low salivary cortisol and<br />

persistent aggression in boys referred for disruptive behavior. Arch. Gen. Psychiatry 57(1), 38–43.<br />

McCormick, M. C. (1985). The contribution of low birth weight to infant mortality and childhood<br />

morbidity. N. Engl. J. Med. 312(2), 82–90.<br />

McDougle, C. J., Holmes, J. P., Carlson, D. C., Pelton, G. H., Cohen, D. J., and Price, L. H. (1998).<br />

A double-blind, placebo-controlled study of risperidone in adults with autistic disorder and other<br />

pervasive developmental disorders. Arch. Gen. Psychiatry 55(7), 633.<br />

Mednick, S. A., and Kandel, E. (1988). Genetic and perinatal factors in violence. In “Biological<br />

Contributions to Crime Causation” (S. A. Mednick and T. E. Moffitt, eds.), pp. 121–131.<br />

Martinus Nijhoff Publishing, Dordrecht, Netherlands.<br />

Miczek, K. A., Fish, E. W., De Bold, J. F., and De Almeida, R. M. (2002). Social and neural<br />

determinants of aggressive behavior: Pharmacotherapeutic targets at serotonin, dopamine and -<br />

aminobutyric acid systems. Psychopharmacology 163(3–4), 434–458.<br />

Miller, N. S., Gold, M. S., and Mahler, J. C. (1991). Violent behaviors associated with cocaine use:<br />

Possible pharmacological mechanisms. Int. J. Addict. 26(10), 1077–1088.<br />

Mirochnick, M., Meyer, J., Cole, J., Herren, T., and Zuckerman, B. (1991). Circulating catecholamine<br />

concentrations in cocaine-exposed neonates: A pilot study. Pediatrics 88, 481–485.<br />

Moffitt, T. E. (1993). Adolescence-limited and life-course-persistent antisocial behavior: A developmental<br />

taxonomy. Psychol. Rev. 100(4), 674–701.<br />

Moffitt, T. E., and Caspi, A. (2001). Childhood predictors differentiate life-course persistent and<br />

adolescence-limited antisocial pathways among males and females. Dev. Psychopathol. 13,<br />

355–375.<br />

Monoghan, E., and Glickman, S. (1992). Hormones and aggressive behavior. In “Behavioral Endocrinology”<br />

(J. Becker, S. Breedlove, and D. Crews, eds.). MIT Press, Cambridge, MA.<br />

Monuteaux, M. C., Blacker, D., Biederman, J., Fitzmaurice, G., and Buka, S. L. (2006). Maternal<br />

smoking during pregnancy and offspring overt and covert conduct problems: A longitudinal study.<br />

J. Child Psychol. Psychiatry 47, 883–890.<br />

Monuteaux, M. C., Biederman, J., Doyle, A. E., Mick, E., and Faraone, S. V. (2009). Genetic risk for<br />

conduct disorder symptom subtypes in an ADHD sample: Specificity to aggressive symptoms.<br />

J. Am. Acad. Child Adolesc. Psychiatry 48, 757–764.<br />

Mulder, E., de Medina, P., Huizink, A., Van den Bergh, B., Buitelaar, J., and Visser, G. (2002). Prenatal<br />

maternal stress: Effects on pregnancy and the (unborn) child. Early Hum. Dev. 70(1–2), 3–14.<br />

Nagin, D., and Tremblay, R. E. (1999). Trajectories of boys’ physical aggression, opposition, and<br />

hyperactivity on the path to physically violent and nonviolent juvenile delinquency. Child Dev.<br />

70, 1181–1196.


250 LaPrairie et al.<br />

Nelson, R. J., and Trainor, B. C. (2007). Neural mechanisms of aggression. Nat. Rev. Neurosci. 8(7),<br />

536–546.<br />

New, A. S., Buchsbaum, M. S., Hazlett, E. A., Goodman, M., Koenigsberg, H. W., Lo, J., et al. (2004).<br />

Fluoxetine increases relative metabolic rate in prefrontal cortex in impulsive aggression. Psychopharmacology<br />

176(3–4), 451–458.<br />

Nilsson, K. W., Sjöberg, R. L., Wargelius, H. L., Leppert, J., Lindström, L., and Oreland, L. (2007).<br />

The monoamine oxidase A (MAO-A) gene, family function and maltreatment as predictors of<br />

destructive behaviour during male adolescent alcohol consumption. Addiction 102, 389–398.<br />

Olweus, D. (1986). Aggression and hormones: Behavioral relationship with testosterone and adrenaline.<br />

In “Development of Antisocial and Prosocial Behavior” (D. Olweus, J. Block, and<br />

M. Radke-Yarrow, eds.), pp. 51–72. Academic press, New York.<br />

Olweus, D., Mattsson, A., Schalling, D., and Low, H. (1988). Circulating testosterone levels and<br />

aggression in adolescent males: A causal analysis. Psychosom. Med. 50, 261–272.<br />

Parsey, R. V., Oquendo, M. A., Simpson, N. R., Ogden, R. T., Van Heertum, R., Arango, V., et al.<br />

(2002). Effects of sex, age, and aggressive traits in man on brain serotonin 5-HT1A receptor<br />

binding potential measured by PET using [C-11] WAY-100635. Brain Res. 954(2), 173–182.<br />

Patrick, C. J. (2008). Psychophysiological correlates of aggression and violence: An integrative<br />

review. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363(1503), 2543–2555.<br />

Patterson, G. R., DeBaryshe, B. D., and Ramsey, E. (1989). A developmental perspective on<br />

antisocial behavior. Am. Psychol. 44, 329–335.<br />

Piquero, A., and Tibbetts, S. (1999). The impact of pre/perinatal disturbances and disadvantaged<br />

familial environment in predicting criminal offending. Stud. Crime Crime Prev. 8(1), 52–70.<br />

Poland, R. E., Lutchmansingh, P., Au, D., Edelstein, M., Lydecker, S., Hsieh, C., and<br />

McCracken, J. T. (1994). Exposure to threshold doses of nicotine in utero: I. Neuroendocrine<br />

response to restraint stress in adult male offspring. Life Sci. 55(20), 1567–1575.<br />

Popma, A., Doreleijers, T. A., Jansen, L. M., Van Goozen, S. H., Van Engeland, H., and<br />

Vermeiren, R. (2007a). The diurnal cortisol cycle in delinquent male adolescents and normal<br />

controls. Neuropsychopharmacology 32(7), 1622–1628.<br />

Popma, A., Vermeiren, R., Geluk, C. A. M. L., Rinne, T., van den Brink, W., Knol, D. L., et al.<br />

(2007b). Cortisol moderates the relationship between testosterone and aggression in delinquent<br />

male adolescents. Biol. Psychiatry 61(3), 405–411.<br />

Potegal, M., and Davidson, R. J. (2003). Temper tantrums in young children: 1. Behavioral composition.<br />

J. Dev. Behav. Pediatr. 24, 140–147.<br />

Radtke, K. M., Ruf, M., Gunter, H. M., Dohrmann, K., Schauer, M., Meyer, A., and Elbert, T.<br />

(2011). Transgenerational impact of intimate partner violence on methylation in the promoter of<br />

the glucocorticoid receptor. Translational Psychiatry 1, e21.<br />

Ragnauth, A. K., Devidze, N., Moy, V., Finley, K., Goodwillie, A., Kow, L. M., et al. (2005). Female<br />

oxytocin gene-knockout mice, in a semi-natural environment, display exaggerated aggressive<br />

behavior. Genes Brain Behav. 4, 229–239.<br />

Raine, A. (1993). The Psychopathology of Crime: Criminal Behavior as a Clinical Disorder.<br />

Academic, San Diego.<br />

Raine, A. (2002a). Biosocial studies of antisocial and violent behavior in children and adults:<br />

A review. J. Abnorm. Child Psychol. 30(4), 311–326.<br />

Raine, A. (2002b). The role of prefrontal deficits, low autonomic arousal and early health factors in<br />

the development of antisocial and aggressive behavior in children. J. Child Psychol. Psychiatry 43<br />

(4), 417–434.<br />

Raine, A., Venables, P., and Williams, M. (1990). Relationships between central and autonomic<br />

measures of arousal at age 15 years and criminality at age 24 years. Arch. Gen. Psychiatry 47(11),<br />

1003.


9. Perinatal Factors in the Development of Aggression 251<br />

Raine, A., Brennan, P., and Mednick, S. A. (1994). Birth complications combined with early<br />

maternal rejection at age 1 predispose to violent crime at age 18 years. Arch. Gen. Psychiatry<br />

51, 984–988.<br />

Retz, W., Rosler, M., Supprian, T., Retz-Junginger, P., and Thome, J. (2003). Dopamine D3 receptor<br />

gene polymorphism and violent behavior: Relation to impulsiveness and ADHD-related psychopathology.<br />

J. Neural Transm. 110(5), 561–572.<br />

Rice, F., Harold, G. T., Boivin, J., Hay, D. F., van den Bree, M., and Thapar, A. (2009). Disentangling<br />

prenatal and inherited influences in humans with an experimental design. Proc. Natl.<br />

Acad. Sci. USA 106(7), 2464–2467.<br />

Robinson, B. W. (1967). Vocalization evoked from forebrain in Macaca mulatta. Physiol. Behav. 2(4),<br />

345–346.<br />

Rolls, E. T., Hornak, J., Wade, D., and McGrath, J. (1994). Emotion-related learning in patients with<br />

social and emotional changes associated with frontal lobe damage. J. Neurol. Neurosurg. Psychiatry<br />

57(12), 1518–1524.<br />

Rose, R. M., Holladay, J. W., and Bernstein, I. S. (1971). Plasma testosterone, dominance rank and<br />

aggressive behavior in male rhesus monkeys. Nature 231, 366–368.<br />

Ross, G., Lipper, E. G., and Auld, P. A. (1990). Social competence and behavior problems in<br />

premature children at school age. Pediatrics 86, 391–397.<br />

Rowe, R., Maughan, B., Worthman, C. M., Costello, E. J., and Angold, A. (2004). Testosterone,<br />

antisocial behavior, and social dominance in boys: Pubertal development and biosocial interaction.<br />

Biol. Psychiatry 55, 546–552.<br />

Sabol, S. Z., Hu, S., and Hamer, D. (1998). A functional polymorphism in the monamine oxidase<br />

A gene promoter. Hum. Genet. 103, 273–279.<br />

Sampson, R. J., and Laub, J. H. (2003). Life-course desisters—Trajectories of crime among delinquent<br />

boys followed to Age 70. Criminology 41, 301–339.<br />

Scarpa, A., and Raine, A. (1997). Psychophysiology of anger and violent behavior. Psychiatr. Clin.<br />

North Am. 20(2), 375–394.<br />

Scerbo, A. S., and Kolko, D. J. (1994). Salivary testosterone and cortisol in disruptive children:<br />

Relationship to aggressive, hyperactive, and internalizing behaviors. J. Am. Acad. Child Adolesc.<br />

Psychiatry 33(8), 1174–1184.<br />

Schneider, M. L. (1992a). Delayed object permanence development in prenatally stressed monkey<br />

infants (Macaca mulatta). Occup. Ther. J. Res. 12, 96–110.<br />

Schneider, M. L. (1992b). Prenatal stress exposure alters postnatal behavioral expression under<br />

conditions of novelty challenge in rhesus monkey infants. Dev. Psychobiol. 25, 529–540.<br />

Sengupta, S. M., Grizenko, N., Schmitz, N., Schwartz, G., Ben Amor, L., Bellingham, J., de<br />

Guzman, R., Polotskaia, A., Ter Stepanian, M., Thakur, G., and Joober, R. (2006). COMT<br />

Val108/158Met gene variant, birth weight, and conduct disorder in children with ADHD.<br />

J. Am. Acad. Child Adolesc. Psychiatry 45, 1363–1369.<br />

Shepard, G. M. (1988). Neurobiology. 2nd edn., Oxford <strong>University</strong> Press, New York.<br />

Siegel, A., and Victoroff, J. (2009). Understanding human aggression: New insights from neuroscience.<br />

Int. J. Law Psychiatry 32(4), 209–215.<br />

Siever, L. J. (2008). Neurobiology of aggression and violence. Am. J. Psychiatry 165(4), 429.<br />

Slotkin, T. A., MacKillop, E. A., Rudder, C. L., Ryde, I. T., Tate, C. A., and Seidler, F. J. (2007).<br />

Permanent, sex-selective effects of prenatal or adolescent nicotine exposure, separately or sequentially,<br />

in rat brain regions: Indices of cholinergic and serotonergic synaptic function, cell signaling,<br />

and neural cell number and size at 6 months of age. Neuropsychopharmacology 32, 1082–1097.<br />

Sobrian, S. K., Vaughn, V. T., Ashe, W. K., Markovic, B., Djuric, V., and Jankovic, B. D. (1997).<br />

Gestational exposure to loud noise alters the development and post-atal responsiveness of<br />

humoral and cellular components of the immune system in offspring. Environ. Res. 73, 227–241.


252 LaPrairie et al.<br />

Soloff, P. H., Meltzer, C. C., Becker, C., Greer, P. J., Kelly, T. M., and Constantine, D. (2003).<br />

Impulsivity and prefrontal hypometabolism in borderline personality disorder. Psychiatry Res. 123<br />

(3), 153–163.<br />

Sood, B., Delaney-Black, V., Covington, C., Nordstrom-Klee, B., Ager, J., Templin, T., et al. (2001).<br />

Prenatal alcohol exposure and childhood behavior at age 6 to 7 years: I. Dose-response effect.<br />

Pediatrics 108(2), 34–44.<br />

Spear, L. P., Campbell, J., Snyder, K., Silveri, M., and Katovic, N. (1998). Animal behavior models.<br />

Increased sensitivity to stressors and other environmental experiences after prenatal cocaine<br />

exposure. Ann. N. Y. Acad. Sci. 846, 76–88.<br />

Stadler, C., Sterzer, P., Schmeck, K., Krebs, A., Kleinschmidt, A., and Poustka, F. (2007). Reduced<br />

anterior cingulate activation in aggressive children and adolescents during affective stimulation:<br />

Association with temperament traits. J. Psychiatr. Res. 41(5), 410–417.<br />

Sterzer, P., Stadler, C., Krebs, A., Kleinschmidt, A., and Poustka, F. (2005). Abnormal neural responses<br />

to emotional visual stimuli in adolescents with conduct disorder. Biol. Psychiatry 57(1), 7–15.<br />

Susman, E. J. (2006). Psychobiology of persistent antisocial behavior: Stress, early vulnerabilities and<br />

the attenuation hypothesis. Neurosci. Biobehav. Rev. 30(3), 376–389.<br />

Susman, E. J., Inoff-Germain, G., Nottelmann, E. D., Loriaux, D. L., Cutler, G. B., and<br />

Chrousos, G. P. (1987). Hormones, emotional dispositions, and aggressive attributes in young<br />

adolescents. Child Dev. 58(4), 1114–1134.<br />

Swanson, L. W. (2000). Cerebral hemisphere regulation of motivated behavior. Brain Res. 886(1–2),<br />

113–164.<br />

Takahashi, L. K., Baker, E. W., and Kalin, N. H. (1990). Ontogeny of behavioral and hormonal<br />

responses to stress in prenatally stressed male rat pups. Physiol. Behav. 47(2), 357–364.<br />

Thapar, A., Fowler, T., Rice, F., Scourfield, J., van den Bree, M., Thomas, H., Harold, G., and<br />

Hay, D. (2003). Maternal smoking during pregnancy and attention deficit hyperactivity disorder<br />

symptoms in offspring. Am. J. Psychiatry 160, 1985–1989.<br />

Thapar, A., Langley, K., Fowler, T., Rice, F., Turic, D., Whittinger, N., Aggleton, J., Van den<br />

Bree, M., Owen, M., and O’Donovan, M. (2005). Catechol O-methyltransferase gene variant and<br />

birth weight predict early-onset antisocial behavior in children with attention-deficit/hyperactivity<br />

disorder. Arch. Gen. Psychiatry 62, 1275–1278.<br />

Toro, R., Leonard, G., Lerner, J. V., Lerner, R. M., Perron, M., Pike, G. B., Richer, L., Veillette, S.,<br />

Pausova, Z., and Paus, T. (2008). Prenatal exposure to maternal cigarette smoking and the<br />

adolescent cerebral cortex. Neuropsychopharmacology 33(5), 1019–1027.<br />

Tow, P. M., and Whitty, C. W. (1953). Personality changes after operations on the cingulate gyrus in<br />

man. J. Neurol. Neurosurg. Psychiatry 16, 186–193.<br />

Tremblay, R. E. (2008). Development of physical aggression from early childhood to adulthood.<br />

In “Encyclopedia on Early Childhood Development” (R. E. Tremblay, R. G. Barr, R. De V. Peters,<br />

and M. Boivin, eds.), Rev. edn., pp. 1–6. Centre of Excellence for Early Childhood Development,<br />

Montreal, Quebec, http://www.child-encyclopedia.com/documents/TremblayANGxp_rev.pdf.<br />

Tremblay, R. E., Nagin, D. S., Séguin, J. R., Zoccolillo, M., Zelazo, P. D., Boivin, M., Pérusse, D., and<br />

Japel, C. (2004). Physical aggression during early childhood: Trajectories and predictors. Pediatrics<br />

114(1), e43–e50.<br />

Tunbridge, E. M., Harrison, P. J., and Weinberger, D. R. (2006). Catechol-o-methyltransferase,<br />

cognition, and psychosis: Val158Met and beyond. Biol. Psychiatry 60, 141–151.<br />

Van Bokhoven, I., Van Goozen, S. H. M., van Engeland, H., Schaal, B., Arseneault, L., Séguin, J. R.,<br />

et al. (2005). Salivary cortisol and aggression in a population-based longitudinal study of adolescent<br />

males. J. Neural Transm. 112(8), 1083–1096.<br />

van Bokhoven, I., van Goozen, S. H. M., van Engeland, H., Schaal, B., Arseneault, L., Séguin, J. R.,<br />

et al. (2006). Salivary testosterone and aggression, delinquency, and social dominance in a<br />

population-based longitudinal study of adolescent males. Horm. Behav. 50(1), 118–125.


