chapter 15 - neuropeptide regulation of stress-induced behavior: … · 2015. 8. 25. · related...

21
CHAPTER 15 Neuropeptide Regulation of Stress-Induced Behavior: Insights from the CRF/Urocortin Family Yehezkel Sztainberg, Alon Chen Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel OUTLINE Introduction: The CRF Family of Peptides and Receptors 355 Central Administration of CRF/Ucn1 356 CNS Sites of Action of CRF/Ucn1 360 Amygdala 360 BNST 360 PVN 360 Hippocampus 360 Locus Coeruleus 361 The CRFR2/Urocortin Central System 361 Genetically Altered Mice 362 CRF-OE 362 CRF-KO 363 CRFR1-KO 363 CRFR2-KO 363 CRF-BP-OE 364 CRF-BP-KO 364 CRFR1/CRFR2 Double KO (dKO) 364 CRFR1 loxp/loxp CamKIIaCRE 364 Ucn1-KO 364 Ucn2-KO 365 Ucn3-KO 365 Ucn1/Ucn2 Double KO (dKO) 365 Ucn1/Ucn2/Ucn3 Triple KO (tKO) 365 Dysregulation of the CRF System: Mood Disorders 365 Depression 365 Anxiety 366 Concluding Remarks 366 Summary The neuropeptide corticotropin-releasing factor (CRF) and the more recently identified family members, the urocortins (Ucns), are proposed to integrate the neuroendocrine, autonomic, metabolic and behavioral responses to stressors. Chronic hyperactivation of the CRF system has been linked to stress- related emotional disorders such as anxiety and depression. The intracerebral administration of CRF or Ucn1 results in behavioral responses that are similar to those observed when animals are exposed to a stressor. The behavioral effects include increased anxiety-like behavior, decreased food consumption, increased arousal, altered locomotor activity, diminished sexual behavior, and sleep disruption. The role of the CRF/Urocortin system in modulating the behavioral responses to stressors was further demonstrated based on the behavioral phenotypes of transgenic mice that overexpress, or are deficient in, the different CRF family members. The CRFeCRFR1 system is suggested as crit- ical for initiating stress responses, while the urocortinseCRFR2 system is suggested to terminate stress responses and restore homeostasis. INTRODUCTION: THE CRF FAMILY OF PEPTIDES AND RECEPTORS Perception of physical or psychological stress by an organism is followed by a series of events that result in changes in emotional and cognitive functions, modu- lation of autonomic activities and the secretion of gluco- corticoids from the adrenal cortex. Both the activation 355 Handbook of Neuroendocrinology, DOI: 10.1016/B978-0-12-375097-6.10015-0 Copyright Ó 2012 Elsevier Inc. All rights reserved.

Upload: others

Post on 31-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

C H A P T E R

15

Neuropeptide Regulation of Stress-InducedBehavior: Insights from the CRF/Urocortin

FamilyYehezkel Sztainberg, Alon Chen

Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel

H

O U T L I N E

Introduction: The CRF Family of Peptides andReceptors 355

Central Administration of CRF/Ucn1 356

CNS Sites of Action of CRF/Ucn1 360

andboo

Amygdala 360

BNST 360 PVN 360 Hippocampus 360 Locus Coeruleus 361

The CRFR2/Urocortin Central System 361

Genetically Altered Mice 362

CRF-OE 362 CRF-KO 363 CRFR1-KO 363

355k of Neuroendocrinology, DOI: 10.1016/B978-0-12-375097-6.10015-0

CRFR2-KO 363

CRF-BP-OE 364 CRF-BP-KO 364 CRFR1/CRFR2 Double KO (dKO) 364 CRFR1loxp/loxpCamKIIaCRE 364 Ucn1-KO 364 Ucn2-KO 365 Ucn3-KO 365 Ucn1/Ucn2 Double KO (dKO) 365 Ucn1/Ucn2/Ucn3 Triple KO (tKO) 365

Dysregulation of the CRF System: Mood Disorders 365

Depression 365 Anxiety 366

Concluding Remarks 366

SummaryThe neuropeptide corticotropin-releasing factor (CRF) and themore recently identified family members, the urocortins (Ucns),are proposed to integrate the neuroendocrine, autonomic,metabolic and behavioral responses to stressors. Chronichyperactivation of the CRF system has been linked to stress-related emotional disorders such as anxiety and depression. Theintracerebral administration of CRF or Ucn1 results in behavioralresponses that are similar to those observed when animals areexposed to a stressor. The behavioral effects include increasedanxiety-like behavior, decreased food consumption, increasedarousal, altered locomotor activity, diminished sexual behavior,and sleep disruption. The role of the CRF/Urocortin system inmodulating the behavioral responses to stressors was furtherdemonstrated based on the behavioral phenotypes of transgenicmice that overexpress, or are deficient in, the different CRF

family members. The CRFeCRFR1 system is suggested as crit-ical for initiating stress responses, while the urocortinseCRFR2system is suggested to terminate stress responses and restorehomeostasis.

INTRODUCTION: THE CRF FAMILY OFPEPTIDES AND RECEPTORS

Perception of physical or psychological stress by anorganism is followed by a series of events that resultin changes in emotional and cognitive functions, modu-lation of autonomic activities and the secretion of gluco-corticoids from the adrenal cortex. Both the activation

Copyright � 2012 Elsevier Inc. All rights reserved.

Page 2: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY356

and the termination of the behavioral, autonomic andadrenocortical stress responses are crucial for adaptationand survival. The neuropeptide corticotropin-releasingfactor (CRF), expressed and secreted from the paraven-tricular nucleus (PVN) of the hypothalamus, representsthe final common pathway for the integration of theneuroendocrine stress response in the brain.1 CRF and,to a lesser extent, arginine vasopressin (AVP) play animportant and well-established role in the regulationof the hypothalamusepituitaryeadrenal (HPA) axisunder basal and stress conditions.2,3 In addition to thePVN, CRF mRNA and peptide have been found inseveral extrahypothalamic brain nuclei, including thecerebral cortex, amygdala, bed nucleus of striaterminalis (BNST), and hippocampus, suggesting that,in addition to its hypophysiotropic action, CRF actsto integrate the neuroendocrine, behavioral, autonomicand metabolic responses to stressors.4e6 Dysregulationof the stress response can have severe psychologicaland physiological consequences,7 and chronic hyperac-tivation of the CRF system has been linked to stress-related emotional disorders such as anxiety, anorexianervosa and melancholic depression.8e10 In additionto CRF, the mammalian CRF-peptide family containsurocortin (Ucn) 1,11 Ucn212,13 and Ucn3.13,14 CRF andits related peptides signal through the activationof two high-affinity G-protein coupled receptors,CRF receptor type 1 (CRFR1)15e17 and CRFR2.18e22

Both CRFR1 and CRFR2 belong to the class B1subfamily of seven-transmembrane domain receptorsthat signal by coupling to G proteins. CRFR1 hasone known functional splice variant (a) expressed inthe central nervous system and the periphery.23,24

The CRFR1 mRNA is widely expressed in mammalianbrain and pituitary, with high levels in the anteriorpituitary, cerebral cortex, cerebellum, globus pallidus,amygdala, hippocampus and olfactory bulb.25 Inthe periphery, CRFR1 is expressed in testes, ovary,skin and spleen. CRFR2 has three functionalmembrane splice variants in the human (a, b and g)and two in the rodent (a and b).18e22,24,26 CRFR type2a (CRFR2a) is the major splice variant in the rodentbrain.27 It is expressed in a more discrete patternthan CRFR1, with highest densities in the lateralseptum, BNST, ventromedial hypothalamic nucleus(VMH), olfactory bulb and mesencephalic dorsal raphenucleus (DRN),25 whereas CRFR type 2b (CRFR2b) isexpressed primarily in peripheral tissues and thechoroid plexus of the brain.18 CRFR1 and CRFR2 differpharmacologically: CRF has relatively lower affinityfor CRFR2 compared to its affinity for CRFR1, Ucn1has equal affinities for both receptors, and Ucn2 andUcn3 appear to be selective for CRFR2.11,12,14 In addi-tion to both CRF receptors, CRF and Ucn1 can bindto the CRF-binding protein (CRF-BP) suggested to

III. HORMONES, BRAIN FUN

function as an endogenous “buffer,” thus addinga further level of complexity to the control of theCRF-related ligands’ actions.28e30

CENTRAL ADMINISTRATION OFCRF/Ucn1

The intracerebroventricular administration of CRF orUcn1 results in behavioral responses that are similar tothose observed when animals are exposed to a stressor(Fig. 15.1). The behavioral effects include increasedanxiety-like behavior, decreased food consumption,increased arousal, altered locomotor activity, dimin-ished sexual behavior, and sleep disruption (for detailedreviews, see references 5,31e35).

One of the earliest and best described effects ofcentral CRF administration is increased arousal andaltered locomotor activity which is dependent on thetesting conditions. In non-stressed rats tested ina familiar environment, i.c.v. injection of CRF elicitsa dose-dependent increase in general behavioralactivation that includes elevated walking, excessivegrooming and rearing.4,36e38 This effect is apparentlymediated by central pathways and independent ofpituitaryeadrenal activation, since CRF increases loco-motor activity in both naı̈ve and hypophysectomizedanimals,39 and the effect is not blocked by dexametha-sone administration.40 The potent stimulatory effect ofCRF in behavioral activation is evolutionary conserved,as has also been described in several vertebratespecies.35 CRF increases swimming and walking inamphibians41e44 and fish,45,46 stepping in birds,47,48

and general motor activity in pigs,49e52 mice36,53,54 andprimates.55,56

In contrast to the behavioral activating effect of CRFin animals under low arousal conditions, when animalsare exposed to a novel stressful environment such as anopen field or an elevated plus maze, CRF administrationeffects changes to behavioral inhibition. Following i.c.v.injection of CRF, rodents show decreased locomotion,rearing and inner square crossings in an open fieldtest,4,57 a reduced percentage of time spent on theopen arms of the elevated plus maze,58,59 and decreasedinvestigatory behavior in a multi-compartment chambertest.53,60,61 In addition to the behavioral suppressionobserved in these exploration tests, the anxiogenic-likeprofile of central CRF administration includes increasedstress-induced freezing behavior,62,63 reduced socialinteraction,64 potentiated acoustic startle response65,66

and increased defensive burying.67 Other stress-relatedbehaviors induced by i.c.v. CRF administration aredecreased food consumption,63,68e71 inhibition ofmaternal behavior,72 decreased sexual behavior,73 andthe induction of place and taste aversion.74,75 The i.c.v.

CTION AND BEHAVIOR

Page 3: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

Locomotion (familiar environment) [ 86, 87 ]

Locomotion (novel environment) [ 88 ]

Startle response [ 104 ]

Anxiety [ 105, 106 ]

Depression [ 94 ]

Social interaction [ 105 ]

Food consumption [ 108 ]

Anxiety [ 121 ]

[ 121 ]FreezingAssociative learning [ 117 ]

Anxiety [ 58, 59, 62-67, 76-78 ]

Startle response [ 65, 66 ]

Behavioral activation (familiar environment) [ 4, 36-56 ]

Behavioral inhibition (novel environment) [ 4, 53, 57, 60, 61 ]

Food consumption [ 63, 68-71 ]

Social interaction [ 64 ]

Sexual behavior [ 73 ]

Maternal behavior [ 72 ]

Hipp

BNST

CeA

BLA

PVN

LC

Anxiety [ 91 ]

Grooming [ 92 ]

Food consumption [ 92 ]

Social interaction [ 89, 90 ]

Grooming [ 111 ]

Locomotion (low dose) [ 111 ]

Locomotion (high dose) [ 111 ]

Food consumption [ 112 ]

cc

LV

Arousal [ 128 ]

Anxiety [ 128 ]

Memory [ 131 ]

FIGURE 15.1 A schematic representation of a sagittal view of the rodent brain, showing the behavioral consequences of site-specific CRF/Urocortin administration. LV, lateral ventricle; CC, corpus callosum; Hipp, hippocampus; BNST, bed nucleus of stria terminalis; CeA, centralamygdala; BLA, basolateral amygdala; PVN, paraventricular nucleus of the hypothalamus; LC, locus coeruleus.

BOX 15.1

H OW DO WE KNOW TH E G E N E T I C A P P R O A CH F O RI N T R A C E R E B R OV E N T R I C U L A R D E L I V E R Y ?

Pharmacological administration of synthetic peptides

or secreted recombinant proteins into the brain ventricles

is a commonmethod used by neuroscientists for exploring

physiological and behavioral functions of novel or known

gene products. Cerebroventricular (rather than systemic),

administration of these proteins is required in order to

bypass the bloodebrain barrier (BBB) and allow the non-

selective transport of peptides or proteins from the

periphery into the central nervous system (CNS).

Current approaches for delivery of peptides or secreted

proteins to the CNS include, for short-term acute admin-

istration, a stereotaxic injection into the ventricular space,

known as intracerebroventricular (i.c.v. administration);

and for chronic delivery, an i.c.v. microinjection pump.

These methods rely heavily on the solubility and half-life

of the injected substance, require chemical or in vitro

synthesis of the administered ligand, and may involve

different purification procedures. The i.c.v. administration

is difficult to use in studies requiring repeated or pro-

longed administration of the ligand, since the microin-

jection pump procedure depends on the ligand stability

and capacity of the reservoir, and complex surgical

procedures are required for installation and manipulation

of the pump. Furthermore, the current procedures require

(Continued)

CENTRAL ADMINISTRATION OF CRF/Ucn1 357

III. HORMONES, BRAIN FUNCTION AND BEHAVIOR

Page 4: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

BOX 15.1 (cont’d)

extensive handling of the experimental animals, which

may create behavioral or physiological disturbances.

We have recently described a genetic approach for the

delivery of secreted peptides or proteins into the cere-

brospinal fluid (CSF) using a choroid plexus-specific and

lentiviral-based genetic system.1 The choroid plexus

plays a critical role in the barrier mechanism regulating

the exchange of molecules between the brain’s internal

milieu and the periphery. This bloodeCSF barrier is

composed of epithelial cells with apical tight junctions

that restrict intercellular passage of molecules from

fenestrated blood vessels. The CSF circulatory system’s

function is to provide micronutrients, neurotrophins,

hormones, neuropeptides and growth factors exten-

sively to neuronal networks. Therefore, neuro-

modulators directed to CSF can modify and adapt

a variety of behavioral, neuroendocrine and immuno-

logic processes.

Using a choroid plexus-specific promoter, we estab-

lished a lentiviral-based system, which offers inducible

and reversible delivery of a gene product into the CSF.