9. Perinatal Factors in the Development of Aggression 253<br />

van Goozen, S., Matthys, W., Cohen-Kettenis, P., Buitelaar, J., and van Engeland, H. (2000).<br />

Hypothalamic-pituitary-adrenal axis and autonomic nervous system activity in disruptive children<br />

and matched controls. J. Am. Acad. Child Adolesc. Psychiatry 39(11), 1438–1445.<br />

Veenema, A. H., Torner, L., Blume, A., Beiderbeck, D. I., and Neumann, I. D. (2007). Low inborn<br />

anxiety correlates with high intermale aggression: Link to ACTH response and neuronal activation<br />

of the hypothalamic paraventricular nucleus. Horm. Behav. 51(1), 11–19.<br />

Veit, R., Flor, H., Erb, M., Hermann, C., Lotze, M., Grodd, W., et al. (2002). Brain circuits involved<br />

in emotional learning in antisocial behavior and social phobia in humans. Neurosci. Lett. 328(3),<br />

233–236.<br />

Virkkunen, M., Rawlings, R., Tokola, R., Poland, R. E., Guidotti, A., Nemeroff, C., et al. (1994). CSF<br />

biochemistries, glucose metabolism, and diurnal activity rhythms in alcoholic, violent offenders,<br />

fire setters, and healthy volunteers. Arch. Gen. Psychiatry 51(1), 20–27.<br />

Volavka, J. (1999). The neurobiology of violence: An update. J. Neuropsychiatry Clin. Neurosci. 11<br />

(3), 307.<br />

Volkow, N. D., Tancredi, L. R., Grant, C., Gillespie, H., Valentine, A., Mullani, N., et al. (1995).<br />

Brain glucose metabolism in violent psychiatric patients: A preliminary study. Psychiatry Res. 61<br />

(4), 243–253.<br />

Vom Saal, F. (1983). Models of early hormonal effects on intrasex aggression in mice. In “Hormones<br />

and Aggressive Behavior” (B. Svare, ed.), pp. 197–222. Plenum, New York.<br />

Wakschlag, L. S., Pickett, K. E., Cook, E., Jr., Benowitz, N. L., and Leventhal, B. L. (2002). Maternal<br />

smoking during pregnancy and severe antisocial behavior in offspring: A review. Am. J. Public<br />

Health 92(6), 966–974.<br />

Wakschlag, L. S., Kistner, E. O., Pine, D. S., Biesecker, G., Pickett, K. E., Skol, A. D., Dukic, V.,<br />

Blair, R. J., Leventhal, B. L., Cox, N. J., Burns, J. L., Kasza, K. E., et al. (2010). Interaction of<br />

prenatal exposure to cigarettes and MAOA genotype in pathways to youth antisocial behavior.<br />

Mol. Psychiatry 15(9), 928–937.<br />

Wallace, J. A., and Lauder, J. M. (1983). Development of the serotonergic system in the rat embryo:<br />

An immunocytochemical study. Brain Res. Bull. 10, 459–479.<br />

Ward, I. L., and Weisz, J. (2011). Differential effects of maternal stress on circulating levels of<br />

corticosterone, progesterone, and testosterone in male and female rat fetuses and their mothers.<br />

Endocrinology 114(5), 1635–1644.<br />

Weaver, K., Campbell, R., Mermelstein, R., and Wakschlag, L. (2007). Pregnancy smoking in<br />

con<strong>text</strong>: The influence of multiple levels of stress. Nicotine Tob. Res. 10(6), 1065–1073.<br />

Weinberg, J., Taylor, A. N., and Gianoulakis, C. (1996). Fetal ethanol exposure: Hypothalamicpituitary-adrenal<br />

and beta-endorphin responses to repeated stress. Alcohol Clin. Exp. Res. 20,<br />

122–131.<br />

White, J. L., Moffitt, T. E., Caspi, A., Bartusch, D. J., Needles, D. J., and Stouthamer-Loeber, M.<br />

(1994). Measuring impulsivity and examining its relationship to delinquency. J. Abnorm. Psychol.<br />

103(2), 192–205.<br />

Wingfield, J. C., and Hahn, T. P. (1994). Testosterone and sedentary behaviour in sedentary and<br />

migratory sparrows. Anim. Behav. 47(1), 77–89.<br />

Young, L. J., Lim, M. M., Gingrich, B., and Insel, T. R. (2001). Cellular mechanisms of social<br />

attachment. Horm. Behav. 40(2), 133–138.


Intentionally left as blank


10<br />

Neurocriminology<br />

Benjamin R. Nordstrom,* Yu Gao, † Andrea L. Glenn, ‡<br />

Melissa Peskin, } Anna S. Rudo-Hutt, } Robert A. Schug, <br />

Yaling Yang, k and Adrian Raine* ,},#<br />

*Department of Psychiatry, <strong>University</strong> of Pennsylvania, Philadelphia, USA<br />

† Department of Psychology, Brooklyn College, New York, USA<br />

‡ Department of Child and Adolescent Psychiatry, Institute of Mental Health,<br />

Singapore, Singapore<br />

} Department of Psychology, <strong>University</strong> of Pennsylvania, Philadelphia, USA<br />

Department of Criminal Justice, California <strong>State</strong> <strong>University</strong>, Long Branch,<br />

USA<br />

k Laboratory of Neuro Imaging, <strong>University</strong> of California, Los Angeles, USA<br />

# Department of Criminology, <strong>University</strong> of Pennsylvania, Philadelphia, USA<br />

I. Introduction<br />

II. Psychodynamic Theories<br />

III. Neuroimaging<br />

A. Structural imaging studies<br />

B. Functional imaging studies<br />

IV. Neuropsychological Testing<br />

V. Psychophysiological Evidence<br />

A. Electrocortical measures<br />

VI. Genetics<br />

A. Twin studies<br />

B. Adoption studies<br />

C. Molecular genetics<br />

D. ACE model<br />

E. Gene–environment interaction<br />

VII. Nongenetic Risk Factors<br />

A. Prenatal<br />

Advances in Genetics, Vol. 75 0065-2660/11 $35.00<br />

Copyright 2011, Elsevier Inc. All rights reserved.<br />

DOI: 10.1016/B978-0-12-380858-5.00006-X


256 Nordstrom et al.<br />

B. Perinatal risk factors<br />

C. Postnatal<br />

VIII. The Limitations and Potential of Neurocriminology<br />

IX. Modifiable Risk Factor Interventions<br />

X. Conclusion<br />

References<br />

ABSTRACT<br />

In the past several decades there has been an explosion of research into the<br />

biological correlates to antisocial behavior. This chapter reviews the state of<br />

current research on the topic, including a review of the genetics, neuroimaging,<br />

neuropsychological, and electrophysiological studies in delinquent and antisocial<br />

populations. Special attention is paid to the biopsychosocial model and<br />

gene–environment interactions in producing antisocial behavior. ß 2011, Elsevier Inc.<br />

I. INTRODUCTION<br />

In 1977, George Engel wrote an essay in Science to advocate for a new model in<br />

medicine that would serve as a corrective to the biomedical reductionism that he<br />

noted in the field and in psychiatry in particular (Engel, 1977). The model he<br />

suggested was called the biopsychosocial model and, in understanding the disease<br />

in question, took into account the biological aspect of the individual, their<br />

psychological state, and the social con<strong>text</strong> in which they exist. This model has<br />

since become the dominant paradigm in psychiatric treatment.<br />

In recent years, a tremendous amount of research has been done to<br />

elucidate the biological correlates and causes of antisocial behavior. This work<br />

has been conducted in an environment that has been, at times, hostile to this<br />

kind of research, as the dominant paradigm in criminology research has focused<br />

on social theories of crime. What we hope to accomplish in this chapter is to<br />

present the evidence for a biopsychosocial model of crime.<br />

We will present the data that argues that there is an inherited propensity<br />

for criminal behavior. The behavioral phenotype of those who criminally<br />

offend is demonstrably and obviously different from those who do not; we will<br />

show that their biological phenotypes are also different. We will marshal the data<br />

that suggest that the various brain areas that perform cognitive processes relevant<br />

to criminal offending are structurally and functionally different in antisocial<br />

people compared to others. We will discuss how these brain differences are also


10. Neurocriminology 257<br />

evident in techniques that elucidate the mind–body connection. We will also<br />

discuss how various social, or environmental, events can have multiplicative<br />

interactions with the biological risk factors to produce criminal offending.<br />

That there is not one standard diagnosis to identify the behavioral<br />

phenotype of interest to criminologists is a limitation of any review of this<br />

literature. Some research teams studying children use the diagnosis of oppositional<br />

defiant disorder or conduct disorder. Others use a broader category of<br />

disruptive behavior, attention deficit hyperactivity disorder. Researchers interested<br />

in adults may use the diagnosis of antisocial personality disorder to identify<br />

participants, while others might use the more stringent diagnosis of psychopathy.<br />

Other teams use self-reports of violence or aggression, a history of arrests, or<br />

various scores on personality inventories to identify the population of interest.<br />

Our stance on this is that although these differences make it difficult to directly<br />

compare the results of studies using different identifying criteria, all add the<br />

potential to better understand the biological correlates of problematic behaviors.<br />

II. PSYCHODYNAMIC THEORIES<br />

For the first part of the twentieth century, psychoanalytic models of crime and/or<br />

criminality (Holmes and Holmes, 1998; Wittels, 1937), cases of murder<br />

(Abrahamsen, 1973; Arieti and Schreiber, 1981; Bromberg, 1951; Cassity,<br />

1941; Evseef and Wisniewski, 1972; Karpman, 1951a,b; Lehrman, 1939;<br />

Morrison, 1979; Revitch and Schlesinger, 1981, 1989; Wertham, 1949, 1950;<br />

Wittels, 1937), and even homicide wound patterns (DeRiver, 1951) appeared in<br />

the psychiatric literature. A common feature of psychoanalytic criminological<br />

theory centers on unconscious processes (i.e., drives, instincts, and motivations,<br />

and the defense mechanisms used to control them which operate outside of a<br />

person’s conscious awareness) which are maladaptive and lead to antisocial and<br />

criminal behavior (Alexander and Staub, 1931).<br />

A number of psychodynamic theorists have posited that early problems<br />

of attachment to parents (especially mothers) can predispose individuals to<br />

unstable personality structure and later criminal offending (Bowlby, 1944,<br />

1969, 1973, 1980). Some theorists have posited that early experiences with<br />

rejecting mothers can lead children to mentally internalize fragments of this<br />

“bad mother,” which can then be externalized onto later female victims (Liebert,<br />

1972). More recent authors have posited that such malformed attachments<br />

might be at work in cases of serial homicide (Whitman and Akutagawa, 2004).<br />

Although some of these notions of psychodynamic theories may appear<br />

quaint compared to the astonishing technological achievements used in the<br />

studies described later, it is worth noting that the psychodynamic theorists may<br />

be using a different language to describe phenomena other researchers frame in


258 Nordstrom et al.<br />

more exacting biological terms. For example, we will later turn to discussions of<br />

how biology and environment can interact in ways that increase the likelihood<br />

of criminal offending. We will see that some of this data involves birth complications,<br />

and in what might be a partial affirmation of attachment theory,<br />

maternal rejection.<br />

III. NEUROIMAGING<br />

Using neuroanatomy as a tool to study criminal propensity is an idea that dates<br />

back to the early eighteenth century when the German physician Franz Joseph<br />

Gall developed phrenology. Phrenology purported to analyze the shape of cranial<br />

bones to make scientific inferences as to both the size and function of underlying<br />

brain areas. Later technological advances replaced the pseudoscience of phrenology,<br />

allowing for the scientific study of how brain structure and function<br />

relate to antisocial behavior. A comprehensive review of the neuroanatomic<br />

literature as it pertains to antisocial behavior is available (Yang et al., 2008).<br />

The two main technological advances that allowed images of the brain<br />

itself to be generated are computerized axial tomography (CAT) scans and<br />

magnetic resonance imaging (MRI) scans. CAT scans are produced using a series<br />

of X-rays taken along the axis of the body. The X-rays pass unevenly through<br />

tissues of different densities, allowing for distinctions between fluid, bone, and<br />

brain tissue to be made. A computer then assembles these slices into a sequence<br />

of cross-sectional images. MRI scans are created by using powerful magnetic<br />

fields to orient all the hydrogen atoms (primarily found in water molecules) in<br />

the brain in the same direction. A radiofrequency electromagnetic field is<br />

introduced which then produces a signal that is detected by the MRI scanner’s<br />

receiver. These signals are then assembled into high-resolution images that can<br />

distinguish the gray matter from the white matter of the brain. MRI scans don’t<br />

use radiation and produce more detailed pictures than do CAT scans, but they<br />

also take much longer to obtain and are much more expensive.<br />

A. Structural imaging studies<br />

Structural neuroimaging studies the size of brain regions of interest (ROI). One<br />

early study found that when CAT scans of the brains of sexual sadists were<br />

studied, about 50% of them had abnormal brain structures, especially in the<br />

temporal lobes (Langevin et al., 1988). Other researchers found that nearly 50%<br />

of 19 murder suspects studied had atrophic brains on CAT scan (Blake et al.,<br />

1995). Later studies using MRI scans found brain atrophy as well, especially in<br />

the frontotemporal region (Aigner et al., 2000; Sakuta and Fukushima, 1998).


10. Neurocriminology 259<br />

Another qualitative structural imaging study found that 6 of 10 of the violent<br />

psychiatric inpatients they studied had atrophic temporal regions (Chesterman<br />

et al., 1994).<br />

A number of studies in structural imaging have used larger samples and<br />

reported their findings in quantitative terms. Raine and colleagues viewed 21<br />

individuals with antisocial personality disorder and compared them to a matched<br />

group of substance users and normal controls (Raine et al., 2000, 2010). This<br />

work reported an 11% reduction in the gray matter of the prefrontal cortices of<br />

the antisocial group. A second study by this group found reduced prefrontal<br />

cortical gray matter volumes in unsuccessful psychopaths (i.e., psychopaths who<br />

had been criminally convicted at least once), compared to successful psychopaths<br />

(i.e., psychopaths who had never been convicted of a crime), and normal<br />

controls (Yang et al., 2005). In addition, Yang et al. revealed reduced cortical<br />

gray matter thickness in the frontal and temporal regions in psychopaths when<br />

compared to normal controls (Yang et al., 2009). Other groups have found that,<br />

compared to normal controls, subjects with antisocial personality disorder have<br />

smaller temporal lobes (Dolan et al., 2002; Laakso et al., 2002), as well as<br />

reductions in their dorsolateral, medial frontal, and orbitofrontal cortices<br />

(Laakso et al., 2002).<br />

One team of researchers demonstrated smaller gray matter volumes in<br />

the orbitofrontal and temporal lobes of children with conduct disorder compared<br />

to normal controls (Huebner et al., 2008). Reductions in gray matter concentration<br />

have also been observed in the frontal and temporal lobes of criminal<br />

psychopaths compared to normal controls (Muller et al., 2008). Along<br />

these same lines, another research group found insignificant prefrontal lobe<br />

volume reductions, but significant temporal lobe volume reductions, in conduct<br />

disordered children (Kruesi et al., 2004).<br />

Buried deep in the temporal lobe is the amygdala, which is associated<br />

with fear conditioning, and the hippocampus, a structure associated with<br />

learning and memory. Laakso and colleagues found, in a group of violent<br />

offenders with alcoholism and antisocial personality disorder, that smaller posterior<br />

hippocampus measures matched higher psychopathy rating scores (Laakso<br />

et al., 2000, 2001). Other researchers report that adolescents with conduct<br />

disorder demonstrate reduced gray matter volumes in the insula and amygdala<br />

compared to normal controls (Sterzer et al., 2007).<br />

A relatively new technique called diffusion tensor imaging (DTI) allows<br />

images to be taken of the structural integrity of the white matter tracts connecting<br />

various parts of the brain. One DTI study showed evidence of abnormal white<br />

matter tract structure in the frontotemporal regions of adolescents with disruptive<br />

behavior compared to normal controls (Li et al., 2005). A second study<br />

showed similar evidence of abnormal white matter tracts connecting the amygdalas<br />

and orbitofrontal cortices of criminal psychopaths when compared to


260 Nordstrom et al.<br />

normal controls (Craig et al., 2008). Other studies looking at abnormalities in<br />

connectivity have focused on white matter structures. Raine et al. (2003a,b)<br />

found that compared to a normal comparison group, psychopathic, antisocial<br />

subjects had a longer, thinner corpus callosum with overall increased volume.<br />

They also found a correlation between psychopathy scores and larger callosal<br />

volumes (Raine et al., 2003a).<br />

B. Functional imaging studies<br />

Not only does current technology allow us to study the structure and connectivity<br />

of brain regions, it also allows us to image the functioning of brain areas as<br />

well. One form of functional neuroimaging is photon emission tomography<br />

(PET). This technique relies on injecting subjects with radioactively labeled<br />

substance such as glucose. Images of their brains can then be obtained. Areas of<br />

higher radioactive signal have more glucose metabolism and are thought to be<br />

more active (Yang et al., 2008). A second form of functional neuroimaging is<br />

single photon emission tomography (SPECT). This form of imaging also<br />

involves the injection of a radioactive tracer. The camera detects the amount<br />

of radiation coming from different parts of the brain. These differences are due to<br />

differences in regional cerebral blood flow (rCBF) and are thought to reflect<br />

different levels of activity in various parts of the brain (Yang et al., 2008)<br />

Functional magnetic resonance imaging (fMRI) studies measure changes in<br />

blood oxygen in ROI in the brain before and after cognitive tasks are undertaken.<br />