The system is composed of two complementary lenti-

viral vectors. The ”Effector” construct consists of

a choroid plexus-specific promoter that drives the

expression of reverse tetracycline transactivator (rtTA)

protein and the reporter green fluorescent protein (GFP)

(see A, upper panel). The “Target” construct includes

the tetracycline-responsive element (TRE) DNA

sequence, upstream to the nucleotide coding sequence

of the requested gene of interest, followed by the

reporter red florescent protein (RFP) (see A, lower

panel). Transcription initiation of the gene of interest

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY358

III. HORMONES, BRAIN FUNCTION AND BEHAVIOR

Page 5: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

BOX 15.1 (cont’d)

and the RFP is mediated only in the presence of the

inducer, doxycycline (Dox). A mixture of the two lentivi-

ruses is injected intracerebroventricularly, and the deliv-

ered genes are incorporated into the DNA of the choroid

plexus cells. Initiation of transcription, limited to the

choroid plexus cells by the choroid plexus-specific

promoter, is induced by administrating Dox-containing

drinking water, and results in secretion of the final pro-

cessed gene product into the CSF (see B). Dox is the

inducer of choice for our purposes, as it has been

demonstrated to cross the bloodebrain barrier. The

functionality of this system was demonstrated using the

overexpression of the two established neuropeptides,

corticotropin-releasing factor and gonadotropin-releasing

hormone, modulating anxiety-like behavior and estrous

cycle, respectively.1

Reference

1. Regev L, Ezrielev E, Gershon E, et al. Genetic approach forintracerebroventricular delivery. Proc Natl Acad Sci. USA.

2010;107:4424e4429.

CENTRAL ADMINISTRATION OF CRF/Ucn1 359

administration of Ucn1 results in a similar behavioralprofile to that of CRF, including increased groomingand motor activity in a familiar environment,38 andincreased anxiety-like behavior in different paradigms,including the open field test, the elevated plus mazetest, the lightedark transfer test, the defensive with-drawal test and the conflict test.76,77

Recently, a new genetic approach has been applied forthe inducible and reversible secretion of CRF into thecerebrospinal fluid (CSF).78 The choroid plexus, a groupof modified ependymal cells in the ventricles of the brainthat produce the CSF, was genetically targeted by i.c.v.

III. HORMONES, BRAIN FUN

injection of a lentiviral vector expressing CRF. The initi-ation of CRF transcription, limited to the choroid plexuscells by a choroid plexus-specific promoter, was inducedby administrating the animals with doxycycline in thedrinking water, which results in the secretion of theCRF peptide into the CSF. Mice conditionally overex-pressing CRF at the choroid plexus showed an increasein anxiety-like behavior in the lightedark transfer,open field and elevated plus maze tests relative to thecontrol group.78 These results further support the prin-cipal role of CRF in the regulation of anxiety, previouslydemonstrated by pharmacological studies.

CTION AND BEHAVIOR

Page 6: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY360

CNS SITES OF ACTION OF CRF/UCN1

Amygdala

The amygdala is an important mediator of fear andanxiety.79e81 In view of the relative high concentrationof CRF in the central nucleus82,83 and the dense concen-tration of CRF receptors in the basal nucleus,25,84,85 theamygdala has been largely investigated for its role inthe effects of CRF on emotion-related behavior. In rats,CRF injected into the central amygdala (CeA) inducesan increase in locomotor activity in a familiar environ-ment,86,87 but a decrease in locomotion in a novel openfield88 e results that resemble those obtained by i.c.v.administration of CRF. CRF or Ucn1 injected into thebasolateral amygdala (BLA) reduced social interactiontime,89,90 led to a persistent increase in anxiety-likebehavior,91 decreased feeding, and increased groom-ing,92 suggesting a specific role for the amygdalar CRFsystem in anxiety-like behavior. Two research groupshave recently used a lentiviral-based system forchronic amygdala-specific overexpression of CRF.Keen-Rhinehart and colleagues found that female ratsoverexpressing CRF in the amygdala for a period of 2weeks showed an increase in acoustic startle responseand attenuated sexual motivation,93 both results indi-cating an increase in anxiety levels. Regev and colleaguesfound that male mice overexpressing CRF in the amyg-dala for a period of 4 months showed reduced levels ofanxiety-like behavior in response to acute stressful stim-ulation in the open field and lightedark transfer tests.94

The discrepancy between the studies may be attributedto the difference in CRF overexpression duration, genderdifferences, species differences and behavioral tests.

Injections of a-hCRF9e41, a synthetic competitiveantagonist of CRF receptor, into the CeA reverses thesocial stress-induced decrease in time spent in the openarms of the EPM95 and attenuates stress-induced freezingbehavior.96 Intra-CeA infusions of antisense oligodeoxy-nucleotides against CRFR1 reduce anxiety-like behaviorin socially defeated rats.97 Intra-BLA administration ofastressin B, a CRFR1/2 antagonist, reverses the anxio-genic effects ofUcn1,98whereas intra-BLAadministrationof the CRFR1-specific antagonist antalarmin preventsdefeat-induced defensive behavior in mice.99 Finally,a recent study has demonstrated that the decrease inanxiety-like behavior in mice reared in an enriched envi-ronment is associated with very low levels of CRFR1 inthe BLA. Knockdown of CRFR1 in the BLA mimickedthe anxiolytic effect of environmental enrichment.100

BNST

Heavily innervated by the amygdala, and projecting toseveral regions involved in fear and anxiety responses,

III. HORMONES, BRAIN FUN

the BNST is considered an additional brain site thatpotentially mediates the behavioral effects of CRF/Ucnadministration.101 Expressing CRF83 and both CRF1and CRF2 receptors,25,85,102 the BNST has been suggestedto play a special role in longer-duration anxiety-likeresponses, in contrast to the CeA implicated in shorter-duration fear responses.103 Microinfusions of CRF intothe BNST increase startle amplitude in a dose-dependentmanner. In the same study, intra-BNST injection ofa-hCRF9e41 attenuated the effect of CRF injected i.c.v. inthe startle response test.104 Moreover, “priming” withsubanxiogenic doses of Ucn1 in the BNSTelicits a persis-tent anxiogenic profile in the social interaction test andthe elevated plus maze test, an effect blocked by priorlocal injection of CRFR1 antagonists.105,106 In anotherstudy, post-training administration of CRF in the BNSTenhanced retention in an inhibitory avoidance task,107

suggesting a role for the CRF system in the BNST inmemory formation processes for affective experience.The BNST is also involved in CRF-induced anorexia, asCRF administrated directly into the BNST, but not theCeA or the LC, induces a marked inhibition of feeding.108

Finally, chronic overexpression of CRF in the dorsolateralsubdivision of the BNST, but not in the CeA, increasesdepressive-like behavior without affecting anxietylevels.94

PVN

The paraventricular nucleus of the hypothalamus(PVN) contains a large number of CRF-immunopositivecell bodies and fibers,83,109 as well as moderate concen-trations of CRF receptors.25,85,102 In addition to its rolein activating the HPA axis by its projections to themedian eminence, the CRF neurons in the PVN maybe involved in CRF-induced behavioral inhibitionthrough its projections to brainstem nuclei.110 CRFadministration directly into the PVN induces a dose-dependent increase in grooming, and an increase inlocomotion at lower doses but a suppression of locomo-tion at higher doses of CRF.111 In addition, intra-PVNinjections of CRF decrease food intake,112 whereasa-hCRF9e41 injections in the PVN potentiate feedinginduced by NPY injected in the same locus.113 A recentstudy has demonstrated that lentiviral-mediated knock-down of CRF in the PVN attenuates social avoidance inchronic social-defeated mice.114