These blood oxygen level dependent (BOLD) signals are used as a proxy for how<br />

active a region of the brain is. By comparing subjects of interest with matched<br />

controls, the patterns of activation or inactivation in the brain can be studied to<br />

learn how the functioning of various brain regions relates to the condition at<br />

hand (Yang et al., 2008).<br />

One early PET scan study showed that, compared to controls, antisocial<br />

subjects demonstrated reduced glucose metabolism in the prefrontal and temporal<br />

areas of their brains (Volkow et al., 1995). A SPECT study of aggressive<br />

psychiatric patients also found reduced rCBF in the prefrontal cortex, as well as<br />

increased blood flow to the left temporal and the anterior medial frontal cortices<br />

(Amen et al., 1996).<br />

Other PET studies have investigated how glucose metabolism responds<br />

to difficult cognitive tests, such as a continuous performance task (CPT). One<br />

group found that the number of impulsive–aggressive acts perpetrated by subjects<br />

with personality disorders, including antisocial personality disorder, was negatively<br />

correlated to glucose metabolism in the orbitofrontal, anterior medial<br />

frontal, and left anterior frontal cortices (Goyer et al., 1994). Raine et al.<br />

(1994a,b) found that after a CPT, a sample of murderers demonstrated reduced<br />

glucose metabolism in the anterior medial prefrontal, orbitofrontal, and superior


10. Neurocriminology 261<br />

frontal cortices compared to a normal comparison group (Raine et al., 1994b).<br />

A follow up study with a larger sample but a similar methodology found the same<br />

pattern of reduced glucose metabolism in the anterior frontal cortices, and in the<br />

amygdalas and hippocampi as well (Raine et al., 1997a).<br />

The amygdala is a structure in the brain that plays a significant role in<br />

emotion processing. This makes it an important structure in associative learning,<br />

in which individuals assign an affective valence to the consequences of their<br />

actions. These associations can be positive, such as learning to feel good after<br />

helping someone, or negative, such as learning to feel guilty or bad after harming<br />

someone. It has been theorized that associating harmful actions with the distress<br />

of others could thus discourage antisocial behavior (Blair, 2006a,b).<br />

A study using PET technology looked at a sample of normal controls, a<br />

sample of schizophrenic patients with a history of repeated violent offending and<br />

a sample of schizophrenic patients with a history of nonrepetitive violent offending<br />

(Wong et al., 1997). This team found that, compared to the normal controls,<br />

the patient samples had reduced glucose metabolism in the anterior inferior<br />

temporal lobes. This reduction was bilateral in the nonrepetitively violent<br />

group, but isolated to the left side in the repetitively violent group. Later, a<br />

research group using SPECT found that, compared to normal controls, antisocial<br />

populations have reduced rCBF to the frontal cortex and temporal cortex, and<br />

that psychopathy scores are negatively correlated with the degree of rCBF<br />

reduction to these areas (Soderstrom et al., 2000, 2002).<br />

One fMRI study looked at patterns of brain activation in 13 adolescent<br />

aggressive conduct disordered males and 14 matched controls as they looked at<br />

neutral pictures and pictures with a strong negative affective valence. It was<br />

found that when the conduct disordered youth viewed the distressing pictures<br />

they had significantly reduced activity to their left amygdalas compared to the<br />

control subjects (Sterzer et al., 2005). Similar findings have been described in<br />

adult populations (Kiehl et al., 2004; Muller et al., 2003).<br />

Another group used a similar methodology to study the reaction of a<br />

sample of 36 children and adolescents as they viewed photographs of neutral,<br />

angry, or fearful faces. 12 of the participants had callous–unemotional traits and<br />

oppositional defiant disorder or conduct disorder, 12 had attention deficit hyperactivity<br />

disorder and 12 were comparison subjects. When compared to the other<br />

two groups, the group with callous–unemotional traits demonstrated significantly<br />

reduced amygdala activation on viewing the fearful (but not the angry or<br />

neutral) faces (Marsh et al., 2008). In addition, in a functional connectivity<br />

analysis, the callous–unemotional children showed reduced connectivity between<br />

the ventromedial prefrontal cortex and the amygdala. Further, the degree<br />

of reduction in this connectivity was negatively correlated with the score on the<br />

scale that measured the degree of callous–unemotional traits. This is particularly<br />

interesting as the ventromedial prefrontal cortex has been implicated in


262 Nordstrom et al.<br />

processing punishment and reward (Rolls, 2000), affective theories of the mind<br />

(Shamay-Tsoory et al., 2005), response inhibition (Aron et al., 2004; Vollm<br />

et al., 2006), and emotional regulation (Ochsner et al., 2005).<br />

IV. NEUROPSYCHOLOGICAL TESTING<br />

Neuropsychological tests provide another method for testing the capabilities and<br />

functioning of various brain areas. One of the most consistent findings in the<br />

neuropsychological aspects of criminality is that antisocial populations have<br />

lower verbal IQs compared to nonantisocial groups (Brennan et al., 2003; Déry<br />

et al., 1999; Raine, 1993; Teichner and Golden, 2000). Researchers have found<br />

that verbal deficits on testing at age 13 predict delinquency at age 18 (Moffitt<br />

et al., 1994). A number of authors have found evidence that such neuropsychological<br />

deficits show interactive effects when they are present in children with<br />

social risk factors (Aguilar et al., 2000; Brennan et al., 2003; Raine, 2002a,b).<br />

Executive functioning is another neuropsychological function of interest<br />

in criminology (Moffitt, 1990, 1993). Executive functioning refers to the<br />

group of cognitive processes that produce goal-directed, flexible, and strategically<br />

effective behavior (Lezak et al., 2004; Luria, 1996; Spreen and Strauss, 1998).<br />

Executive dysfunction involves impairments in impulse control, self-regulation,<br />

abstract reasoning, concept formation, sustained attention, planning, organization,<br />

problem solving, and cognitive flexibility (Raine, 2002a,b). A meta-analysis<br />

of 39 studies incorporating data from 4589 individuals studied the relationship<br />

between executive dysfunction and antisocial behavior(Morgan and Lilienfeld,<br />

2000). These authors found significant effect sizes (d¼0.86 for juvenile delinquency<br />

and d¼0.46 for conduct disorder) for the association between antisocial<br />

behavior and executive dysfunction.<br />

Another neuropsychological test that has been studied in antisocial<br />

populations tests selective attention, or the ability to attend to one or more<br />

stimuli while ignoring others. The dichotic listening test is used to probe selective<br />

attention by having subjects wear headphones and then sending different auditory<br />

stimuli to each ear, while instructing them to respond to only 1 ton and ignore<br />

others. Both adult (Hare and Jutai, 1988) and juvenile (Raine et al., 1990a)<br />

populations with psychopathic traits have been shown to have abnormalities on<br />

this test when verbal stimuli are used. These researchers have hypothesized that<br />

this reduced lateralization of linguistic processes might indicate that people with<br />

psychopathic traits have a reduced use of language to regulate their behavior.<br />

Other neuropsychological tests have focused on how antisocial populations<br />

respond to affectively charged stimuli. Loney et al. (2003) found that<br />

juveniles with callous–unemotional traits showed slower reaction times after<br />

being presented with emotionally negative words, while those with impulsive<br />

traits showed faster reaction times to such stimuli. Adult psychopaths have been


10. Neurocriminology 263<br />

found to have deficits in passive-avoidance learning tasks (Newman and Kosson,<br />

1986) and adolescent psychopaths have been shown to demonstrate hyperresponsivity<br />

to rewards (Scerbo et al., 1990). Taken together, these data suggest<br />

that psychopathic individuals will be less sensitive to punishment and more<br />

sensitive to the possibility of rewards as a consequence to their behavior. Also,<br />

given the executive functioning literature, they may be less able to plan, act in a<br />

rationally self-interested fashion, control their impulses and respond flexibly to<br />

the various problems encountered in everyday life.<br />

V. PSYCHOPHYSIOLOGICAL EVIDENCE<br />

The autonomic underarousal and hyporesponsivity noted in various electrophysiological<br />

studies have given rise to fearlessness theory. This theory posits that the<br />

low level of arousal noted in the somewhat stressful testing situations can be<br />

taken as evidence of a lack of normal fear (Raine, 1993, 1997). An alternative to<br />

fearlessness theory is the stimulation-seeking theory, which presumes that the<br />

observed hypoarousal is experienced by affected individuals as unpleasant, and is<br />

compensated for using risk-taking/thrill-seeking behaviors. Supporting this hypothesis<br />

is the observation that 3-year-old children who show high levels of<br />

sensation seeking and lower levels of fearlessness demonstrate increased levels of<br />

aggression at age 11 (Raine et al., 1998).<br />

It is likely that stimulation-seeking and fearlessness explain some part of<br />

the low resting heart rate shown in antisocial youth, but a causal link between<br />

the low resting heart rate and criminal behavior is more elusive (Raine, 2002a,b).<br />

A third theory, the prefrontal deficit theory, argues that the low arousal seen<br />

arises from abnormalities in the prefrontal cortical–subcortical circuits involved<br />

with arousal and stress response (Raine, 2002a,b).<br />

A number of psychophysiological studies have also elucidated biological<br />

correlates of criminal behavior. These studies have typically focused on heart<br />

rate, skin conductance and electrocortical measurements. In-depth descriptions<br />

of the methodologies used in psychophysiological research are available<br />

(Cacioppo et al., 2007).<br />

A. Electrocortical measures<br />

1. Electroencephalogram (EEG)<br />

The electrical activity in the cerebral cortex can be measured by a noninvasive<br />

test, the EEG (Hugdahl, 2001). In an EEG, the subject has electrodes placed in<br />

specific points over the scalp. These electrodes detect the brain’s electrical<br />

impulses, which are then recorded and analyzed by a computer. The frequency<br />

and amplitude of the resultant signals are then interpreted.


264 Nordstrom et al.<br />

Increasing frequency is associated with increasing arousal, and lower<br />

frequency is associated with lower arousal (Hugdahl, 2001). Slower EEG activity<br />

in children and adolescents is associated with later criminal behavior (Mednick<br />

et al., 1981; Petersen et al., 1982). Raine and colleagues demonstrated that,<br />

compared to their peers with higher arousal, 15-year-old boys with lower arousal<br />

as measured by resting EEG were more likely to become criminals at age 24<br />

(Raine et al., 1990b). Children with externalizing and antisocial behaviors have<br />

been noted to demonstrate abnormal patterns of EEG asymmetry in their frontal<br />

lobes (Ishikawa and Raine, 2002; Santesso et al., 2006).<br />

It has been noted that dominant EEG frequencies increase with age<br />

(Dustman et al., 1999). The EEG abnormalities noted with respect to criminal<br />

behavior have been hypothesized to be due to cortical immaturity (Volavka,<br />

1987). It has been suggested that abnormal frontal EEG asymmetry might belie<br />

language and analytic reasoning deficits, thus impairing emotion regulation<br />

(Santesso et al., 2006).<br />

2. Event-related potentials (ERPs)<br />

A stimulus perceived by the brain will cause a change in the brain’s electrical<br />

activity. An ERP is a measure of the magnitude of that change after the<br />

presentation of specific stimuli. The change, or deflection, may be positive or<br />

negative in direction, and occurs within milliseconds of the onset of the stimulus.<br />

Typically an ERP is measured several times, and the average of all the trials is<br />

taken (Hugdahl, 2001). The P300 is a waveform that typically occurs approximately<br />

300 ms after the presentation of a stimulus. Early onset of drug abuse and<br />

criminal behavior has been shown to be related to smaller P300 amplitudes<br />

(Iacono and McGue, 2006). Other studies have demonstrated that greater<br />

negative amplitude at 100 ms and faster latency at 300 ms at age 15 are predict<br />

criminal behavior at age 24 (Raine et al., 1990b). A meta-analysis of studies of<br />

ERP in antisocial populations found that, in general, antisocial individuals have<br />

smaller P300 amplitudes and longer latencies (Gao and Raine, 2009).<br />

3. Low resting heart rate<br />

Low resting heart rate is the best-replicated biological correlate of antisocial<br />

behavior in juvenile samples (Ortiz and Raine, 2004). In a meta-analytic review<br />

of 29 samples, the average effect size was 0.56. This effect was demonstrated in<br />

both genders and irrespective of measurement technique (Raine, 1996). This<br />

relationship is not artifactual, as confounding variables such as height, weight,<br />

body composition, muscle tone, poor school performance, low IQ, hyperactivity,


10. Neurocriminology 265<br />

low attention, drug and alcohol use, participation in sports and exercise, social<br />

class, and family size and composition have all been ruled out (Farrington, 1997;<br />

Raine et al., 1990b, 1997b; Wadsworth, 1976).<br />

The finding that low resting heart rate predicts later crime has been<br />

replicated in the United <strong>State</strong>s, Germany, England, Canada, Mauritius, and New<br />

Zealand (Farrington, 1997; Mezzacappa et al., 1997; Moffitt and Caspi, 2001;<br />

Raine et al., 1997b; Rogeness et al., 1990; Schmeck and Poustra, 1993). In<br />

longitudinal studies, low resting heart rate has been shown to accurately identify<br />

individuals who are at risk for later developing antisocial behavior. This finding<br />

is specific for antisocial behavior (Rogeness et al., 1990) and has not been shown<br />

in other psychiatric syndromes.<br />

In the Cambridge Study in Delinquent Development, a series of six<br />

regression analyses were used to identify the best independent risk factors of<br />

violence (Farrington, 1997). Only two risk factors, low resting heart rate and<br />

poor concentration, were found, independently of all other risk factors, to predict<br />

violence. This same study found evidence of an interaction between low resting<br />

heart rate and several environmental risk factors (e.g., coming from a large<br />

family, having a teenaged mother, being of low socioeconomic status) in producing<br />

violent behavior. Lastly, it has been shown that having a high resting heart<br />

rate is negatively correlated with later violent behavior (i.e., a high resting heart<br />

rate is a protective factor against developing crime development) (Raine et al.,<br />

1995).<br />

4. Skin conductance<br />

The ease with which the skin can conduct electrical impulses is a function of<br />

sympathetic nervous system activity. Increased sweating leads to improved<br />

electrical conductance along the surface of the skin. In times of stress, sympathetic<br />

nervous system activity increases, and skin conductance will also increase.<br />

A classically conditioned fear response (as measured by an increase in skin<br />

conductance) can be produced by pairing a stressful stimulus, such as a noxious<br />

sound, with a neutral stimulus, such as a light turning on. Studying skin conductance<br />

under different paradigms can thus provide insight into the functioning of<br />

the sympathetic nervous system.<br />

Low skin conductance has been shown to be associated with conduct<br />

problems (Lorber, 2004). Boys with conduct disorder have been shown to have<br />

reduced fluctuations in skin conductance and impairments in conditioned fear<br />

responses (Fairchild et al., 2008; Herpertz et al., 2005). Longitudinal studies have<br />

demonstrated that reduced skin conductance arousal at age 15 has been associated<br />

with criminal offending at age 24 (Raine et al., 1995) and that low skin<br />

conductance at age 11 predicts institutionalization at age 13 (Kruesi et al., 1992).


266 Nordstrom et al.<br />

Impaired fear conditioning as measured by skin conductance at age 3 has been<br />

shown to predict aggression at age 8 and criminal behavior at age 23 (Gao et al.,<br />

2010a,b).<br />

Low sympathetic reactivity has been shown in psychopathy-prone adolescents<br />

and in children with conduct disorder and callous–unemotional traits<br />

(Anastassiou-Hadjichara and Warden, 2008; Kimonis et al., 2006; Loney et al.,<br />

2003). At age 3, having an abnormal skin conductance response to unpleasant<br />

stimuli is a risk factor for displaying psychopathy in adulthood (Glenn et al.,<br />

2007).<br />

VI. GENETICS<br />

As described in more detail in a separate chapter in this volume, a growing body<br />

of evidence has shown that there is a strong genetic contribution to juvenile<br />

delinquency (Popma and Raine, 2006). Although a number of genes have been<br />

shown to have an association with antisocial behavior, no one gene seems to<br />

“explain” criminal behavior (Goldman and Ducci, 2007). Investigating the<br />

potential genetic basis for complex behaviors is inherently complicated as they<br />

are likely to involve multiple genes, in contrast to conditions where there is a<br />

single-gene effect, as in classic Mendelian genetics (Uhl and Grow, 2004).<br />

Studies report heritability estimates that range widely, although the majority of<br />

investigators find heritability estimates that fall between 40% and 60%<br />

(Arsenault et al., 2003; Beaver et al., 2009; Jaffee et al., 2004, 2005; Lyons<br />

et al., 1995; Moffitt, 2005; Rhee and Waldman, 2002; Slutske et al., 1997).<br />

A. Twin studies<br />

One way to investigate a genetic component to a behavior is by comparing the<br />

frequency with which the disease occurs in different kinds of siblings. Monozygotic<br />

(also called identical) twins arise from a single fertilized ovum, meaning<br />

they have exactly the same genetic material. Dizygotic (also called fraternal)<br />

twins arise from two separate fertilized ova. Like any siblings, they share 50% of<br />

the same genes. A twin pair demonstrates concordance when both individuals<br />

demonstrate the condition in question, while twin pairs with only one affected<br />

individual are said to show discordance. The heritability of a disease can be<br />

estimated by comparing the rates of concordance and discordance in both<br />

monozygotic and dizygotic twins (Jorde et al., 1995).<br />

One study that investigated the genetic contribution to childhood<br />

antisocial and aggressive behavior investigated 605 families of 9- to 10-yearold<br />

twins and triplets (Baker et al., 2007). In this economically and ethnically<br />

diverse sample, such behavior was strongly heritable. Another study analyzed


10. Neurocriminology 267<br />

self-report measures of aggression in 182 monozygotic and 118 dizygotic twins<br />

(Coccaro et al., 1997a). The investigators found significant heritability for three<br />

out of the four forms of aggression studied. Although twin studies provide the<br />

opportunity study individuals with identical genetic make-ups or identical prenatal<br />

histories, other methodologies have also sought to gain understanding into<br />

the relative contribution of genes and parenting on later problematic behavior.<br />

B. Adoption studies<br />

Adoption studies provide another mechanism for studying the genetic versus the<br />

environmental contributions to antisocial behavior. In such studies, the characteristics<br />

of a child’s biological and adoptive parents are considered relative to<br />

the child’s own behavior. One early such study (Bohman, 1978) found evidence<br />

for a genetic predisposition to alcohol, but not to criminality, while another<br />

study from that same year (Cadoret, 1978) found evidence for heritability of<br />

antisocial behavior. A study of 862 Swedish male adoptees found that genetic<br />

influences were the most significant contributor to later criminal behavior<br />

(Cloninger et al., 1982). Another large sample of adoptees in Denmark similarly<br />

found strong evidence for a genetic propensity for criminal behavior (Gabrielli<br />

and Mednick, 1984).<br />

C. Molecular genetics<br />

Although it was previously noted complex behavioral syndromes don’t follow<br />

ordinary Mendelian patterns of inheritance, there is a notable exception to this.<br />

One group of researchers identified a family that demonstrated X-linked inheritance<br />

of borderline intellectual functioning and “abnormal behavior.” (Brunner<br />

et al., 1993) The behaviors exhibited by affected males included aggression, rape,<br />

exhibitionism, and arson. The researchers found that all had inherited a deficiency<br />

in the gene that coded for monoamine oxidase A (MAO-A).<br />

D. ACE model<br />

In behavioral genetic research, the heritability (i.e., the portion of the phenotypic<br />

variance explained by genetic factors) is represented by the letter “A.” The<br />

letter “C” is used to represent the family-wide, common, or shared environment.<br />

This includes influences that siblings would share, such as parenting styles or<br />

neighborhood characteristics. The letter “E” is used to represent environmental<br />

conditions uniquely encountered by an individual, such as getting a head injury.<br />

These are also called nonshared environmental influences.