Hippocampus

The hippocampus contains scattered CRF-stainedinterneurons,83 and moderate concentrations of CRFreceptors.25,85,102 The dorsal hippocampus has been sug-gested to be involved in context-dependent115 and tone-dependent fear conditioning.116 Injections of CRF in the

CTION AND BEHAVIOR

Page 7: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

THE CRFR2/UROCORTIN CENTRAL SYSTEM 361

dorsal hippocampus before or after training enhance bothcontext-dependent and tone-dependent fear acquisition.This effect seems to be CRFR1-mediated, as it is pre-vented by local injections of astressin B, a non-selectiveCRF receptor antagonist, but not by antisauvagine-30,a specific CRFR2 antagonist.117 Furthermore, recoveryfrom stress-induced increased anxiety and impairmentof context-dependent fear conditioning requires hippo-campal CRFR1, as the recovery is prevented by intra-hippocampal injections of DMP696, a highly selectiveCRFR1 antagonist, but not by antisauvagine-30.118

Finally, the ventral hippocampus has been implicated inthe modulation of anxiety-related behaviors.119,120 Micro-infusions of CRF into the ventral hippocampus increaseanxiety-like behavior in the EPM test, as well as stress-induced freezing.121

Locus Coeruleus

Both physiologic and psychogenic stressors activatethe noradrenergic cells in the locus coeruleus(LC).122e124 These cells project to the forebrain and thespinal cord,125e127 and induce a state of arousal requiredfor the correct behavioral and physiological response tostress. Several studies suggest that CRF plays an impor-tant role in this effect. CRF infused into the LC increasesnon-ambulatory motor activity (shifting in body posi-tion), whereas locomotor activity is not altered. Inaddition, CRF infused into the LC produces a dose-dependent decrease in floating behavior in a modifiedPorsolt swim test, whereas struggling behavior is notaffected. Both results suggest an increased arousal andagitation effect of CRF in the LC.128 In the same study,CRF was injected into the LC decreased inner and outercrossings, free and wall rearing, as well as explorationoutside of a darkened compartment in an open fieldtest, indicating an anxiogenic effect.128 Furthermore,injection of a-hCRF9e41 into the LC reduces the levelsof shock-induced freezing behavior129 and immobiliza-tion stress-induced defensive withdrawal.130 Finally,CRF injected directly into the LC improves memoryretention in a passive-avoidance task,131 suggestinginvolvement of the LC in the learning and memory-facil-itating effects of CRF.

THE CRFR2/UROCORTIN CENTRALSYSTEM

The CRF-peptide family includes, in addition to CRF,the three urocortin (Ucn) peptides (Ucn1, Ucn2 andUcn3) that bind and activate the CRF receptor type 2(CRFR2) with high affinity.11e14,132e134 CRF has a rela-tively lower affinity for CRFR2 than for CRFR1, Ucn1

III. HORMONES, BRAIN FUN

has equal affinities for both, and Ucns 2 and 3 appearto be selective for CRFR2.11e14 These receptors aredistributed differently throughout the brain, whileCRFR1 is widely expressed, CRFR2 expression is morelocalized to selected stress-related brain nuclei, such asthe medial amygdala, the bed nucleus of the stria termi-nalis (BNST), the lateral septum (LS) and the dorsalraphe nucleus (DRN).25,102

While the role of CRF and CRFR1 in the activation ofthe HPA axis and the regulation of emotional and cogni-tive functions following exposure to stressful challengeare well established,9,135e138 the role of CRFR2 is stillcontroversial. Studies exploring the role of CRFR2 usingknockout mice models, antisense oligonucleotide, orantagonist administration are less clear, and show con-flicting behavioral results. Central administration ofUcn2 or Ucn3,139e142 CRFR2 antisense oligonucleo-tide,143,144 and antagonist studies117,145,146 have showncontrasting, dose-dependent and localization-dependentresults. These results suggest a central role for CRFR2that may vary between different brain nuclei and underdifferent stress conditions. Several pharmacologicalstudies suggest anxiolytic-like effects of CRFR2activation. Intracerebroventricular administration ofUcn2139,147,148 or Ucn3,140,149 the endogenous ligandsfor CRFR2, exerts an anxiolytic-like phenotype in thetested rodents. In contrast, others have shown a possibleanxiogenic role for CRFR2 activation, specifically in theLS,117,141,150 the DRN,151 and when using CRFR2-specificantisense oligonucleotides or CRFR2 agonists.146,152e155

Furthermore, CRFR2 activity in the septum was shownto vary under different stress conditions, being lessinvolved under low stress conditions, and significantlyincreasing anxiety-like behavior under high stressconditions.140

A growing body of evidence suggests that Ucn1 andUcn2 may influence stress-related physiology andbehavior by modulating the DRN serotonergicsystem.156 Ucn2 i.c.v. administration caused an increasein c-Fos immunostaining in topographically organizedsubpopulations of serotonergic neurons in the DRN,specifically within the dorsal part of themid-rostrocaudalDRN (DRD) and the caudal part of the DRN(DRC).157,158 Moreover, while injection of Ucn2 to theDRC leads to increased 5-HT release in the BLA159 andpotentiation of conditioned fear, as well as escape defi-cits in a model of learned helplessness,151 injection ofthe CRFR2 antagonist antisauvagine-30, and not ofCRFR1 antagonists, showed anxiolytic effects, includingreversal of the potentiation of conditioned fear and theescape deficits following exposure to inescapablestress.146,151,154 Thus, these behaviors indicating height-ened anxiety in response to uncontrollable stress seemto be mediated by CRFR2 receptors on serotonergicneurons in the DRC.160

CTION AND BEHAVIOR

Page 8: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY362

GENETICALLY ALTERED MICE

Amajor approach for the study of the involvement ofthe CRF/urocortin system in the regulation of stress-induced behavior has been the use of transgenic micemodels, overexpressing or knocking out the differentfamily members. To date, all genes of the CRF/urocortinfamily have been successfully targeted, generating defi-cient animal models that have been crucial for theunderstanding of the role of the CRF/Urocortin familyof peptides and receptors in mediating stress-relatedbehaviors.

In the case of developmental transgenic mice model,it should be remarked that conclusions regarding their

TABLE 15.1 Summary of Behavioral and Endocrinological Consequ

Genotype Phenotype

CRF OE (Stenzel-Poore et al.) High plasma levels of ACTH and corbehavior, decrease in sexual behavioattention.

CRF OE (Dirks et al.) Reduced acoustic startle reactivity, imbehavioral adaptation to a novel env

CRF OE (CNS restricted) Stress-induced hypersecretion of ACimmobility in the forced swim test a

CRF KO Normal basal and stress-induced anxstartle response and learning.

CRFR1 KO Disrupted HPA axis activation in resimpaired spatial recognition memory

CRFR2 KO (Bale et al.) Increased ACTH and corticosteronerelease. Increased anxiety and depre

CRFR2 KO (Coste et al.) Increased ACTH and corticosteronerelease. No differences in anxiety-lik

CRFR2 KO (Kishimoto et al.) Increased anxiety and depression-lik

CRF-BP OE Increased locomotion and rearing inin the EPM test.

CRF-BP KO Increased anxiety-like behavior. Defi

CRFR1/CRFR2 dKO Impaired stress-induced HPA systemEPM test (only in females).

CRFR1loxp/loxpCamKIIaCRE Reduced anxiety-like behavior. Norm

Ucn1 KO (Vetter et al.) Increased anxiety-like behavior.