268 Nordstrom et al.<br />

One such study investigated the genetic basis for psychopathy (Larsson<br />

et al., 2006). The researchers found that “A” accounted for 63% of the variance,<br />

“C” accounted for 0%, and “E” accounted for 37% of the variance.<br />

One meta-analytic study that used the ACE model found that, in<br />

children, genes (“A”) and shared environment (“C”) were equally important in<br />

explaining aggressive behavior (Miles and Carey, 1997). The researchers also<br />

found that heritability was slightly higher for males than for females, and that in<br />

adulthood the role of heritability increased while the role of shared environment<br />

fell to inconsequential levels.<br />

Another meta-analytic study of over 100 behavioral genetic studies<br />

showed that 40–50% of the variance of antisocial behavior is due to heritability,<br />

30% is due to the nonshared environmental influences, and 15–20% is due to<br />

shared environmental influences (Rhee and Waldman, 2002).<br />

It has been demonstrated that the influence of genes on criminal<br />

behavior varies over the life course (Goldman and Ducci, 2007). The majority<br />

of reports find that heritability estimates for antisocial behavior are lower, and<br />

shared environmental effects on antisocial behavior are higher, in childhood<br />

than in adolescence (Jacobson et al., 2002; Lyons et al., 1995; Miles and Carey,<br />

1997). It further seems that some genes affect the propensity for criminal<br />

involvement in adolescence, while others exert their effects in adulthood<br />

(Goldman and Ducci, 2007).<br />

The effects of genetics are also moderated by the type of criminal<br />

offending being considered. Heritability estimates for aggressive offending are<br />

higher than those for nonaggressive offending, such as rule breaking and theft<br />

(Eley et al., 2003). The opposite appears to be true for nonaggressive offending,<br />

which may be influenced more by shared environmental factors, such as family<br />

criminality, family poverty, and poor parenting, although research suggests that<br />

genetic influences also affect several of these risk factors (Moffitt, 2005).<br />

E. Gene–environment interaction<br />

Other studies have focused on how a person’s genetic endowment interacts with<br />

the environment in which the person lives. In the Swedish adoption study<br />

described above (Cloninger et al., 1982), the researchers found that if a person<br />

had both a biological parent and an adoptive parent who were criminals, then<br />

the person’s likelihood of criminal behavior was greater than the sum of the<br />

individual risks. In other words, there was a multiplicative effect of having a<br />

biological predisposition to crime and then being raised in a criminogenic<br />

environment.<br />

Another large study of gene–environment interaction identified people<br />

who carried a genotype that conferred a low expression of MAO-A activity<br />

(Caspi et al., 2002). The researchers looked at the people with high versus low


10. Neurocriminology 269<br />

MAO-A activity and also whether or not the individual had been abused as a<br />

child. They found evidence of a strong interaction between low MAO-A activity<br />

and childhood maltreatment in the likelihood of developing conduct disorder.<br />

VII. NONGENETIC RISK FACTORS<br />

There are many different types of the kinds of environmental risk factors<br />

captured by the ACE model. Researchers have identified a number of intriguing<br />

risk factors, some of which could be shared by siblings, some of which are less<br />

likely to be, which have been associated with later problematic behavior. These<br />

risk factors can be broken into those that arise during pregnancy (prenatal),<br />

those that arise during birth (perinatal) and those that arise in childhood<br />

(postnatal).<br />

A. Prenatal<br />

1. Minor physical anomalies (MPAs)<br />

MPAs are subtle physical defects such as having a curved fifth finger, a single<br />

palmar crease, low seated ears, or a furrowed tongue, are thought to arise from<br />

abnormalities in fetal development. These are thought to serve as biomarkers for<br />

abnormalities in neural development as well. MPAs may have a genetic basis, but<br />

they might also be due to anoxia, bleeding, or infection (Guy et al., 1983). Early<br />

studies showed an increase in the prevalence of MPAs in school-aged boys<br />

exhibiting behavioral problems (Halverson and Victor, 1976). MPAs have also<br />

been shown to be correlated to aggressive behaviors in children as young as 3<br />

years old (Waldrop et al., 1978). It has also been shown that MPAs identified at<br />

age 14 predict violence at age 17 (Arsenault et al., 2000).<br />

Mednick and Kandel studied MPAs in a sample of 129 12-year-old boys<br />

(Mednick and Kandel, 1988). They found MPAs were related to violent offending<br />

as assessed 9 years later when subjects were 21 years old. Interestingly, when<br />

subjects were divided into those from unstable (i.e., non-intact) homes versus<br />

those from stable homes, a biosocial interaction was observed. MPAs only<br />

predicted violence in those individuals raised in unstable home environments.<br />

Similarly, a study of 72 male offspring of psychiatrically ill parents found<br />

that those with both MPAs and family adversity had especially high rates of adult<br />

violent offending (Brennan et al., 1997). Another study showed that the presence<br />

of MPAs significantly interacted with environmental risk factors (e.g.,<br />

poverty, marital conflict) to predict conduct problems in adolescence (Pine<br />

et al., 1997).


270 Nordstrom et al.<br />

2. Tobacco<br />

There is a significant body of evidence that demonstrates that maternal smoking<br />

during pregnancy predisposes children to developing antisocial behavior<br />

(Wakschlag et al., 2002). Maternal prenatal smoking predicts externalizing behaviors<br />

in childhood and criminal behavior in adolescence (Fergusson et al., 1993,<br />

1998; Orlebeke et al., 1997; Rantakallio et al., 1992b;Wakschlaget al., 1997).<br />

Researchers have elucidated a clear dose-dependent relationship between smoking<br />

and later criminal behavior (Brennan et al., 1999; Maughan et al., 2001, 2004).<br />

Although the mechanism by which smoking produces these effects is<br />

unknown, basic science research has shown that the byproducts of smoking may<br />

affect the brain’s dopaminergic and noradrenergic systems (Muneoka et al.,<br />

1997) and glucose metabolism (Eckstein et al., 1997). Smoking may affect<br />

various brain structures, for example, the basal ganglia, cerebral, and cerebellar<br />

cortices—implicated in the deficits observed in violent offenders (Olds, 1997;<br />

Raine, 2002a,b).<br />

3. Alcohol<br />

There is also a great deal of evidence that prenatal exposure to alcohol predisposes<br />

individuals to antisocial behavior (Fast et al., 1999; Olson et al., 1997;<br />

Streissguth et al., 1996). Although Fetal Alcohol Syndrome (FAS) does not arise<br />

in all children exposed to alcohol in utero, evidence shows that children who do<br />

not display the <strong>full</strong> FAS syndrome can have some of the functional deficits<br />

characteristic of the syndrome (Schonfeld et al., 2005). Children who do not<br />

meet diagnostic criteria for FAS, yet were exposed to high levels of alcohol in<br />

utero, are at increased risk of antisocial behavior (Roebuck et al., 1999).<br />

B. Perinatal risk factors<br />

Obstetrical complications are untoward events that occur at the time of delivery<br />

and include such things as maternal preeclampsia, premature birth, low birth<br />

weight, use of forceps in delivery, transfer to a neonatal intensive care unit,<br />

anoxia, and low Apgar scores. Maternal complications have been shown to have<br />

deleterious effects on neonatal brain function (Liu, 2004; Liu and Wuerker,<br />

2005). Newborns who suffer obstetrical complications are more likely to exhibit<br />

externalizing behaviors at age 11 than those without complications (Liu et al.,<br />

2009). Obstetrical complication was found to mediate the relationship between<br />

low IQ and externalizing behaviors.<br />

It has also been demonstrated that obstetrical complications interact<br />

with other environmental factors to predict later antisocial behavior. Raine et al.<br />

(1994a,b) investigated a cohort 4269 Danish men. The investigators found that


10. Neurocriminology 271<br />

birth complication significantly interacted with severe maternal rejection (e.g.,<br />

efforts to abort the pregnancy, reporting the pregnancy as unwanted, or attempting<br />

to give up custody of the baby) to predict violent crime in adolescence (Raine<br />

et al., 1994a). This study has since been replicated in the United <strong>State</strong>s, Sweden,<br />

Finland, and Canada, and has repeatedly shown that birth complications interact<br />

with a number of psychosocial risk factors to produce antisocial behavior<br />

(Arsenault et al., 2002; Hodgins et al., 2001; Kemppainen et al., 2001; Tibbetts<br />

and Piquero, 1999).<br />

C. Postnatal<br />

Poor nutrition has been investigated as a risk factor for criminal behavior for<br />

some time. An association between aggressive behavior and vitamin and mineral<br />

deficiency has been described (Breakey, 1997; Werbach, 1995). The exact<br />

mechanism by which malnutrition affects later antisocial behavior is not well<br />

understood, it has been hypothesized that proteins or minerals may either<br />

regulate neurotransmitters and hormones, or ameliorate neurotoxins (Coccaro<br />

et al., 1997b; Liu and Raine, 2006).<br />

Although most studies have focused on nutrition in the postnatal period,<br />

one study investigated the role of malnutrition in the prenatal period in producing<br />

antisocial behavior (Neugebauer et al., 1999). This group studied the offspring of<br />

women who were pregnant during the German food blockade of Dutch cities in<br />

World War II. The blockade produced near starvation and severe food shortages.<br />

The researchers found that the male offspring of women who were in the first and<br />

second trimesters (but not the third trimester) of pregnancy during this time had<br />

two and a half times the rate of antisocial personality disorder than did the<br />

offspring of women who were not affected by food shortages.<br />

Another study of prenatal nutrition studied a sample of 11,875 pregnant<br />

women. Those women who ate less seafood (i.e., less than 340 g a week), which<br />

is rich in omega-3 fatty acids, had offspring that demonstrated significantly lower<br />

scores on a number of neurodevelopmental outcomes, including prosocial behavior,<br />

than the offspring of mothers who ate more seafood (Hibbeln et al.,<br />

2007).<br />

Studies have also shown that deficiency in nutrients such as proteins,<br />

zinc, iron, and docosahexaenoic acid (a component of omega-3 fatty acid) can<br />

lead to impaired brain functioning and a predisposition to antisocial behavior in<br />

childhood and adolescence (Arnold et al., 2000; Breakey, 1997; Fishbein, 2001;<br />

Lister et al., 2005; Liu and Raine, 2006; Rosen et al., 1985).<br />

Longitudinal studies have shown that malnutrition in infancy is associated<br />

with aggressive behavior and attentional deficits in childhood (Galler and<br />

Ramsey, 1989; Galler et al., 1983a,b). Liu et al. conducted a prospective longitudinal<br />

study to investigate how early malnutrition can predispose to behavior


272 Nordstrom et al.<br />

problems later in life (Liu and Raine, 2006). The researchers found that, compared<br />

to controls, children with protein, iron, or zinc deficiencies at age 3 had significantly<br />

more aggressive and hyperactive behavior at age 8, more antisocial behavior<br />

at age 11, and more excessive motor activity and conduct disorder at age 17.<br />

Significantly, this team also found a dose-dependent relationship between the<br />

extent of malnutrition and the extent of later behavior problems.<br />

1. Traumatic brain injury (TBI)<br />

Another risk factor that has been studied in relation to antisocial behavior is<br />

TBI. One group of investigators found that half of the juvenile delinquents in<br />

their sample had a history of TBI, and a third of the delinquents with TBI were<br />

thought by their parents to have neuropsychological sequelae from their injuries<br />

(Hux et al., 1998). Another study, which used more severe criteria in the<br />

definition of TBI than the previous study, found that 27.7% of the delinquents<br />

in their sample had a history of TBI (Carswell et al., 2004). A number of large,<br />

longitudinal studies of have repeatedly shown an increased incidence of delinquent<br />

behavior among youth with a history of TBI (Asarnow et al., 1991; Bloom<br />

et al., 2001; Butler et al., 1997; McAllister, 1992; Rantakallio et al., 1992a; Rimel<br />

et al., 1981; Rivera et al., 1994).<br />

VIII. THE LIMITATIONS AND POTENTIAL OF NEUROCRIMINOLOGY<br />

The field of neurocriminology has struggled to free itself from associations to<br />

earlier efforts to incorporate biology into the field of criminology. The reductionism<br />

of Lombroso’s biological positivism, the pseudoscience of phrenology,<br />

and the appalling racism of social Darwinists have all cast long shadows that<br />

have affected how contemporary efforts have been received by sociologically<br />

oriented criminologists.<br />

There is a danger that the kind of neurocriminological data could be<br />

used in a sensationalistic or superficial manner to implicate or exculpate individual<br />

offenders. Although the current state of imaging and other forms of biological<br />

research have not advanced to the point where an individual’s data could be<br />

confidently compared against a reliable database of normal controls, the possibility<br />

exists that such databases could be created and validated (Yang et al.,<br />

2008). Until such time, however, it is important to note that studies of the sort<br />

reviewed in this chapter cannot be taken to imply that any one biological factor<br />

causes criminal behavior. Rather, the presence of these factors only increases the<br />

probability that problematic behavior will be present in people with a given<br />

biological risk factor.


10. Neurocriminology 273<br />

We have now reviewed a number of studies that describe such increased<br />

probabilities of biological risk factors in criminal behavior. The sociological roots<br />

have crime have also been widely studied. This chapter has also reviewed a<br />

number of examples of how biological and environmental forces can interact to<br />

produce problematic behavior. We can see that criminal behavior can be investigated<br />

and explained at many different levels of abstraction.<br />

The psychiatrist and philosopher Kenneth Kendler illustrates this phenomenon<br />

with a hypothetical case of a pharmacologist running a randomized<br />

controlled trial of a medication for a psychiatric condition (Kendler, 2005).<br />

Although it is undoubtedly true that the medication is a molecule, and molecules<br />

are made up of atoms and atoms are made up of subatomic particles, it does not<br />

necessarily make sense to consult with a particle physicist in conducting the<br />

study. Thus, some levels of abstraction may be more or less efficient in explaining<br />

the phenomenon in question. However, each time a new level is identified, new<br />

possibilities for intervention arise as well. In uncovering biological leads relevant<br />

to crime the potential for new strategies for crime prevention are created as well.<br />

IX. MODIFIABLE RISK FACTOR INTERVENTIONS<br />

Not all risk factors for criminal behavior (e.g., male gender, having a biological<br />

parent with a history of criminal behavior) are modifiable. There are a number of<br />

risk factors (e.g., smoking, nutrition), however, that can be modified. Successful<br />

interventions have been developed to reduce prenatal alcohol exposure (Chang<br />

et al., 1999, 2005). Interventions have also been designed to reduce smoking in<br />

pregnancy, but these have been notably less effective than the interventions for<br />

alcohol use (Ershoff et al., 2004). Of note, women who persist in smoking<br />

throughout pregnancy are more likely than those who quit to have a personal<br />

history of conduct disorder (Kodl and Wakschlag, 2004).<br />

Other studies have sought to correct nutritional deficits. One randomized,<br />

double blind, placebo-controlled study was performed in a sample of 486<br />

public schoolchildren to see if a daily multivitamin and mineral supplement<br />

could reduce antisocial behavior (Schoenthaler and Bier, 2000). The researchers<br />

found that, compared to the placebo group, the treatment group had a 47%<br />

reduction in antisocial behavior after 4 months. Previously, this team had<br />

investigated the effect of vitamin and mineral supplementation in a group of<br />

juvenile delinquents confined to a correctional setting. The results of this<br />

randomized, double blind, placebo-controlled trial showed that, compared to<br />

the placebo group, the treatment group had significantly less violent and nonviolent<br />

antisocial behaviors (Schoenthaler et al., 1997). Another randomized,<br />

double blind, placebo-controlled trial of omega-3 fatty acid supplementation<br />

was done in a sample of 50 children. Compared to the placebo condition, the


274 Nordstrom et al.<br />

intervention group had a 42.7% reduction in conduct disorder problems (Stevens<br />

et al., 2003). A study using omega-3 fatty acid supplementation in ADHD failed<br />

to reveal a benefit (Hirayama et al., 2004).<br />

Other interventions address more than one risk factor at a time. For<br />

example, one highly successful intervention for prevention of later criminal and<br />

antisocial behavior involves home nursing visits for pregnant and new mothers.<br />

Parenting, health, and nutritional guidance are provided in the sessions (Olds<br />

et al., 1998). Other authors have also shown that prenatal education on nutrition,<br />

health, and parenting can lead to reductions in juvenile delinquency at age<br />

15 (Lally et al., 1988).<br />

Another multidimensional intervention was tested in a randomized,<br />

controlled fashion (Raine et al., 2003b). In this study, an intervention consisting<br />

of physical exercise and nutritional and educational enrichment was tested on a<br />

sample of 3–5 year olds. The study found that the intervention significantly<br />

reduced antisocial behavior at age 17 and criminal behavior at age 23. The<br />

intervention was found to be especially effective for the subgroup of children<br />

who displayed signs of malnutrition at age 3, suggesting the nutritional aspect of<br />

the treatment was particularly beneficial. The intervention was shown to produce<br />

lasting psychophysiological changes at age 11, including increased skin<br />

conductance, more orienting, and more arousal on EEG (Raine et al., 2001,<br />

2003b). These changes might then protect against the development of criminal<br />

offending (Raine et al., 1995, 1996).<br />

X. CONCLUSION<br />

Human beings are biological creatures. Whatever the truest essence of our souls<br />

may be, our subjective mental lives are mediated by and expressed through a<br />

system that is undeniably biological. This biological self exists in a specific social<br />

reality, which, in turn, shapes and alters the biological self in ways that will find<br />

some biological expression. What this chapter has sought to do is clarify how<br />

these biological aspects of the self can be used to understand, identify and,<br />

hope<strong>full</strong>y, predict individuals who criminally offend. Understanding these processes<br />

is the first step in then being able to modify risk factors or target at-risk<br />

individuals for services designed to attenuate their criminal propensity.<br />

References<br />

Abrahamsen, D. (1973). The Murdering Mind. Harper & Row, New York.<br />

Aguilar, B., Sroufe, A., et al. (2000). Distinguishing the early-onset/persistent and adolescent-onset<br />

antisocial behavior types: From birth to 6 years. Dev. Psychopathol. 12, 109–132.<br />

Aigner, M. R., Eher, R., et al. (2000). Brain abnormalities and violent behavior. J. Psychol. Human<br />

Sex. 11, 57–64.