Ucn1 KO (Wang et al.) No effect in anxiety-like behavior.

Ucn2 KO Increased nocturnal ACTH and cortibehavior (only in females). No effect

Ucn3 KO Normal HPA axis regulation, anxietyIncreased social recognition memory

Ucn1/Ucn2 dKO Increased stress-induced corticosterolike behavior.

Ucn1/Ucn2/Ucn3 tKO Increased anxiety-like behaviors 24 hinduced acoustic startle-response.

III. HORMONES, BRAIN FUN

physiology and behavior must be drawn cautiously,due to compensatory changes in other related or non-related genes. In addition, in the case of gene overex-pression the expression of the gene of interest is oftendriven under the control of a general promoter, resultingin overexpression of the gene of interest in non-endoge-nous brain regions and peripheral organs (for detailedreviews on CRF family mutant mice, see references161e163) (Table 15.1).

CRF-OE

To study the effects of chronic exposure to CRF, trans-genic mice overexpressing CRF under the control of

ences of CRF System Gene Targeting

Reference(s)

ticosterone. Increased anxiety and depression-liker (only in females) and deficits in learning and

164e69

paired pre-pulse inhibition, and abnormalironment.

170e172

TH and corticosterone (only in males). Reducednd tail suspension test.

173

iety-like behavior, locomotor activity, exploration, 175e176

ponse to stress. Reduced anxiety-like behavior,, and deficiencies in nurturing behavior (females).

177e182

response to stress and early termination of ACTHssion-like behavior.

183, 186

response to stress and early termination of ACTHe behavior.

184

e behavior. 185, 187

the open field test, and increased total arm entries 188

cits in maternal aggression. 190,191

activation. Reduced anxiety-like behavior in the 192, 193

al basal plasma ACTH and corticosterone levels. 196

197

198

costerone levels, and reduced depressive-likein anxiety-like behavior.

199

-like behavior and depressive-like behavior..

201

ne response (only in males). Decreased anxiety- 156

following stress exposure. Increased stress- 202

CTION AND BEHAVIOR

Page 9: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

GENETICALLY ALTERED MICE 363

metallothionein (mMT1) promoter have been devel-oped.164 CRF-overexpressing mice (CRF-OE) exhibithigh plasma levels of ACTH and corticosterone, anddevelop physical changes typical of Cushing’ssyndrome. At the behavioral level these mutants displaya phenotype characteristic of increased anxiety anddepression levels, such as decreased baseline andstress-induced locomotor activity in a novel environ-ment, decreased time spent in the open arms of anelevated plus maze (EPM),165 decreased time spent inthe light area of a lightedark transfer box, reducedtime spent rearing in home cages, and increased immo-bility in a forced swim test.166 In terms of sexualbehavior, CRF-OE female mice display a profounddecrease in sexual receptivity, whereas the sexualperformance of CRF-OE males remains intact.167 CRF-OE mice also show significant deficits in learning,168

and reduced attention.169

An additional CRF-OE transgenic mouse was gener-ated using the murine Thy-1.2 promoter.170 In contrastto the mMT1 CRF-OE mice, the overexpression inThy1 CRF-OE mice starts 4e8 days after birth and isrestricted to the central nervous system and thespinal cord.170 These mice display reduced acousticstartle reactivity, impaired pre-pulse inhibition,171

and abnormal behavioral adaptation to a novelenvironment.172

Finally, three different mouse models of conditionalCRF overexpression have been generated using the nes-tin (Nes) promoter (CNS-restricted overexpression),CamKIIa promoter (forebrain-restricted overexpres-sion) and Dlx5/6 promoter (forebrain GABAergicneurons-restricted overexpression).173 CRF overexpres-sion in the entire central nervous system, but not inspecific forebrain regions, resulted in stress-inducedhypersecretion of ACTH and corticosterone in malesbut not in females, and reduced immobility in the forcedswim test and tail suspension test in both males andfemales.173

CRF-KO

The development of CRF knockout (CRF-KO) micehas been important in addressing the physiologic andpathologic roles of CRF.3,174 However, in terms of behav-ioral effects, CRF-KO mice exhibit normal basal andstress-induced anxiety-like behavior. In addition, base-line locomotor activity, exploration, startle responseand learning appear to be unaffected.175,176 CRF receptorantagonists have an anxiolytic effect in CRF-KO mice,suggesting that whereas CRFR1 activation is crucialto induce anxiety, CRF itself is not.176 Therefore, it hasbeen proposed that another CRF-related peptide,such as Ucn1, which has high affinity to CRFR1, may

III. HORMONES, BRAIN FUN

contribute to the observed stress-induced behavior inthese animals.175,176

CRFR1-KO

In order to elucidate the relative contribution of theCRF receptor type 1 to stress-induced physiology andbehavior, two independent groups generated a mouseline null for the CRFR1 gene.177,178 Mice deficient forCRFR1 (CRFR1-KO) display a disrupted HPA axis acti-vation in response to stress, causing glucocorticoid defi-ciency.177,178 However, basal plasma ACTH was foundto be similar to that in wild-type controls, probablydue to the action of AVP.179 Behaviorally, CRFR1-KOmice display reduced anxiety-like behavior in thelightedark transfer test and the EPM test.177,178,180 Inaddition, it has been reported that CRFR1-KO mice areimpaired in spatial recognition memory in a Y-mazeparadigm.180 In terms of social behavior, male CRFR1-KO mice show intact isolation-induced inter-maleaggression,181 whereas female CRFR1-KO mice exhibitdeficiencies in nurturing behavior, such as decreasedtime spent nursing, licking and grooming the pups.182

It should be remarked that, in both studies,181,182

CRFR1-KO mice with C57BL/6 X 129 background,have been crossed into an outbred hsd:ICR strain selec-tively bred to exhibit high levels of maternal aggression,to improve aggressive performance.

CRFR2-KO

In contrast to the clear and consistent phenotype ofthe two CRFR1-KO mouse lines, the three indepen-dently generated CRFR2-KO mouse lines183e185 presentseveral inconsistencies in their neuroendocrine andbehavioral profiles. Coste and colleagues184 found anincreased ACTH and corticosterone response to stressand an early termination of ACTH release in CRFR2-KO mice. However, no differences in anxiety-likebehavior184 were found between CRFR2-KO and theirwild-type littermates. Bale et al.183 also showed increasedACTH and corticosterone response to stress and an earlytermination of ACTH release in their CRFR2-KO mouseline. However in contrast to the Coste et al.184 findings,Bale and colleagues183 found that CRFR2-KO micedisplay a significant increase in anxiety-like behaviorin the EPM and the open field tests, but not in thelightedark transfer test.183 Finally, Kishimoto et al.185

showed that only male CRFR2-KO mice are moreanxious in the EPM test and the lightedark transfertest; however, in the open field test, CRFR2-KO micespent more time in the center of the arena.185 Thesedifferences could be a result of the different geneticbackgrounds of the mouse lines. Both the Bale et al.183

CTION AND BEHAVIOR

Page 10: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY364

and the Kishimoto et al.185 CRFR2-KO mouse linesexhibited a depression-like phenotype in the forcedswim test and the tail suspension test.186,187