10. Neurocriminology 275<br />

Alexander, F., and Staub, H. (1931). The Criminal, the Judge and the Public: A Psychological<br />

Analysis. The Macmillan Company, New York.<br />

Amen, D. G., Stubblefield, M., et al. (1996). Brain SPECT findings and aggressiveness. Ann. Clin.<br />

Psychiatry 8, 129–137.<br />

Anastassiou-Hadjichara, X., and Warden, D. (2008). Physiologically-indexed and self-perceived<br />

affective empathy in conduct-disordered children high and low on callous-unemotional traits.<br />

Child Psychiatry Hum. Dev. 39, 503–517.<br />

Arieti, S., and Schreiber, F. R. (1981). Multiple murders of a schizophrenic patient: A psychodynamic<br />

interpretation. J. Am. Acad. Psychoanal. 9(4), 501–524.<br />

Arnold, L. E., Pinkham, S. M., et al. (2000). Does zinc moderate essential fatty acid and amphetamine<br />

treatment of attention-deficit/hyperactivity disorder J. Child Adolesc. Psychopharmacol. 10,<br />

111–117.<br />

Aron, A. R., Robbins, T. W., et al. (2004). Inhibition and the right infrerior frontal cortex. Trends<br />

Cogn. Sci. 8(4), 170–177.<br />

Arsenault, L., Tremblay, R. E., et al. (2000). Minor physical anomalies and family adversity as risk<br />

factors for violent delinquency in adolescence. Am. J. Psychiatry 157, 917–923.<br />

Arsenault, L., Tremblay, R. E., et al. (2002). Obstetrical complications and violent delinquency:<br />

Testing two developmental pathways. Child Dev. 73, 496–508.<br />

Arsenault, L., Moffitt, T. E., et al. (2003). Strong genetic effects on cross-situational antisocial<br />

behaviour among 5-year-old children according to mothers, teachers, examiners-observers, and<br />

twins’ self-reports. J. Child Psychol. Psychiatry 44(6), 832–848.<br />

Asarnow, R., Satz, P., et al. (1991). Behavior problems and adaptive functioning in children with<br />

mild and severe closed head injury. J. Pediatr. Psychol. 16, 543–555.<br />

Baker, L. A., Jacobson, K. C., et al. (2007). Genetic and environmental bases of childhood antisocial<br />

behavior: A multi-informant twin study. J. Abnorm. Psychol. 116(2), 219–235.<br />

Beaver, K. M., DeLisi, M., et al. (2009). Gene environment interplay and delinquent involvement:<br />

Evidence of direct and indirect, and interactive effects. J. Adolesc. Res. 24, 147–168.<br />

Blair, R. J. R. (2006a). The emergence of psychopathy: Implications for the neuropsychological<br />

approach to developmental disorders. Cognition 101, 414–442.<br />

Blair, R. J. R. (2006b). Subcortical brain systems in psychopathy. In “Handbook of Psychopathy”<br />

(C. J. Patrick, ed.), pp. 296–312.<br />

Blake, P. Y., Pincus, J. H., et al. (1995). Neurologic abnormalities in murderers. Neurology 45,<br />

1641–1647.<br />

Bloom, D. R., Levin, H. S., et al. (2001). Lifetime and novel psychiatric disorders after pediatric brain<br />

injury. J. Am. Acad. Child Adolesc. Psychiatry 40, 572–579.<br />

Bohman, M. (1978). Some genetic aspects of alcoholism and criminality. A population of adoptees.<br />

Arch. Gen. Psychiatry 35(3), 269–276.<br />

Bowlby, J. (1944). Forty-four thieves: Their character and home life. Int. J. Psychoanal. 25, 19–52.<br />

Bowlby, J. (1969). Attachment and Loss: Volume 1 Attachment. Basic Books, New York.<br />

Bowlby, J. (1973). Attachment and Loss. Vol. 2. Separation: Anxiety and Anger. Basic Books,<br />

New York.<br />

Bowlby, J. (1980). Attachment and Loss: Vol. 3. Loss: Sadness and Depression. Basic Books,<br />

New York.<br />

Breakey, J. (1997). The role of diet and behaviour in childhood. J. Paediatr. Child Health 33, 190–194.<br />

Brennan, P. A., Mednick, S. A., et al. (1997). Biosocial interactions and violence: A focus on<br />

perinatal factors. In “Biosocial Bases of Violence” (A. Raine, P. Brennan, D. P. Farrington, and<br />

S. A. Mednick, eds.). Plenum, New York.<br />

Brennan, P. A., Grekin, E. R., et al. (1999). Maternal smoking during pregnancy and adult male<br />

criminal outcomes. Arch. Gen. Psychiatry 56, 215–219.


276 Nordstrom et al.<br />

Brennan, P. A., Hall, J., et al. (2003). Integrating biological and social processes in relation to earlyonset<br />

persistent aggression in boys and girls. Dev. Psychopathol. 39(2), 309–323.<br />

Bromberg, W. (1951). A psychological study of murder. Int. J. Psychoanal. 32, 117–127.<br />

Brunner, H. G., Nelen, M., et al. (1993). Abnormal behavior associated with a point mutation in the<br />

structural gene for monoamine oxidase A. Science 262(5133), 578–580.<br />

Butler, G., Chadwick, O., et al. (1997). A typology of psychosocial functioning in pediatric closedhead<br />

injury. Child Neuropsychol. 3, 98–133.<br />

Cacioppo, J. T., and Tassinary, L. G. et al. (eds.) (2007). In “Handbook of Psychophysiology”.<br />

Cambridge <strong>University</strong> Press, New York.<br />

Cadoret, R. J. (1978). Psychopathology in adopted-away offspring of biologic parents with antisocial<br />

behavior. Arch. Gen. Psychiatry 35(2), 176–184.<br />

Carswell, K., Maughan, B., et al. (2004). The psychosocial needs of young offenders and adolescents<br />

from an inner city area. J. Adolesc. 27, 415–428.<br />

Caspi, A., McClay, J., et al. (2002). Role of genotype in the cycle of violence in maltreated children.<br />

Science 297(5582), 851–854.<br />

Cassity, J. H. (1941). Personality study of 200 murderers. J. Crim. Psychopathol. 2, 296–304.<br />

Chang, G., Wilkins-Haug, L., et al. (1999). Brief intervention for alcohol use in pregnancy:<br />

A randomized trial. Addiction 94(10), 1499–1508.<br />

Chang, G., McNamara, T. K., et al. (2005). Brief intervention for prenatal alcohol use: A randomized<br />

trial. Obstet. Gynecol. 10(5 Pt. 1), 991–998.<br />

Chesterman, L., Taylor, P., et al. (1994). Multiple measures of cerebral state in dangerous mentally<br />

disordered inpatients. Crim. Behav. Ment. Health 4, 228–239.<br />

Cloninger, C. R., Sigvardsson, S., et al. (1982). Predisposition to petty criminality in Swedish<br />

adoptees. II. Cross-fostering analysis of gene-environment interaction. Arch. Gen. Psychiatry<br />

39(11), 1242–1247.<br />

Coccaro, E. F., Bergeman, C. S., et al. (1997a). Heritability of aggression and irritability: A twin study<br />

of the Buss-Durkee aggression scales in adult male subjects. Biol. Psychiatry 41(3), 273–284.<br />

Coccaro, E. F., Kavoussi, R. J., et al. (1997b). Serotonin function and anti-aggressive response to<br />

fluoxetine: A pilot study. Biol. Psychiatry 42, 546–552.<br />

Craig, M. C., Catani, M., et al. (2008). Altered connections on the road to psychopathy. Mol.<br />

Psychiatry 14(10), 946–953.<br />

DeRiver, J. P. (1951). The Sexual Criminal: A Psychoanalytic Study. Charles C. Thomas, Springfield,<br />

IL.<br />

Déry, M., Toupin, J., et al. (1999). Neuropsychological characteristics of adolescents with conduct<br />

disorder: Association with attention-deficit-hyperactivity and aggression. J. Abnorm. Child<br />

Psychol. 27(3), 225–236.<br />

Dolan, M., Deakin, J. F. W., et al. (2002). Quantitative frontal and temporal structural MRI studies in<br />

personality-disordered offenders and control subjects. Psychiatry Res. 116, 133–149.<br />

Dustman, R. E., Shearer, D. E., et al. (1999). Life-span changes in EEG spectral amplitude, amplitude<br />

variability and mean frequency. Clin. Neurophysiol. 110, 1399–1409.<br />

Eckstein, L. W., Shibley, I. J., et al. (1997). Changes in brain glucose levels and glucose transporter<br />

protein isoforms in alcohol- or nicotine- treated chick embryos. Brain Res. Dev. Brain Res. 15,<br />

383–402.<br />

Eley, T. C., Lichtenstein, P., et al. (2003). A longitudinal behavioral genetic analysis of the etiology<br />

of aggressive and nonaggressive antisocial behavior. Dev. Psychopathol. 15, 383–402.<br />

Engel, G. L. (1977). The need for a new medical model: A challenge for biomedicine. Science<br />

196(4286), 129–136.<br />

Ershoff, D. H., Ashford, T. H., et al. (2004). Helping pregnant women quit smoking: An overview.<br />

Nicotine Tob. Res. 6(2), S101–S105.


10. Neurocriminology 277<br />

Evseef, G. S., and Wisniewski, E. M. (1972). A psychiatric study of a violent mass murderer.<br />

J. Forensic Sci. 17(3), 371–376.<br />

Fairchild, G., Van Goozen, S. H. M., et al. (2008). Fear conditioning and affective modulation of the<br />

startle reflex in male adolescents with early-onset or adolescence-onset conduct disorder and<br />

health control subjects. Biol. Psychiatry 63, 279–285.<br />

Farrington, D. P. (1997). The relationship between low resting heart rate and violence. In “Biosocial<br />

Bases of Violence” (A. Raine, P. A. Brennan, D. P. Farrington, and S. A. Mednick, eds.),<br />

pp. 89–106. Prenum Press, New York.<br />

Fast, D. K., Conry, J., et al. (1999). Identifying Fetal Alcohol Syndrome among youth in the criminal<br />

justice system. J. Dev. Behav. Pediatr. 20, 370–372.<br />

Fergusson, D. M., Horwood, L. J., et al. (1993). Maternal smoking before and after pregnancy.<br />

Pediatrics 92, 815–822.<br />

Fergusson, D. M., Woodward, L. J., et al. (1998). Maternal smoking during pregnancy and psychiatric<br />

adjustment in late adolescence. Arch. Gen. Psychiatry 55, 721–727.<br />

Fishbein, D. (2001). Biobehavioral Perspectives in Criminology. Wadsworth/Thomson Learning,<br />

Belmont, CA.<br />

Gabrielli, W. F., and Mednick, S. A. (1984). Urban environment, genetics, and crime. Criminology<br />

22(4), 645–652.<br />

Galler, J. R., and Ramsey, F. (1989). A follow-up study of the influence of early malnutrition on<br />

development. J. Am. Acad. Child Adolesc. Psychiatry 26, 23–27.<br />

Galler, J. R., Ramsey, F., et al. (1983a). The influence of early malnutrition on subsequent behavioral<br />

development. II. Classroom behavior. J. Am. Acad. Child Adolesc. Psychiatry 22, 16–22.<br />

Galler, J. R., Ramsey, F., et al. (1983b). The influence of early malnutrition on subsequent behavioural<br />

development. I. Degree of impairment of intellectual performance. J. Am. Acad. Child<br />

Adolesc. Psychiatry 22, 8–15.<br />

Gao, Y., and Raine, A. (2009). P3 event-related potential impairments in antisocial and psychopathic<br />

individuals: A meta-analysis. Biol. Psychiatry 83, 199–210.<br />

Gao, Y., Raine, A., et al. (2010a). Poor childhood fear conditioning predisposes to adult crime. Am. J.<br />

Psychiatry 167, 56–60.<br />

Gao, Y., Raine, A., et al. (2010b). Reduced electrodermal fear conditioning from ages 3 to 8 years is<br />

associated with aggressive behavior at age 8 years. J. Child Psychol. Psychiatry 51(5), 550–558.<br />

Glenn, A. L., Raine, A., et al. (2007). Early temperamental and psychophysiological precursors of<br />

adult psychopathic personality. J. Abnorm. Psychol. 116, 508–518.<br />

Goldman, D., and Ducci, F. (2007). The genetics of psychopathic disorders. In “International<br />

Handbook of Psychopathic Disorders and the Law” (A. R. Felthous and H. Sa, eds.), Vol. 1,<br />

pp. 149–169. John Wiley and Sons, Ltd., West Sussex, England.<br />

Goyer, P. F., Andreason, P. J., et al. (1994). Positron-emission tomography and personality disorders.<br />

Neuropsychopharmacology 10, 21–28.<br />

Guy, J. D., Majorski, L. V., et al. (1983). The incidence of minor physical anomalies in adult male<br />

schizophrenics. Schizophr. Bull. 9, 571–582.<br />

Halverson, C. F., and Victor, J. B. (1976). Minor physical anomalies and problem behavior in<br />

elementary schoolchildren. Child Dev. 47, 281–285.<br />

Hare, R. D., and Jutai, J. W. (1988). Psychopathy and cerebral asymmetry in semantic processing.<br />

Pers. Indiv. Differ. 9, 329–337.<br />

Herpertz, S. C., Mueller, B., et al. (2005). Emotional responses in boys with conduct disorder. Am. J.<br />

Psychiatry 162, 1100–1107.<br />

Hibbeln, J. R., Davis, J. M., et al. (2007). Maternal seafood consumption in pregancy and neurodevelopmental<br />

outcomes in childhood (ALSPAC study): An observational cohort study. Lancet<br />

369(9561), 578–585.