CRF-BP-OE

Two different mouse models of CRF binding proteinoverexpression (CRF-BP-OE) have been independentlygenerated.188,189 The CRF-BP-OE line created by Lovejoyand colleagues expresses the transgene not only in thebrain and pituitary, but also ectopically in peripheraltissues.189 However, this mouse line has not yet beenbehaviorally phenotyped. The CRF-BP-OE line createdby Burrows et al. expresses the transgene under thecontrol of the pituitary glycoprotein hormone a-subunit(a-GSU) promoter, resulting in specific overexpressionof CRF-BP in the anterior pituitary gland.188 Thesemice exhibit increased locomotion and rearing in theopen field test, increased total arm entries in the EPM,and a tendency to spend more time in the open armsof the EPM.188

CRF-BP-KO

Mice deficient for CRF-BP (CRF-BP-KO) displayincreased anxiogenic-like behavior when tested in theEPMand the lightedark transfer test, and a trend towardincreased anxiety-like behavior in the open field test.190

These results suggest an increase in “free” CRF or Ucn1levels, leading to an anxiogenic phenotype; however,this hypothesis has not yet been examined in thesemutants. The same line of CRF-BP-KO mice has beencrossed into an outbred hsd:ICR strain selectively bredto exhibit high levels of maternal aggression, to investi-gate aggressive behavior.191 In this study, CRF-BP-KOmice exhibited significant deficits inmaternal aggression(offspring protection) relative to wild-type mice. FemaleCRF-BP-KO mice were also tested in the forced swimtest, where no differences were found between thegroups. In the lightedark transfer test, female CRF-BP-KOmice exhibited higher levels of anxiety-like behavior.For males, no significant differences in lightedark trans-fer, swim test and isolation-induced residenteintrudermale aggression were found between the groups.However, increased anxiety-like behavior in mutantmales was detected in the approach to a novel objectplaced in the center of an open field arena.191

CRFR1/CRFR2 Double KO (dKO)

To further elucidate the role of both CRF receptors inthe activation and modulation of the neuroendocrinestress response and anxiety-like behavior, two mousemodels deficient of both CRFR1 and CRFR2 (CRFR1/CRFR2double knockout; dKO) have been independently

III. HORMONES, BRAIN FUN

generated.192,193 Bothmouse lines display impairment instress-induced HPA system activation. In terms ofanxiety-like behavior, double-mutant mice are sexuallydichotomous. Whereas no difference was found inanxiety-like behavior between male double-mutantsand wild-type mice, female CRFR1/CRFR2 dKO micedisplay reduced anxiety-like behavior in the EPM test.However, no difference was found in the number ofcenter visits in the open field test.192 The behavioralphenotype of the Preil et al. mouse model193 has not yetbeen reported.

CRFR1loxp/loxpCamKIIaCRE

As glucocorticoids are known to be involved in themodulation of fear and anxiety-like behavior,194,195 theanxiolytic-like profile observed in the developmentalCRFR1 KO mice may result either from central CRFR1deficiency, or from the low circulating levels of gluco-corticoids. In order to differentiate the CRF/CRFR1pathways that control stress-induced behavior fromthose regulating the HPA axis, a conditional CRFR1knockout mouse line was generated using the Cre/loxPsystem. In these mutants the CRFR1 expression, drivenby the calcium calmodulin-kinase IIa (CamKIIa)promoter, is postnatally ablated in the anterior fore-brain and limbic system including the hippocampus,amygdala and neocortex, but remains intact in the pitu-itary gland.196 The CRFR1 conditional mutants(CRFR1loxp/loxpCamKIIaCRE) display reduced anxiety-like behavior levels in the EPM and the lightedarktransfer tests. In contrast, the basal plasma ACTH andcorticosterone levels are similar to those in wild-typemice. Moreover, the hormone levels in conditionalmutants remain significantly elevated 30 and 90 minfollowing restraint stress, indicating that CRFR1 condi-tional mutants are hypersensitive to stress, and thatlimbic CRFR1 is required for central control of HPAsystem feedback and hormonal adaptation to stress.196

Ucn1-KO

Two mouse lines deficient for the Ucn1 gene (Ucn1-KO) were independently generated.197,198 Ucn1-KOmice show normal endocrine stress responses in bothmouse lines,197,198 supporting the view that Ucn1 doesnot play a role (or has only a minor one) in stress-induced HPA axis regulation. Similar to the case ofCRFR2-KO, the behavioral phenotype of Ucn1-KOmice is controversial. Whereas Wang and colleaguesshowed no differences in anxiety-like behavior,198 Vetteret al. demonstrated that Ucn1-KO mice have increasedanxiety-like behavior in the EPM and the openfield tests; however, no differences were found in thelightedark transfer test.197

CTION AND BEHAVIOR

Page 11: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

DYSREGULATION OF THE CRF SYSTEM: MOOD DISORDERS 365

Ucn2-KO

Mice deficient for Ucn2 (Ucn2-KO) exhibit gender-specific alterations in the basal circadian rhythms ofACTH and corticosterone secretion, and in depressive-like behavior.199 The nocturnal ACTH and corticoste-rone levels were found to be higher in female Ucn2-KOmice compared to their wild-type littermates, but not inmale. No differences were found in stress-inducedhormone response in both genders.199 In addition,female mutant mice display reduced depressive-likebehavior assessed by both the forced swim test and thetail suspension test. The altered performance of femaleUcn2 null mice in tests of antidepressant activity wasbehaviorally specific, because Ucn2 null mutant micedid not differ in their anxiety-like behavior in the EPMor lightedark transfer tests.199 In addition, the differ-ences observed in the forced swim test could not beattributed to deficits in learning abilities, as no differ-ences were found between the genotypes in a cuedcontextual fear conditioning test.199

Ucn3-KO

Mice deficient for the Ucn3 gene (Ucn3-KO)were independently generated by two groups.200,201 Liet al.200 revealed an important role for Ucn3 in the regu-lation of glucose-induced insulin secretion in thepancreas,200 while not reporting any behavioral pheno-type. Deussing et al.201 recently assessed the secondUcn3 KO mouse model in several behavioral tests. Nodifferences were found in HPA axis regulation, anxiety-like behavior or depressive-like behavior between themutants and their wild-type littermates. However, ina social discrimination task, Ucn3-KO mice were ableto recognize previously encountered conspecifics fora longer time than wild-type littermates e an observa-tion that was also confirmed in CRFR2-KO mice in thesame study.201 Taken together, these findings suggesta specific role of the Ucn3/CRFR2 system in socialrecognition.

Ucn1/Ucn2 Double KO (dKO)

To further explore the physiological role of Ucn1 andUcn2 in mediating the central stress response, a doubleUcn1 and Ucn2-deficient mouse line (Ucn1/Ucn2dKO) was recently generated.156 Ucn1/Ucn2 dKOmale mice show a higher stress-induced corticosteroneresponse and an anxiolytic profile in the EPM, theopen field and the lightedark transfer tests, relative towild-type mice. Female Ucn1/Ucn2 dKO mice showno significant changes in basal or stress-induced cortico-sterone levels. However, similar to male mutants, ananxiolytic-like phenotype was observed in the EPM

III. HORMONES, BRAIN FUN

and the open field tests.156 Whereas both female andmale wild-type mice exhibited an expected increase inanxiety-like behavior when subjected to restraint stressbefore the tests, Ucn1/Ucn2 dKO mice showed a signifi-cantly smaller stress-induced change in behavior.156

Ucn1/Ucn2/Ucn3 Triple KO (tKO)

To better understand the role of the endogenousCRFR2 ligands Ucn1, Ucn2 and Ucn3 in regulating thecentral stress response, a triple knockout (tKO) mousemodel lacking all three urocortin genes has been gener-ated.202 Intriguingly, these urocortin tKO mice exhibitincreased anxiety-like behaviors in the open field andlightedark transfer tests, 24 h following stress exposure,but not under unstressed conditions or immediatelyfollowing exposure to acute stress. In addition, urocortintKO mice exhibit a significantly higher stress-inducedincrease in acoustic startle-response. Together, theseresults suggest that lacking all urocortins has a limitedeffect on anxiety under non-challenged conditions butrenders the mice susceptible to the effects of stress,possibly by impairing recovery mechanisms.