278 Nordstrom et al.<br />

Hirayama, S., Hamazaki, T., et al. (2004). Effect of docosahexaenoic acid-containing food administration<br />

on symptoms of attention-deficit/hyperactivity disorder- a placebo-controlled doubleblind<br />

study. Eur. J. Clin. Nutr. 58, 467–473.<br />

Hodgins, S., Kratzer, L., et al. (2001). Obstetric complications, parenting, and risk of criminal<br />

behavior. Arch. Gen. Psychiatry 58, 746–752.<br />

Holmes, R. M., and Holmes, S. T. (1998). Serial Murder. Thousand Oaks, CA, Sage.<br />

Huebner, T., Vloet, T. D., et al. (2008). Morphometric brain abnormalities in boys with conduct<br />

disorder. J. Am. Acad. Child Adolesc. Psychiatry 47, 540–547.<br />

Hugdahl, K. (2001). Psychophysiology: The Mind-Body Perspective. Harvard <strong>University</strong> Press,<br />

Cambridge, MA.<br />

Hux, K., Bond, V., et al. (1998). Parental report of occurrences and consequences of traumatic brain<br />

injury among delinquent and non-delinquent youth. Brain Inj. 12, 667–681.<br />

Iacono, W. G., and McGue, M. (2006). Association between P3 event-related brain potential<br />

amplitude and adolescent problem behavior. Psychophysiology 43, 465–469.<br />

Ishikawa, S. S., and Raine, A. (2002). Psychophysiological correlates of antisocial behavior: A<br />

central control hypothesis. In “The Neurobiology of Criminal Behavior” (J. Glicksohn, ed.),<br />

pp. 187–229. Kluwer, Boston, MA.<br />

Jacobson, K. C., Prescott, C. A., et al. (2002). Sex differences in the genetic and environmental<br />

influences on the development of antisocial behavior. Dev. Psychopathol. 14, 395–416.<br />

Jaffee, S. R., Caspi, A., et al. (2004). Physical maltreatment victim to antisocial child: Evidence of an<br />

environmentally-mediated process. J. Abnorm. Psychol. 113, 44–55.<br />

Jaffee, S. R., Caspi, A., et al. (2005). Nature x nurture: Genetic vulnerabilities interact with physical<br />

maltreatment to promote conduct problems. Dev. Psychopathol. 17, 67–84.<br />

Jorde, L. B., Carey, J. C., et al. (1995). Medical Genetics. Mosby, New York.<br />

Karpman, B. (1951a). A psychoanalytic study of a case of murder. Psychoanal. Rev. 38(2), 139–157.<br />

Karpman, B. (1951b). A psychoanalytic study of a case of murder. Psychoanal. Rev. 38(2), 245–270.<br />

Kemppainen, L., Jokelainen, J., et al. (2001). The one-child family and violent criminality: A 31-year<br />

follow-up study of the Northern Finland 1966 birth cohort. Am. J. Psychiatry 158, 960–962.<br />

Kendler, K. S. (2005). Toward a philosophical structure for psychiatry. Am. J. Psychiatry 162, 433–440.<br />

Kiehl, K., Smith, A. M., et al. (2004). Temporal lobe abnormalities in semantic processing by<br />

criminal psychopaths as revealed by functional magnetic resonance imaging. Psychiatry Res.<br />

130, 27–42.<br />

Kimonis, E. R., Frick, P. J., et al. (2006). Psychopathy, aggression, and the processing of emotional<br />

stimuli in non-referred girls and boys. Behav. Sci. Law 24, 21–37.<br />

Kodl, M. M., and Wakschlag, L. S. (2004). Does a childhood history of externalizing problems predict<br />

smoking during pregnancy Addict. Behav. 29(2), 273–279.<br />

Kruesi, M. J. P., Hibbs, E. D., et al. (1992). A 2-year prospective follow-up study of children and<br />

adolescents with disruptive behavior disorders. Prediction by cerebrospinal fluid 5-hydroxyindoleacetic<br />

acid, homovanillic acid, and autonomic measures Am. J. Psychiatry 49(6), 429–435.<br />

Kruesi, M. J. P., Casanova, M. V., et al. (2004). Reduced temportal lobe volume in early-onset<br />

conduct disorder. Psychiatry Res. 132, 1–11.<br />

Laakso, M. P., Vaurio, O., et al. (2000). A volumetric MRI study of the hippocampus in type 1 and<br />

2 alcoholism. Behav. Brain Res. 109, 117–186.<br />

Laakso, M. P., Vaurio, O., et al. (2001). Psychopathy and the posterior hippocampus. Behav. Brain<br />

Res. 118, 187–193.<br />

Laakso, M. P., Gunning-Dixon, F., et al. (2002). Prefrontal volume in habitually violent subjects with<br />

antisocial personality disorder and type 2 alcoholism. Psychiatry Res. 114, 95–102.<br />

Lally, J. R., Mangione, P. L., et al. (1988). Long-range impact of an early intervention with low<br />

income children and their families. In “Parent Education as Early Childhood Intervention”<br />

(D. R. Powell, ed.), pp. 79–104. Ablex, Norwood, NJ.


10. Neurocriminology 279<br />

Langevin, R., Ben-Aron, M., et al. (1988). The sex killer. Ann. Sex Res. 1, 263–301.<br />

Larsson, H., Andershed, H., et al. (2006). A genetic factor explains most of the variation in the<br />

psychopathic personality. J. Abnorm. Psychol. 115(2), 2211–2230.<br />

Lehrman, P. R. (1939). Some unconscious determinants in homicide. Psychoanal. Q. 13(4),<br />

606–621.<br />

Lezak, M. D., Howieson, D. B., et al. (2004). Neuropsychological Assessment. 4th edn., Oxford<br />

<strong>University</strong> Press, New York.<br />

Li, T. Q., Mathews, V. P., et al. (2005). Adolescents with disruptive behavior disorder investigated<br />

using an optimized MR diffusion tensor imaging protocol. Ann. N. Y. Acad. Sci. 1064, 184–192.<br />

Liebert, J. A. (1972). Contributions of psychiatric consultation in the investigation of serial murder.<br />

Int. J. Offender Ther. Comp. Criminol. 29, 187–200.<br />

Lister, J. P., Blatt, G. J., et al. (2005). Effect of prenatal protein malnutrition on numbers of neurons in<br />

the principal cell layers of the adult rat hippocampal formation. Hippocampus 15, 393–403.<br />

Liu, J. (2004). Childhood externalizing behavior: Theory and implications. J. Child Adolesc. Psychiatr.<br />

Nurs. 17, 93–103.<br />

Liu, J., and Raine, A. (2006). The effect of childhood malnutrition on externalizing behaviors. Curr.<br />

Opin. Pediatr. 18, 565–570.<br />

Liu, J., and Wuerker, A. (2005). Biosocial bases of aggressive and violent behavior- implications for<br />

nursing studies. Int. J. Nurs. Stud. 42, 229–241.<br />

Liu, J., Raine, A., et al. (2009). The association of birth complications and externalizing behavior in<br />

early adolescents. J. Res. Adolesc. 19, 93–111.<br />

Loney, B. R., Frick, P. J., et al. (2003). Callous-unemotional traits, impulsivity and emotional<br />

processing in adolescents with antisocial behavior problems. J. Clin. Child Adolesc. Psychol. 32,<br />

66–80.<br />

Lorber, M. F. (2004). Psychophysiology of aggression, psychopathy, and conduct problems: A metaanalysis.<br />

Psychol. Bull. 130, 531–552.<br />

Luria, A. (1996). Higher Cortical Functions in Man. Basic Books, New York.<br />

Lyons, M. J., True, W. R., et al. (1995). Differential heritability of adult and juvenile traits. Arch.<br />

Gen. Psychiatry 52, 906–915.<br />

Marsh, A. A., Finger, E. C., et al. (2008). Reduced amygdala response to fearful expressions in<br />

children and adolescents with callous-unemotional traits and disruptive behavior disorders. Am. J.<br />

Psychiatry 165, 712–720.<br />

Maughan, B., Taylor, C., et al. (2001). Pregnancy smoking and childhood conduct problems: A causal<br />

association J. Child Psychol. Psychiatry 42, 1021–1028.<br />

Maughan, B., Taylor, A., et al. (2004). Prenatal smoking and early childhood conduct problems.<br />

Arch. Gen. Psychiatry 61, 836–843.<br />

McAllister, T. (1992). Neuropsychiatric sequelae of head injuries. Psychiatr. Clin. North Am. 15,<br />

661–665.<br />

Mednick, S. A., and Kandel, E. S. (1988). Congential determinants of violence. Bull. Am. Acad.<br />

Psychiatry Law 16, 101–109.<br />

Mednick, S. A., Volavka, J., et al. (1981). EEG as a predictor of antisocial behavior. Criminology 19,<br />

219–229.<br />

Mezzacappa, E., Tremblay, R. E., et al. (1997). Anxiety, antisocial behavior and heart rate regulation<br />

in adolescent males. J. Child Psychol. Psychiatry 38, 457–468.<br />

Miles, D. R., and Carey, G. (1997). Genetic and environmental architecture of human aggression.<br />

J. Pers. Soc. Psychol. 72, 207–217.<br />

Moffitt, T. E. (1990). Juvenile delinquency and attention-deficit disorder: Developmental trajectories<br />

from age three to fifteen. Child Dev. 61, 893–910.<br />

Moffitt, T. E. (1993). Adolescence-limited and life-course-persistent antisocial behavior: A developmental<br />

taxonomy. Psychol. Rev. 100(2), 674–701.


280 Nordstrom et al.<br />

Moffitt, T. E. (2005). The new look of behavioral genetics in developmental psychopathology: Geneenvironment<br />

interplay in antisocial behaviors. Psychol. Bull. 533–554.<br />

Moffitt, T. E., and Caspi, A. (2001). Childhood predictors differentiate life-course persistent and<br />

adolescence-limited antisocial pathways among males and females. Dev. Psychopathol. 13,<br />

355–375.<br />

Moffitt, T. E., Lynam, D. R., et al. (1994). Neuropsychological tests predicting persistent male<br />

delinquency. Criminology 32(2), 277–300.<br />

Morgan, A. B., and Lilienfeld, S. O. (2000). A meta-analytic review of the relationship between<br />

antisocial behavior and neuropsychological measures of executive function. Clin. Psychol. Rev.<br />

20, 113–136.<br />

Morrison, H. L. (1979). Psychiatric observations and interpretations of bite mark evidence in<br />

multiple murders. J. Forensic Sci. 24(2), 492–502.<br />

Muller, J. L., Sommer, M., et al. (2003). Abnormalities in emotion processing within cortical and<br />

subcortical regions in criminal psychopaths: Evidence from a functional magnetic imaging study<br />

using pictures with emotional content. Psychiatry Res. 54, 152–162.<br />

Muller, J. L., Ganssbauer, S., et al. (2008). Gray matter changes in right superior temporal gyrus in<br />

criminal psychopaths. Evidence from voxel-based morphometry. Psychiatry Res. 163, 213–222.<br />

Muneoka, K., Ogawa, T., et al. (1997). Prenatal nicotine exposure affects the development of the<br />

central serotonergic system as well as the dopaminergic system in rat offspring: Involvement of<br />

route of drug administrations. Brain Res. Dev. Brain Res. 102(1), 117–126.<br />

Neugebauer, R., Hoek, H. W., et al. (1999). Prenatal exposure to wartime famine and development of<br />

antisocial personality disorder in early adulthood. J. Am. Med. Assoc. 4, 479–481.<br />

Newman, J. P., and Kosson, D. S. (1986). Passive avoidance learning in psychopathic and nonpsychopathic<br />

offenders. J. Abnorm. Psychol. 95, 252–256.<br />

Ochsner, K. N., Beer, J. S., et al. (2005). The neural correlates of direct and reflected self-knowledge.<br />

Neuroimage 28(4), 797–814.<br />

Olds, D. (1997). Tobacco exposure and impaired development: A review of the evidence. Ment.<br />

Retard. Dev. Disabil. Res. Rev. 3, 257–269.<br />

Olds, D., Henderson, C. R. J., et al. (1998). Long-term effects of nurse home visitation on children’s<br />

criminal and antisocial behavior: 15-year follow-up of a randomized controlled trial. J. Am. Med.<br />

Assoc. 280, 1238–1244.<br />

Olson, H. C., Streissguth, A. P., et al. (1997). Association of prenatal alcohol exposure with<br />

behavioral and learning problems in early adolescence. J. Am. Acad. Child Adolesc. Psychiatry<br />

36, 1187–1194.<br />

Orlebeke, J. F., Knol, D. L., et al. (1997). Increase in child behavior problems resulting from maternal<br />

smoking during pregnancy. Arch. Environ. Health 52, 317–321.<br />

Ortiz, J., and Raine, A. (2004). Heart rate level and antisocial behavior in children and adolescents:<br />

A meta analysis. J. Am. Acad. Child Adolesc. Psychiatry 43, 154–162.<br />

Petersen, K. G. I., Matousek, M., et al. (1982). EEG antecedents of thievery. Acta Psychiatr. Scand.<br />

65, 331–338.<br />

Pine, D. S., Shaffer, D., et al. (1997). Minor physical anomalies: Modifiers of environmental risks for<br />

psychiatric impairment J. Am. Acad. Child Adolesc. Psychiatry 36, 395–403.<br />

Popma, A., and Raine, A. (2006). Will future forensic assessment be neurobiologic Child Adolesc.<br />

Psychiatr. Clin. N. Am. 36, 395–444.<br />

Raine, A. (1993). The Psychopathology of Crime: Criminal Behavior as a Clinical Disorder.<br />

Academic Press, San Diego, CA.<br />

Raine, A. (1996). Autonomic nervous system activity and violence. In “Neurobiological Approaches<br />

to Clinical Aggression Research” (D. M. Stoff and R. B. Cairns, eds.), pp. 145–168.Lawrence<br />

Erlbaum, Mahwah, NJ.


10. Neurocriminology 281<br />

Raine, A. (1997). Psychophysiology and antisocial behavior: A biosocial perspective an a prefrontal<br />

dysfunction hypothesis. In “Handbook of Antisocial Behavior” (D. M. Stoff, J. Breiling, and<br />

J. D. Maser, eds.), pp. 289–304. Wiley, New York.<br />

Raine, A. (2002a). Annotation: The role of prefrontal deficits, low autonomic arousal and early<br />

health factors in the development of antisocial and aggressive behavior in children. J. Child<br />

Psychol. Psychiatry 43, 417–434.<br />

Raine, A. (2002b). Biosocial studies of antisocial and violent behavior in children and adults:<br />

A review. J. Abnorm. Child Psychol. 304(4), 311–326.<br />

Raine, A., O’Brien, M., et al. (1990a). Reduced lateralization in verbal dichotic listening in adolescent<br />

psychopaths. J. Abnorm. Psychol. 99, 272–277.<br />

Raine, A., Venables, P. H., et al. (1990b). Relationships between CNS and ANS measures of arousal<br />

at age15 years as protective factors against criminal behavior at age 29 years. Am. J. Psychiatry<br />

152, 1595–1600.<br />

Raine, A., Brennan, P., et al. (1994a). Birth complications combined with early maternal rejection at<br />

age 1 year predispose to violent crime at age 18 years. Arch. Gen. Psychiatry 51, 984–988.<br />

Raine, A., Buchsbaum, M., et al. (1994b). Selective reductions in prefrontal glucose metabolism in<br />

murderers. Biol. Psychiatry 36, 365–373.<br />

Raine, A., Venables, P. H., et al. (1995). High autonomic arousal and electrodermal orienting at age<br />

15 years as protective factors against criminal behavior at age 29 years. Am. J. Psychiatry 152,<br />

1595–1600.<br />

Raine, A., Venables, P. H., et al. (1996). Better autonomic conditioning and faster electrodermal<br />

half-recovery time at age 15 years as possible protective factors against crime at age 29 years. Dev.<br />

Psychol. 32, 624–630.<br />

Raine, A., Buchsbaum, M., et al. (1997a). Brain abnormalities in murderess indicated by positron<br />

emission tomography. Biol. Psychiatry 42, 495–508.<br />

Raine, A., Venables, P. H., et al. (1997b). Low resting heart rate age 3 years predisposes to aggression<br />

at age 11 years: Evidence from the Mauritius Child Health Project. J. Am. Acad. Child Adolesc.<br />

Psychiatry 36, 1457–1464.<br />

Raine, A., Reynolds, C., et al. (1998). Fearlessness, stimulation-seeking, and large body size at age 3<br />

years as early predispositions to childhood aggression at age 11 years. Arch. Gen. Psychiatry 55,<br />

745–751.<br />

Raine, A., Lencz, T., et al. (2000). Reduced prefrontal gray matter volume and reduced autonomic<br />

activity in antisocial personality disorder. Arch. Gen. Psychiatry 57, 119–127.<br />

Raine, A., Venables, P. H., et al. (2001). Early educational and health enrichment at age 3–5 years is<br />

associated with increased autonomic and central nervous system arousal and orienting at age 11<br />

years: Evidene from the Mauritius Child Health Project. Psychophysiology 38, 254–266.<br />

Raine, A., Lencz, T., et al. (2003a). Corpus callosum abnormalities in psychopathic individuals. Arch.<br />

Gen. Psychiatry 160, 1627–1635.<br />

Raine, A., Mellingen, K., et al. (2003b). Effects of environmental enrichment at ages 3–5 years on<br />

schizotypal personality and antisocial behavior at ages 17 and 23 years. Am. J. Psychiatry 160,<br />

1627–1635.<br />

Raine, A., Lee, L., et al. (2010). Neurodevelopmental marker for limbic maldevelopment in antisocial<br />

personality disorder and psychopathy. Br. J. Psychiatry 197, 186–192.<br />

Rantakallio, P., Koiranen, M., et al. (1992a). Association of prenatal events, epilepsy, and central<br />

nervous system trauma with juvenile delinquency. Arch. Dis. Child. 67, 1459–1461.<br />

Rantakallio, P., Laara, E., et al. (1992b). Maternal smoking during pregnancy and delinquency of the<br />

offspring: An association without causation Int. J. Epidemiol. 21, 1106–1113.<br />

Revitch, E., and Schlesinger, L. B. (1981). The Psychopathology of Homicide. Charles C. Thomas,<br />

Springfield, IL.