DYSREGULATION OF THE CRF SYSTEM:MOOD DISORDERS

Depression

Several human studies have suggested that abnormalCRF neurotransmission and CRFR1 receptor signaltransduction play a central role in the pathophysiologyof depression (reviewed in Holsboer and Ising203; Binderand Nemeroff.204). For instance, the CSF concentrationsof CRF are elevated in depressive patients205e207 aswell as in suicide victims208 when compared to normalcontrols. In contrast, electroconvulsive therapy209 orantidepressant administration results in reduced levelsof CRF in the CSF.210e213 Following CRF administration,depressive patients display blunted ACTH release, sug-gesting a desensitization of CRF receptors caused byCRF hypersecretion.214e216 Consistent with this hypoth-esis, it was found that CRF binding sites are reduced inthe forebrain of depressive suicide victims,217,218

whereas CRF concentrations are elevated in the PVN,pontine nucleus, locus coeruleus and cortex.219e223

Moreover, in depressive patients Ucn1 is upregulatedin the Edinger-Westphal nucleus224 and Ucn3 in theprefrontal cortex,225 suggesting a possible role forCRFR2 in the development of depression.

Several genetic association studies have linked poly-morphisms in the CRF family genes with depressionand suicidality.226,227 A CRFR1 haplotype with allelesG-A-G for the single nucleotide polymorphisms (SNP)

CTION AND BEHAVIOR

Page 12: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY366

rs1876828, rs242939 and rs242941, respectively, wasfound to be significantly associated with a higherresponse to antidepressants in major depressivepatients,228,229 whereas the haplotype defined by thealleles G-G-T for the same SNPs was found to be over-represented in major depressive patients.230 Further-more, the CRFR1 intronic SNP rs4792887 has beenlinked to suicidal attempts in major depressivemen,231,232 whereas the A-allele of rs110402 has beenassociated with onset and seasonal pattern of majordepression among females233 and depression in malesuicide attempters.232 Interestingly, multiple individualSNPs, as well as a common haplotype spanning theCRFR1 intron 1 locus, appear to attenuate the effect ofchildhood abuse on the risk for adult depressive symp-tomatology.234 Regarding the CRFR2 gene, several posi-tive associations have been reported. A CRFR2haplotype consisting of three microsatellites has beenassociated with high severity suicidal behavior in bipolardisorder.235 In addition, allele G carriers of the rs2270007SNP in the CRFR2 gene showed a worse overall responseto antidepressant treatment.233 Finally, one study foundthat the rs10473984 SNP within the CRFBP locusshowed a significant association with both remissionand reduction in depressive symptoms followingantidepressant treatment.226

A large number of selective CRFR1 antagonists fromdifferent pharmaceutical companies have been devel-oped in the past two decades as novel therapeutics foranxiety and depression. However, few of them haveentered clinical development and, despite major efforts,to date no CRFR1 antagonist has successfully completeda Phase III trial.34,203 An open-label Phase IIa clinicaltrial assessed the effects of NBI-30775/R121919, anon-peptide high-affinity CRFR1 antagonist, indepressed patients.236e238 The group receiving NBI-30775/R121919 exhibited a safety and tolerabilityprofile,237,238 increased slow-wave sleep and decreasedREM density,236 and reduced depression and anxietyscores.238 Several other CRFR1 antagonists are currentlyin clinical development for the treatment of anxiety anddepression.

Anxiety

The anxiogenic effects of CRF administration in labo-ratory animals have led to the suggestion that the CRFsystem may also be involved in the pathophysiologyof anxiety disorders.138,161,239 Altered HPA axis functionhas been reported in PTSD240,241 and panic disorderpatients.242,243 In addition, several clinical studies havefound elevated CRF concentrations in the CSF of post-traumatic stress disorder (PTSD) patients.244e246 Incontrast, no alterations in CSF CRF concentrationshave been found in patients with generalized anxiety

III. HORMONES, BRAIN FUN

disorder (GAD),247e249 panic disorder247e249 or obses-sive-compulsive disorder (OCD).248,250

A few association studies have linked polymor-phisms in the CRF family with anxiety disorders.204 ACRF-linked microsatellite marker situated 23 kb 30 ofthe gene, and three SNPs, rs6999100, rs6159 andrs1870393, have been associated with behavioral inhibi-tion, a heritable temperamental phenotype considereda risk factor for anxiety disorders.251,252 This associationwas particularly evident in the families in which at leastone of the parents was diagnosed with panic disorder.251

Significant associations between SNPs in the CRFR1gene and panic disorder have been reported in two inde-pendent samples.253 Intriguingly, the results were evenstronger when the effects of the CRFR1 rs878886 SNPwere combined with the AVPR1B (vasopressin receptor1B) rs28632197 SNP, suggesting a potential genetic inter-action between CRFR1 and AVPR1B polymorphisms inthe development of panic disorder.253

CONCLUDING REMARKS

Stress-related psychopathologies, such as depressionand anxiety disorders, represent some of the mostcommon and proliferating health problems world-wide.254,255 Better understanding of the etiology of thesedisorders may fill the urgent requirement for develop-ment of novel and improved therapeutics. The currentview on the etiology of mental disorders is of a complexinteraction between genetic and environmental factors.Certain genetic backgrounds might constitute a predis-position for the development of mental disorder, whichwill onset with the occurrence of additional environ-mental factors.256,257

One of the key environmental risk factors thatcontribute to the onset of a variety of psychopathologiesis stress.7,258e260 When a situation is perceived asstressful, the brain activates many neuronal circuits,linking centers involved in sensory, motor, autonomic,neuroendocrine, cognitive and emotional functions inorder to adapt to the demand. However, the details ofthe pathways by which the brain translates stressfulstimuli into the final, integrated biological response arenot completely understood. Nevertheless, it is clearthat dysregulation of these physiological responses tostress can have severe psychological and physiologicalconsequences, and there is growing evidence to suggestthat inappropriate regulation, disproportional intensity,or chronic and/or irreversible activation of the stressresponse is linked to the etiology and pathophysiologyof anxiety disorders and depression.204,261,262

Previous studies suggested that the CRF/urocortinsystems in the brain have a unique role in mediatingbehavioral and physiological responses to diverse

CTION AND BEHAVIOR

Page 13: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

REFERENCES 367

stressors.1,5,8e10,137,162,204,263,264 However, while theinvolvement of the CRF/CRFR1 system in regulatingthe activation of the HPA axis and the regulation ofstress-linked behaviors is well established, the role ofthe central urocortins/CRFR2 system is less understood.The urocortins/CRFR2 system may be particularlyimportant in situations where an organism must mobi-lize not only the HPA system but also the central nervoussystem in response to environmental challenge. Clearly,dysfunction in such a fundamental brain-activatingsystemmay be the key to a variety of pathophysiologicalconditions involving abnormal responses to stressors,such as anxiety disorders, affective disorders andanorexia nervosa.