282 Nordstrom et al.<br />

Revitch, E., and Schlesinger, L. B. (1989). Sex Murder and Sex Aggression: Phenomenology,<br />

Psychopathology, Psychodynamics and Prognosis. Charles C. Thomas, Springfield, IL.<br />

Rhee, S. H., and Waldman, I. D. (2002). Genetic and environmental influences on antisocial<br />

behavior: A meta-analysis of twin and adoption studies. Psychol. Bull. 128, 490–529.<br />

Rimel, R., Giordani, B., et al. (1981). Disability caused by minor head injury. Neurosurgery 9,<br />

1459–1461.<br />

Rivera, J., Jaffee, K., et al. (1994). Family functioning and children’s academic performance and<br />

behavior problems in the year following brain injury. Arch. Phys. Med. Rehabil. 75, 369–379.<br />

Roebuck, T. M., Mattson, S. N., et al. (1999). Behavioral and psychosocial profiles of alcohol-exposed<br />

children. Alcohol. Clin. Exp. Res. 23, 1070–1076.<br />

Rogeness, G. A., Cepeda, C., et al. (1990). Differences in heart rate and blood pressure in children<br />

with conduct disorder, major depression and separation anxiety. Psychiatry Res. 33, 199–206.<br />

Rolls, E. T. (2000). The orbitofrontal cortex and reward. Cereb. Cortex 10, 284–294.<br />

Rosen, G. M., Deinard, A. S., et al. (1985). Iron deficiency among incarcerated juvenile delinquents.<br />

J. Adolesc. Health Care 6, 419–423.<br />

Sakuta, A., and Fukushima, A. (1998). A study on abnormal findings pertaining to the brain in<br />

criminals. Int. Med. J. 5, 283–292.<br />

Santesso, D. L., Reker, D. L., et al. (2006). Frontal electroencephalogram activation asymmetry,<br />

emotional intelligence, and externalizing behaviors in 10-year-old children. Child Psychiatry Hum.<br />

Dev. 36, 311–328.<br />

Scerbo, A., Raine, A., et al. (1990). Reward dominance and passive avoidance learning in adolescent<br />

psychopaths. J. Abnorm. Child Psychol. 18, 451–463.<br />

Schmeck, K., and Poustra, F. (1993). Psychophysiologische Reacktionsmuster und psychische auffalligkeiten<br />

im kindesalter. In “Bioloogische Psychiatrie Der Gegenwart” (P. Baumann, ed.).<br />

Springer-Verlag, Wien.<br />

Schoenthaler, S. J., and Bier, I. D. (2000). The effect of vitamin-mineral supplementation on<br />

juvenile delinquency among American schoolchildren: A randomized double blind placebocontrolled<br />

trial. J. Altern. Complement. Med. 6, 19–29.<br />

Schoenthaler, S. J., Amos, S. P., et al. (1997). The effect of randomized vitamin-mineral supplementation<br />

on violent and non-violent antisocial behavior among incarcerated juveniles. J. Nutr.<br />

Environ. Med. 7, 343–352.<br />

Schonfeld, A. M., Mattson, S. N., et al. (2005). Moral maturity and delinquency after prenatal<br />

alcohol exposure. J. Stud. Alcohol 6, 19–29.<br />

Shamay-Tsoory, S. G., Tomer, R., et al. (2005). Impaired ’affective theory of mind’ is associated with<br />

right ventromedial prefrontal damage. Cogn. Behav. Neurol. 18(1), 55–67.<br />

Slutske, W. S., Heath, A. C., et al. (1997). Modeling genetic and environmental influences in the<br />

etiology of conduct disorder: A study of 2,682 adult twin pairs. J. Abnorm. Psychol. 106, 266–279.<br />

Soderstrom, H., Tullberg, M., et al. (2000). Reduced regional cerebral blood flow in non-psychotic<br />

violent offenders. Psychiatry Res. 98, 29–41.<br />

Soderstrom, H., Hultin, L., et al. (2002). Reduced frontotemporal perfusion in psychopathic personality.<br />

Psychiatry Res. 114, 81–94.<br />

Spreen, O., and Strauss, E. (1998). A Compendium of Neuropsychological Tests. 2nd edn., Oxford<br />

<strong>University</strong> Press, New York.<br />

Sterzer, P., Stadler, C., et al. (2005). Abnormal neural responses to emotional visual stimuli in<br />

adolescents with conduct disorder. Biol. Psychiatry 57, 7–15.<br />

Sterzer, P., Stadler, C., et al. (2007). A structural neural deficit in adolescents with conduct disorder<br />

and its association with lack of empathy. Neuroimage 37, 335–342.<br />

Stevens, L., Zhang, W., et al. (2003). EFA supplementation in children with inattention, hyperactivity,<br />

and other disruptive behaviors. Lipids 38, 1007–1021.


10. Neurocriminology 283<br />

Streissguth, A. P., Barr, H. M., et al. (1996). Understanding the Occurrence of Secondary Disabilities<br />

in Clients with Fetal Alcohol Syndrome (FAS) and Fetal Alcohol Effects (FAE). Centers for<br />

Disease Control and Prevention, Washington, D.C.<br />

Teichner, G., and Golden, C. J. (2000). The relationship of neuropsychological impairment to<br />

conduct disorder in adolescence: A conceptual review. Aggress. Violent Behav. 5(6), 509–528.<br />

Tibbetts, S. G., and Piquero, A. R. (1999). The influence of gender, low birth weight, and disadvantaged<br />

environment in predicting early onset of offending: A test of Moffitt’s interactional<br />

hypothesis. Criminology 37(4), 843–878.<br />

Uhl, G. R., and Grow, R. W. (2004). The burden of complex genetics in brain disorders. Arch. Gen.<br />

Psychiatry 61, 223–229.<br />

Volavka, J. (1987). Electroencephalogram among criminals. In “The Causes of Crime:<br />

New Biological Approaches” (S. A. Mednick, T. E. Moffitt, and S. Stack, eds.), pp. 137–145.<br />

Cambridge <strong>University</strong> Press, Cambridge.<br />

Volkow, N. D., Tancredi, L. R., et al. (1995). Brain glucose metabolism in violent psychiatric<br />

patients: A preliminary study. Psychiatry Res. 61, 243–253.<br />

Vollm, B., Richardson, P., et al. (2006). Serotonergic modulation of neuronal responses to behavioural<br />

inhibition and reinforcing stimuli: An fMRI study in healthy volunteers. Eur. J. Neurosci.<br />

23, 552–560.<br />

Wadsworth, M. E. J. (1976). Delinquency, pulse rate and early emotional deprivation. Br. J. Criminol.<br />

16, 245–256.<br />

Wakschlag, L. S., Lahey, B. B., et al. (1997). Maternal smoking during pregnancy and the risk of<br />

conduct disorder in boys. Arch. Gen. Psychiatry 54, 670–676.<br />

Wakschlag, L. S., Pickett, K. E., et al. (2002). Maternal smoking during pregnancy and severe<br />

antisocial behavior in offspring: A review. Am. J. Public Health 92, 966–974.<br />

Waldrop, M. F., Bell, R. Q., et al. (1978). Newborn minor physical anomalies predict short attention<br />

span, peer aggression, and impulsivity at age 3. Science 199, 563–564.<br />

Werbach, M. (1995). Nutritional influences on aggressive behavior. J. Orthomol. Med. 7, 45–51.<br />

Wertham, F. (1949). The Show of Violence. Doubleday & Company, Garden City, NY.<br />

Wertham, F. (1950). Dark Legend. Doubleday & Company, Garden City, NY.<br />

Whitman, T. A., and Akutagawa, D. (2004). Riddles in serial murder: A synthesis. Aggress. Violent<br />

Behav. 9, 693–703.<br />

Wittels, F. (1937). The criminal psychopath in the psychoanalytic system. Psychoanal. Rev. 24(1),<br />

276–291.<br />

Wong, M. T., Fenwick, P. B., et al. (1997). Positron emission tomography in male violent offenders<br />

with schizophrenia. Psychiatry Res. 68, 111–123.<br />

Yang, Y., Raine, A., et al. (2005). Volume reduction in prefrontal gray matter in unsuccessful criminal<br />

psychopaths. Biol. Psychiatry 57, 1103–1108.<br />

Yang, Y., Glenn, A. L., et al. (2008). Brain abnormalities in antisocial individuals: Implications for<br />

the law. Behav. Sci. Law 26, 65–83.<br />

Yang, Y., Raine, A., et al. (2009). Localization of deformities within the amygdala in individuals with<br />

psychopathy. Arch. Gen. Psychiatry 66, 986–994.


Intentionally left as blank


Index<br />

Adoption study, human aggression<br />

assumption and generalizability, 193–194<br />

forms of<br />

BDHI subscales, 192–193<br />

etiology, 191–192<br />

reactive and proactive aggression, 192<br />

relational, 192<br />

G x E interaction<br />

alcohol usage, 202–203<br />

family adversity and social disadvantages,<br />

199–201<br />

violent media exposure, 201–202<br />

heritability and environmental factors<br />

biological resemblance, 175<br />

effect size for, 175–184<br />

meta-analysis of, 175<br />

phenotype, 184<br />

sex-limitation, 185–186<br />

vs. twin study, 193<br />

Adrenocorticotropin (ACTH), 231–232<br />

Aggression<br />

analysis of, 3<br />

conflict situations, 1<br />

definition, 2<br />

drug-induced<br />

amphetamine, 138<br />

cocaine, 137<br />

in human, 137<br />

neural reorganization, 137<br />

in prairie vole, 136–137<br />

in Syrian hamsters, 137–138<br />

evolutionary aspects, 8<br />

in human (see Human aggression)<br />

hyperaggressive phenotypes, 2–3<br />

impulsive, 152<br />

incidence, 216<br />

in mammals<br />

mating opportunities (see Mating)<br />

A<br />

sexual dimorphism, 16–20<br />

(see also Mammals, aggression)<br />

sexual selection (see Sexual selection)<br />

mechanism, 4<br />

perinatal risk factors<br />

alcohol exposure, 230–232<br />

birth complications, 228–229<br />

drug, 232–233<br />

environmental factors, 237–238<br />

epigenetics, role of, 241–242<br />

genetic factors, 238–239<br />

G x E interactions, 239–241<br />

maternal psychological stress, 236–237<br />

preterm birth and low birth weight,<br />

229–230<br />

smoking, 233–235<br />

signaling (see Signaling aggression)<br />

in songbirds (see Songbirds)<br />

terminology, 5<br />

types of, 1–2, 152<br />

and violence, regulation of<br />

autonomic arousal, 225–226<br />

electroencephalography (EEG), 226–227<br />

hormones, 223–225<br />

neurobiological base, 218–221<br />

neurochemical signals, 221–223<br />

types of, 218<br />

in voles (see Prairie vole, selective aggression)<br />

Agonistic behavior<br />

in cephalopods, 37<br />

definition, 2<br />

game theory models, 38<br />

in prairie vole, 123–125<br />

Amphetamine (AMPH), 138<br />

Anabolic-androgenic steroids (AASs), 137<br />

Androgens, aggressive behavior, 134–135<br />

Anterior hypothalamus (AH), 96–97<br />

Antisocial personality disorder (AsPD), 152, 259<br />

Arginine vasopressin (AVP)<br />

in hamsters, 130–132<br />

285


286 Index<br />

Arginine vasopressin (AVP) (cont.)<br />

in human, 129–130<br />

in marmoset monkey, 130–132<br />

in vole, neuropeptide regulation<br />

distribution pattern of, 128–129<br />

mating-induced aggression, 129–130<br />

selective aggression, 130–131<br />

signaling and structural, 130–131<br />

Aromatase, 93<br />

Astatolapia burtoni, 61<br />

Autonomic arousal<br />

heart rate and electrodermal activity, 225–226<br />

measurement of, 225<br />

Bateman’s principles, 10–11<br />

Bed nucleus of the stria terminalis (BNST)<br />

high Fos expression, 126–127<br />

in prairie voles, 128–129<br />

social behavior regulation, 127<br />

3-hydroxysteroid dehydeogenase, 94<br />

Black-throated blue warblers, 35<br />

Borderline personality disorder (BPD), 152<br />

Catechol O-methyltransferase (COMT),<br />

240–241<br />

Cephalopods, visual signaling<br />

chromatic skin patterns, 38<br />

cuttlefish agonistic bouts, 38–39<br />

facultative nature, 37<br />

during fight, aggressive motivation, 38<br />

parallel positioning and arm posture, 38<br />

rapid adaptive polyphenism, 37<br />

squid agonistic bouts, 40–41<br />

in squid, contact pheromon, 40–43<br />

Challenge hypothesis, 86<br />

Chemical signals, 25<br />

Chickens, dominance hierarchies in, 56<br />

Child Behavior Checklist (CBCL), 184<br />

Cichlid fish, 14–15, 67–68<br />

Clozapine, dibenzodiazepine antipsychotic<br />

agent, 160<br />

Cocaine, 137<br />

Cognitive-behavioral therapy (CBT), 235<br />

Computerized axial tomography (CAT) scan,<br />

258<br />

Corticosterone (CORT), 231–232<br />

B<br />

C<br />

Crex crex, 36–37<br />

Crocuta crocuta,60<br />

Crustaceans, 57<br />

D<br />

Dendroica caerulescens,35<br />

Dendroica pensylvanica, 88<br />

Diffusion tensor imaging (DTI), 259–260<br />

Dominance relationships and hierarchies<br />

animal models<br />

chickens, 56<br />

crustaceans, 57<br />

fish, 56–57<br />

primates, 57<br />

behavioral process<br />

animal cognitive abilities and interaction<br />

process, 74<br />

jigsaw puzzle model (see Jigsaw puzzle<br />

model)<br />

social organization, 70<br />

definition<br />

behavioral measure of, 53–54<br />

individual majority, 54–55<br />

obtaining desired food, by animal pairs,<br />

54–55<br />

two hens interactions, music notation graph<br />

of, 54–55<br />

factors, animal pairs<br />

behavioral states, 62–63<br />

genetic variations, 60–62<br />

physical differences, 58–59<br />

physiological differences, 59–60<br />

in groups<br />

hierarchy formation, 64–67<br />

linear hierarchy formation, 67–70<br />

prior attributes hypothesis, 64<br />

Dopamine (DA)<br />

human aggression<br />

in BPD patients, 157–158<br />

catecholamine neurotransmitter, 157<br />

in CNS, 157<br />

nucleus accumbens (NAc), 158<br />

receptor antagonist, 158<br />

role, 157<br />

specific gene polymorphism, 157–158<br />

in male, aggressive behavior, 99<br />

perinatal risk factors, 240<br />

prairie vole, selective aggression<br />

pathways, 132


Index 287<br />

receptors, 132–133<br />

regulation of, 132–134<br />

Drug abuse and aggression<br />

amphetamine, 138<br />

cocaine, 137<br />

in human, 137<br />

neural reorganization, 137<br />

in prairie vole, 136–137<br />

in Syrian hamsters, 137–138<br />

Electroencephalogram (EEG)<br />

aggression and violence, regulation of,<br />

226–227<br />

neurocriminology, 263–264<br />

Endogenous opioid, 100<br />

Environmental Risk Longitudinal Twin study<br />

(E-risk study), 175–184<br />

Equal environment assumption (EEA), 195<br />

Estradiol (E2) blocker, 93<br />

Estrildid finches, 101–102<br />

Ethnographic Atlas Codebook, 20<br />

Event-related potentials (ERPs)<br />

neurocriminology, 264<br />

perinatal aggression, 226<br />

Evolutionarily stable strategy (ESS), 15–16,<br />

26–27<br />

Fetal alcohol spectrum disorder (FASD),<br />

230–231<br />

Fetal alcohol syndrome (FAS), 230–231, 270<br />

Field endocrinology, 89<br />

Functional neuroimaging, neurocriminology<br />

fMRI study, 260, 261<br />

PET technique, 260<br />

reduced glucose metabolism, 260–261<br />

reduced rCBF, 260<br />

schizophrenic patients, 261<br />

SPECT technique, 260<br />

Gallus gallus domesticus, 91–92<br />

Game theory, 3–4<br />

Gamma-aminobutryic acid (GABA)<br />

human aggression<br />

behavioral aggression, role in, 160<br />

E<br />

F<br />

G<br />

in CNS, 160<br />

deficiency in, 161<br />

in human aggression, 161<br />

and serotonin, 161<br />

subtypes, 161<br />

neurochemical regulation, selective<br />

aggression, 135<br />

Gene by environment (G x E) interactions<br />

human aggression<br />

alcohol use, 202–203<br />

family adversity and social disadvantage,<br />

199–201<br />

specific genes, 204–205<br />

violent media exposure, 201–202<br />

neurocriminology, 268–269<br />

perinatal aggression<br />

catechol O-methyltransferase, 240–241<br />

dopaminergic function, 240<br />

monoamine oxidase, 239–240<br />

Glutamate, 135<br />

Golden hamsters, 154–155<br />

Gonadotropin-releasing hormone (GnRH1), 61<br />

H<br />

Hawk–Dove game, 26–28<br />

Hormones<br />

aggression<br />

cortisol, 224–225<br />

oxytocin, 225<br />

testosterone, 223–224<br />

norepinephrine (NE), 158–159<br />

prairie vole, selective aggression, 134–135<br />

songbirds (see Songbirds)<br />

territoriality (see Territoriality)<br />

Human aggression<br />

in certain circumstances, 172<br />

genetic and environmental influences<br />

adoption study (see Adoption study, human<br />

aggression)<br />

forms of, 191–193<br />

G x E interaction (see Gene by environment<br />

(G x E) interactions)<br />

specific gene, genetic risk at, 203–205<br />

study design, 193<br />

twin study (see Twin study, human<br />

aggression)<br />

multidimensional behavior, 152<br />

neurochemistry of<br />

dopamine (see Dopamine)


288 Index<br />

Human aggression (cont.)<br />

GABA (see Gamma-aminobutryic acid<br />

(GABA))<br />

impulsive, 152<br />

norepinephrine (see Norepinephrine)<br />

peptides, 162<br />

serotonin/5-HT (see Serotonin (5-HT),<br />

human aggression)<br />

neurotransmitters, 152<br />

signaling<br />

natural selection, 44<br />

vocal and facial expressions, 43–44<br />

vocal frequency, 43–44<br />

social factors, 172<br />

6-Hydroxydopamine, 159<br />

5-Hydroxytryptamine (5-HT). See Serotonin<br />

(5-HT), human aggression<br />

I<br />

Impulsive aggression, serotonin<br />

in animal, 156<br />

in human, 153<br />

norepinephrine, 160<br />

Intermittent explosive disorder (IED), 152<br />

International Center for the Study of Aggression,<br />

5–6<br />

Jigsaw puzzle model<br />

in hens, 72<br />

intransitive dominance relationship, 72<br />

modification of, 73<br />

self-structuring, 71<br />

sequences, 71<br />

transitive relationship, 71<br />

Lekking birds, 14–15<br />

Linear hierarchy<br />

behavioral process<br />

cognitive abilities and interactions, 74<br />

jigsaw puzzle model (see Jigsaw puzzle<br />

model)<br />

social factors influence on<br />

in animal group, 70<br />

cichlid fish, 67–68<br />

individual attributes, 68<br />

J<br />

L<br />

isolated vs. embedded fish pairs, 69–70<br />

round-robin competition, 67–68<br />

winner, loser and bystander effects, 68–69<br />

Loligo pealei, 40–42<br />

Low resting heart rate, 264–265<br />

M<br />

Magnetic resonance imaging (MRI) scan, 258<br />

Mammals, aggression<br />

mating opportunities<br />

cost-benefit analysis, 13<br />

harem holder, paternities by, 14–15<br />

hawk-dove game, 15–16<br />

sequential assessment game, 13–14<br />

strategies, 15–16<br />

pattern, 20<br />

sexual dimorphism<br />

brain structure, 19–20<br />

harem size and body weight, 16–17<br />

limited resources, 19<br />

male-biased, 19<br />

patterns of, 16–18<br />

phylogenetic tree, 17–19<br />

sexual selection<br />

large body size, 11<br />

male domination, 11<br />

weapons, presence and absence of, 11<br />

Mating<br />

male domination, 61<br />

mammals, opportunity in<br />

cost-benefit analysis, 13<br />

harem holder, paternities by, 14–15<br />

hawk-dove game, 15–16<br />

sequential assessment game, 13–14<br />

strategies, 15–16<br />

selective aggression, 123–125<br />

squids and cuttlefish, 37<br />

VMH contribution, 93–94<br />

Melospiza georgiana,35<br />

Melospiza melodia, 31, 87–88<br />

Mesocricetus auratus, 130<br />

Methylation, 241–242<br />

Microtus ochrogaster. See Prairie vole, selective<br />

aggression<br />

Minnesota Study of Twins and Families (MFTS),<br />

175–184<br />

Minor physical anomalies (MPAs), 269<br />

Monoamine oxidase (MAO), 239–240


Index 289<br />

N<br />

Neural circuits<br />

aggression<br />

dopamine, 222–223<br />

norephinephrine, 223<br />

serotonin, 222<br />

prairie vole, selective aggression, 124, 127<br />

in selective aggression, 127<br />

songbirds<br />

in BSTm, VT-Zenk colocalization, 101–102<br />

dopaminergic mechanism, 100<br />

IEG labelling, 103<br />

neuropeptides, 100<br />

stress-related process, 100–101<br />

V1a antagonist, 102<br />

Neurobiology, aggression and violence<br />

amygdala, 219<br />

in animal and human, 218<br />

anterior cingulate cortex (ACC), 219–220<br />

hypothalamus, 221<br />

prefrontal cortex, 220–221<br />

Neurocriminology<br />

CAT scan, 258<br />

functional neuroimaging<br />

fMRI study, 260, 261<br />

PET technique, 260<br />

reduced glucose metabolism, 260–261<br />

reduced rCBF, 260<br />

schizophrenic patients, 261<br />

SPECT technique, 260<br />

genetic contribution<br />

ACE model, 267–268<br />

adoption, 267<br />

gene-environment interaction, 268–269<br />

molecular genetics, 267<br />

twins, 266–267<br />

limitations and potential, 272–273<br />

modifiable risk factor interventions<br />

home nurse visiting, 274<br />

nutritional deficits, 273–274<br />

during pregnancy, reduce smoking, 273<br />

MRI scan, 258<br />

neuropsychological testing, 262–263<br />

nongenetic risk factors<br />

perinatal, 270–271<br />

postnatal, 271–272<br />

prenatal, 269–270<br />

psychodynamic theory<br />

concepts of, 257–258<br />

early problems, 257<br />

psychophysiological evidence<br />

electroencephalogram, 263–264<br />

event-related potentials, 264<br />

low resting heart rate, 264–265<br />

skin conductance, 265–266<br />

structural neuroimaging<br />

amygdala and hippocampus, 259<br />

antisocial personality disorder, 259<br />

atrophic brains, 258–259<br />

brain ROI, size of, 258–259<br />

conduct disoder patient, 259<br />

reduced gray matter volumes/thickness, 259<br />

white matter tracts, 259–260<br />

Neuropeptides<br />

in prairie vole, selection aggression, 128–132<br />

somatostatin, 62<br />

VT and VIP, 100<br />

Neuropsychology, 262–263<br />

Neurotransmitters. See specific<br />

Neurotransmitters<br />

New World sparrows, 87–88<br />

Nicotine replacement therapy (NRT), 235<br />

Norepinephrine (NE)<br />

human aggression<br />

activity, 160<br />

clozapine, schizophrenia treatment, 160<br />

fight-or-flight response, 159<br />

impulsive aggression, role in, 160<br />

shock-induced aggression, 159<br />

stress hormone, 159<br />

impulsive aggression, serotonin, 160<br />

perinatal aggression, 232<br />

Orthoptera, 25<br />

Oxytocin<br />

and human aggression, 162<br />

perinatal aggression, 225<br />

selective aggression, neurochemical, 128<br />

Papio anubis,57<br />

Paraventricular nucleus of the hypothalamus<br />

(PVN), 97<br />

Parus major,88<br />

Passer domesticus, 89–90<br />

Peptides, in human aggression, 162<br />

O<br />

P


290 Index<br />

Perinatal aggression<br />

alcohol exposure<br />

animal models, 231–232<br />

in children, 230<br />

disorders, 230–231<br />

birth complications, 228–229<br />

drugs exposure<br />

CNS alterations, 232<br />

cocaine, 232<br />

dopamine activity, 233<br />

on fetal organogenesis, 232<br />

5-HT and norepinephrine, 232<br />

on social relationship, 233<br />

environmental con<strong>text</strong>, 237–238<br />

epigenetic, role of, 241–242<br />

genetic risk factors, 238–239<br />

G x E interactions<br />

catechol O-methyltransferase, 240–241<br />

dopaminergic function, 240<br />

monoamine oxidase, 239–240<br />

maternal psychological stress, 236–237<br />

neurocriminology, 270–271<br />

preterm birth and low birth weight<br />

CNS damage, 229<br />

neuropsychological deficits, 229<br />

prevalence of, 230<br />

sex, depends on, 229–230<br />

smoking<br />

animal models, 235<br />

in childern’s, 234<br />

effects of, 235<br />

nAChRs, 234–235<br />

nicotine exposure, 234<br />

women, in United <strong>State</strong>s, 233–234<br />

P300 event-related potentials, 264<br />

Photon emission tomography (PET), 260<br />

Postnatal, neurocriminology, 271–272<br />

Prairie vole, selective aggression<br />

drug abuse, 136–137<br />

function, 123–125<br />

inter- and intrasexual, 122<br />

mating, 123–125<br />

molecular genetics of, 136<br />

neural circuitry, 124, 127<br />

neural correlates of, 124, 125–127<br />

neurochemical regulation of<br />

dopamine, 132–134<br />

neuropeptides, 128–132<br />

neurotransmitters, 135<br />

steroid hormones, 134–135<br />

offensive and defensive types, 123<br />

pair bonding, 123<br />

resident–intruder test (RIT), 123<br />

Pregnancy, risk factors during<br />

alcohol, 270<br />

minor physical anomalies, 269<br />

obstetrical complications, 270–271<br />

in postnatal period, nutrition deficiency,<br />

271–272<br />

tobacco, 269<br />

traumatic brain injury, 272<br />

Prenatal<br />

alcohol exposure, 230–232<br />

birth complications, 228, 229<br />

drug exposure, 232–233<br />

neurocriminology, 269–270<br />

Primates, aggression in<br />

dominance hierarchies, 57<br />

male-male aggression, 19<br />

nonhuman, 221<br />

prenatal alcohol exposure, 231<br />

sexual selection, 8<br />

Prior attributes hypothesis, 64<br />

Procambarus clarkii, 57<br />

Prolactin, 154–155<br />

Psychodynamic theory<br />

concepts of, 257–258<br />

early problems, 257<br />

Psychopathy<br />

genetic basis, 268<br />

instrumental aggression, 218<br />

scores, 259–260<br />

skin conductance, 266<br />

Psychophysiology<br />

electro cortical response measures, 226–227<br />

electroencephalogram (EEG), 263–264<br />

event-related potentials, 264<br />

low resting heart rate, 264–265<br />

neurocriminology<br />

electroencephalogram, 263–264<br />

event-related potentials, 264<br />

low resting heart rate, 264–265<br />

skin conductance, 265–266<br />

skin conductance, 265–266<br />

Quinpirole D2 receptor agonist, 99–100<br />

Q


Index 291<br />

R<br />

Reactive and Proactive Aggression<br />

Questionnaire (RPQ), 184<br />

Resident–intruder test (RIT), 123<br />

Regional cerebral blood flow (rCBF), 260<br />

Sepia officinalis, 38<br />

Serotonin (5-HT), human aggression<br />

binding potential, 155–156<br />

in central nervous system, 156<br />

components, 153<br />

D,L-fenfluramine, releasing agent, 154<br />

enhancing agent, 153<br />

expression, 155<br />

failure, 156–157<br />

functions, 152–153<br />

metabolism, 153<br />

neuroimaging studies, 156<br />

postsynaptic function, 154<br />

prolactin, 154–155<br />

subtypes, 155<br />

Sex<br />

birth control methods, 8–9<br />

evolutionary explanation for, 8–9<br />

humans, 8–9<br />

Sexual selection<br />

Bateman’s principles, 10–11<br />

conflicts of interest, 9<br />

inter- and intrasexual selection, 9–10<br />

limiting resources, 10<br />

in mammals<br />

large body size, 11<br />

male domination, 11<br />

weapons, presence and absence of, 11<br />

sex role reversal, 10<br />

Signaling aggression<br />

animal models and techniques, 30<br />

cephalopods, visual display in<br />

chromatic skin patterns, 38<br />

cuttlefish agonistic bouts, 38–39<br />

facultative nature, 37<br />

during fight, aggressive motivation, 38<br />

parallel positioning and arm posture, 38<br />

rapid adaptive polyphenism, 37<br />

squid agonistic bouts, 40–41<br />

in squid, contact pheromon, 40–43<br />

chemical signals, 25<br />

S<br />

classic game theory model<br />

ESS strategies, 26–27<br />

Hawk–Dove game, 26–27<br />

ethological approach, 25–26<br />

evolutionary issues, 29–30<br />

games<br />

Hawk/Dove models, 28<br />

mutual, 27–28<br />

in humans<br />

natural selection, 44<br />

vocal and facial expressions, 43–44<br />

vocal frequency, 43–44<br />

incomplete honesty challenge, 30<br />

songbirds<br />

aggressive escalation, 33<br />

black-throated blue warblers, 35<br />

display measures, 33–34, 34<br />

mate attraction, 31<br />

musculature function, 31<br />

signaler’s advantages, 36–37<br />

singing behaviors, 32–33<br />

soft song, 36<br />

song sparrow, spectrograms of, 31–32<br />

song types and variants, 31<br />

swamp sparrows, 35<br />

territory defense, 31<br />

threat displays<br />

aggressiveness motivation, 28<br />

modulates, social interaction, 28<br />

performance signals, 28–29<br />

resource-holding potential, 28<br />

strategic signals, 29<br />

visual sign, 25<br />

Single photon emission tomography (SPECT),<br />

260<br />

Skin conductance, 265–266<br />

Sociogenomics, definition of, 60<br />

Somatostatin, 62<br />

Songbird model, 4<br />

Songbirds<br />

causality, 107<br />

con<strong>text</strong>, 84–87<br />

endocrine and neuroendocrine correlation,<br />

105–106<br />

hormonal mechanism<br />

evolution and life history, 95<br />

flocks, nonbreeding season, 91–93<br />

territoriality (see Territoriality)<br />

neural circuits, brain<br />

in BSTm, VT-Zenk colocalization, 101–102


292 Index<br />

Songbirds (cont.)<br />

dopaminergic mechanism, 100<br />

IEG labelling, 103<br />

neuropeptides, 100<br />

stress-related process, 100–101<br />

V1a antagonist, 102<br />

phentypic engineering, 107<br />

signaling<br />

aggressive escalation, 33<br />

black-throated blue warblers, 35<br />

display measures, 33–34<br />

mate attraction, 31<br />

musculature function, 31<br />

signaler’s advantages, 36–37<br />

singing behaviors, 32–33<br />

soft song, 36<br />

song sparrow, spectrograms of, 31–32<br />

song types and variants, 31<br />

swamp sparrows, 35<br />

territory defense, 31<br />

transcriptional activity<br />

arginine vasotocin (VT), 97–99<br />

Fos-ir cell count, correlations between,<br />

97–98<br />

IEG transcript, 95–96<br />

limbic areas, similarities in, 95–96<br />

paraventricular nucleus, 97–99<br />

social behavior network, 96–97<br />

white-throated sparrow<br />

genetic basis of, 104<br />

plumage polymorphism, 103–104<br />

social behavior, 103–104<br />

ZAL2 m mapping, 107–109<br />

Song sparrow. See also Songbirds<br />

aromatase mRNA expression, 93–94<br />

soft song, 33<br />

song rate, 33<br />

song-type matching, 33<br />

spectrograms of, 31–32<br />

T treatment of, 95<br />

Spizella pusilla, 87–88<br />

Steroid hormone<br />

and central DA, 135<br />

role of, 94<br />

testosterone, 134–135<br />

in voles (see Prairie vole, selective aggression)<br />

Sturnus vulgaris, 100<br />

Swamp sparrows<br />

types of calls, 35<br />

wing-waving, 35<br />

Syrian hamsters<br />

aggressive behavior in, 130<br />

drug abuse, during adolescence, 137<br />

T<br />

Taeniopygia guttata, 94<br />

Teacher’s Report Form (TRF), 184<br />

Teratogens, 230<br />

Territoriality<br />

in breeding season<br />

central perch, 88<br />

distinct species’ song, 88<br />

feather puffing, 88<br />

high-quality territory establishment, 87–88<br />

physical fights, 88<br />

simulated territorial intrusion, 88–89<br />

hormones<br />

gonadal hormone secretion, 89<br />

and plasma T, correlation between, 89<br />

plasma T profile, male breeding season,<br />

89–90<br />

in nonbreeding season<br />

aromatase activity, in brain, 93–94<br />

autumnal aggression, 93<br />

flocking behavior, 91–93<br />

gonads and ovaries regression, 91<br />

reduced plasma gonadal steroids, 91<br />

steroid hormones, role of, 94<br />

Testosterone (T)<br />

aggression and territoriality, 89–90<br />

aggressive behavior, 134–135<br />

low levels of, 85–86<br />

perinatal aggression, 223–224<br />

Traumatic brain injury (TBI), 272<br />

Twin study, human aggression<br />

additive and nonadditive, 174–175<br />

assumption and generalizability<br />

EEA, 195<br />

genetic and environmental influences,<br />

196–197<br />

psychiatric disorders, 194<br />

random mating, 195–196<br />

vs. singletons, 194–195<br />

in child, adolescent and samples, 175<br />

correlation across<br />

age groups, 187–188<br />

assessment methods, 189–191<br />

effect size for, 175–184<br />

between family members, 174


Index 293<br />

forms of, 191–193<br />

G x E interaction<br />

alcohol usage, 202–203<br />

family adversity and social disadvantages,<br />

199–201<br />

violent media exposure, 201–202<br />

indirect hostility, 192–193<br />

meta-analysis of, 175<br />

monozygotic and dizygotic, 174–175<br />

phenotype, 184<br />

sex-limited effects, 185–186<br />

shared and nonshared environmental factors,<br />

174–175<br />

vs. sibling adoption design, 193<br />

verbal hostility, 192–193<br />

in voles, selective aggression (see Arginine<br />

vasopressin (AVP))<br />

Vasotocin (VT), 97–99<br />

Violence. See Aggression<br />

W<br />

White-throated sparrow<br />

dominance hierarchies, 91–92<br />

territorial aggression in, 86–87<br />

genetic basis of, 104<br />

plumage polymorphism, 103–104<br />

social behavior, 103–104<br />

VT administration, 105–106<br />

Winner-loser models, 62–63<br />

Wood warblers, 31<br />

Vasoactive intestinal polypeptide (VIP), 100<br />

Vasopressin (VP)<br />

PVN, 97–99<br />

V<br />

Z<br />

Zebra finch, 94, 102<br />

Zonotrichia leucophrys, 87–88<br />

Zonotrichia querula, 91–92


Intentionally left as blank


0.1<br />

Human (Homo sapien)<br />

Other primate (consensus)<br />

Pig (Sus scrofa)<br />

Rat (Rattus norvegicus)<br />

Mouse (Mus musculus)<br />

Red jungle fowl (Gallus gallus)<br />

Zebra finch (Taeniopygia guttata)<br />

Ostrich (Struthio camelus)<br />

Northern pike (Esox lucius)<br />

Atlantic salmon (Salmo salar)<br />

Zebrafish (Danio rerio)<br />

Lancet (Branchiostoma belcheri)<br />

California mussel (Mytilus californianus)<br />

Pacific oyster (Crassostrea gigas)<br />

Bay scallop (Argopecten irradians)<br />

Limpet (Lottia gigantea)<br />

Longfin squid (Loligo pealeii)<br />

Abalone (Haliotis diversicolor)<br />

Bobtail squid (Euprymna scolopes)<br />

Rotifer (Adineta vaga)<br />

Human (Homo sapien) CRISP<br />

Mouse (Mus musculus) CRISP<br />

Snake (Rhinoplocephalus nigrescens) CRISP<br />

Frog (Xenopus laevis) CRISP<br />

Phylum Chordata Phylum Mollusca Phylum Rotifera<br />

Chapter 3, Figure 3.6. (See Page 43 of this volume).<br />

Chapter 5, Figure 5.5. (See Page 104 of this volume).


A B C<br />

10<br />

200<br />

b<br />

F<br />

AVP-ir/Fos-ir cells (density)<br />

Aggression (# attacks/10 min)<br />

8<br />

6<br />

4<br />

2<br />

0<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Control Partner Stranger<br />

female<br />

Naive<br />

a<br />

a<br />

CSF<br />

a<br />

a<br />

AVP AVP+<br />

V1aR Ant<br />

Paired<br />

b<br />

Stranger<br />

male<br />

D E F<br />

b<br />

*<br />

CSF V1aR Ant<br />

Naive<br />

AH<br />

OT<br />

AH<br />

Paired<br />

AVP release (% baseline)<br />

Aggression (# attacks/10 min)<br />

150<br />

100<br />

50<br />

0<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Partner<br />

*<br />

Stranger<br />

female<br />

*<br />

Control AAV-V1aR<br />

Chapter 6, Figure 6.2. (See Page 131 of this volume).

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!