Evidence from studies employing competitivepeptides, or small-molecule CRF/urocortin receptorantagonists, suggested that the brain CRF/urocortinsystems play diverse roles in mediating behavioralresponses to stress. Based on the complementary behav-ioral phenotypes of CRFR1- and CRFR2-deficient (KO)mice, opposing roles were suggested for the two CRFreceptor systems in modulating anxiety-like behaviors.CRFR1-KO mice display decreased anxiety-like behav-iors coupled with an impaired HPA axis stress response,while CRFR2-KO mice show increased anxiety-likebehaviors and an accelerated HPA axis response tostress. Thus, the CRFeCRFR1 system was suggested ascritical for initiating stress responses, while the urocor-tinseCRFR2 system was suggested to terminate it orrestore allostasis. Nevertheless, the anxiety-relatedeffects of CRFR2 agonist and antagonist administrationinto the cerebral ventricles or into specific brain regionswere less consistent, with some evidence for brain-siteor ligand specificity.

Mice lacking all three urocortin genes (tKO) exhibitincreased anxiety-like behaviors 24 hours followingstress exposure, but not under unstressed conditionsor immediately following exposure to acute stress.This suggests that the urocortins might play an essentialrole in the stress-recovery process. Mouse models ofUcn1, Ucn2 and Ucn3 individual KOs have not indi-cated clear and robust changes in stress-related behav-iors; this may reflect differences in the time pointfollowing the stress exposure at which these mice weretested. Therefore, further “dissection” of the contribu-tion of each urocortin to the tKO phenotype using longi-tudinal comparative studies in both sexes, underdifferent time points following stress exposure, maypromote further understanding of the role of each uro-cortin gene product in the regulation of the central stressresponse.

Conventional gene knockout models, generated forthe different urocortin genes, have provided importantinformation toward elucidating the function of thesegenes. However, these mice showed significant changes

III. HORMONES, BRAIN FUN

in the expression levels of the other CRF familymembers in the CNS, likely due to developmentalcompensatory mechanisms, which may have contrib-uted to the observed stress-related phenotypes. In orderboth to avoid the developmental compensatory changesand to genetically target the gene of interest withinspecific brain nuclei, future studies will need to usemore specific transgenic mice models and viral toolsthat will allow the manipulation of both the levels andsite of urocortin gene expression in adult mice.

References

1. Vale W, Spiess J, Rivier C, Rivier J. Characterization of a 41-residue ovine hypothalamic peptide that stimulates secretion ofcorticotropin and beta-endorphin. Science. 1981;213:1394e1397.

2. Rivier C, Vale W. Modulation of stress-induced ACTH releaseby corticotropin-releasing factor, catecholamines and vaso-pressin. Nature. 1983;305:325e327.

3. Muglia L, Jacobson L, Dikkes P, Majzoub JA. Corticotropin-releasing hormone deficiency reveals major fetal but not adultglucocorticoid need. Nature. 1995;373:427e432.

4. Sutton RE, Koob GF, Le Moal M, Rivier J, Vale W. Corticotropinreleasing factor produces behavioural activation in rats. Nature.

1982;297:331e333.5. Koob GF, Heinrichs SC. A role for corticotropin releasing factor

and urocortin in behavioral responses to stressors. Brain Res.1999;848:141e152.

6. Brown MR, Fisher LA, Spiess J, Rivier C, Rivier J, Vale W.Corticotropin-releasing factor: actions on the sympatheticnervous system and metabolism. Endocrinology. 1982;111:928e931.

7. de Kloet ER, Joels M, Holsboer F. Stress and the brain: fromadaptation to disease. Nat Rev Neurosci. 2005;6:463e475.

8. Chrousos GP, Gold PW. The concepts of stress and stress systemdisorders. Overview of physical and behavioral homeostasis. JAm Med Assoc. 1992;267:1244e1252.

9. Holsboer F. The rationale for corticotropin-releasing hormonereceptor (CRH-R) antagonists to treat depression and anxiety. JPsychiatr Res. 1999;33:181e214.

10. Holmes A, Heilig M, Rupniak NM, Steckler T, Griebel G.Neuropeptide systems as novel therapeutic targets for depres-sion and anxiety disorders. Trends Pharmacol Sci. 2003;24:580e588.

11. Vaughan J, Donaldson C, Bittencourt J, Perrin MH, Lewis K,Sutton S, Chan R, Turnbull AV, Lovejoy D, Rivier C, et al.Urocortin, a mammalian neuropeptide related to fish urotensin Iand to corticotropin-releasing factor. Nature. 1995;378:287e292.

12. Reyes TM, Lewis K, Perrin MH, Kunitake KS, Vaughan J,Arias CA, Hogenesch JB, Gulyas J, Rivier J, Vale WW,Sawchenko PE. Urocortin II: a member of the corticotropin-releasing factor (CRF) neuropeptide family that is selectivelybound by type 2 CRF receptors. Proc Natl Acad Sci USA.2001;98:2843e2848.

13. Hsu SY, Hsueh AJ. Human stresscopin and stresscopin-relatedpeptide are selective ligands for the type 2 corticotropin-releasing hormone receptor. Nat Med. 2001;7:605e611.

14. Lewis K, Li C, Perrin MH, Blount A, Kunitake K, Donaldson C,Vaughan J, Reyes TM, Gulyas J, Fischer W, Bilezikjian L,Rivier J, Sawchenko PE, Vale WW. Identification of urocortin III,an additional member of the corticotropin-releasing factor(CRF) family with high affinity for the CRF2 receptor. Proc Natl

Acad Sci USA. 2001;98:7570e7575.

CTION AND BEHAVIOR

Page 14: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY368

15. Chen R, Lewis KA, Perrin MH, Vale WW. Expression cloning ofa human corticotropin-releasing-factor receptor. Proc Natl Acad

Sci USA. 1993;90:8967e8971.16. Vita N, Laurent P, Lefort S, Chalon P, Lelias JM, Kaghad M, Le

Fur G, Caput D, Ferrara P. Primary structure and functionalexpression of mouse pituitary and human brain corticotrophinreleasing factor receptors. FEBS Lett. 1993;335:1e5.

17. Chang CP, Pearse II RV, O’Connell S, Rosenfeld MG. Identifi-cation of a seven transmembrane helix receptor for cortico-tropin-releasing factor and sauvagine in mammalian brain.Neuron. 1993;11:1187e1195.

18. Perrin M, Donaldson C, Chen R, Blount A, Berggren T,Bilezikjian L, Sawchenko P, Vale W. Identification of a secondcorticotropin-releasing factor receptor gene and characterizationof a cDNA expressed in heart. Proc Natl Acad Sci USA.

1995;92:2969e2973.19. Stenzel P, Kesterson R, Yeung W, Cone RD, Rittenberg MB,

Stenzel-Poore MP. Identification of a novel murine receptor forcorticotropin-releasing hormone expressed in the heart. Mol

Endocrinol. 1995;9:637e645.20. Kishimoto T, Pearse II RV, Lin CR, Rosenfeld MG. A sauvagine/

corticotropin-releasing factor receptor expressed in heart andskeletal muscle. Proc Natl Acad Sci USA. 1995;92:1108e1112.

21. Lovenberg TW, Liaw CW, Grigoriadis DE, Clevenger W,Chalmers DT, De Souza EB, Oltersdorf T. Cloning and charac-terization of a functionally distinct corticotropin-releasing factorreceptor subtype from rat brain. Proc Natl Acad Sci USA.

1995;92:836e840.22. Chen A, Perrin M, Brar B, Li C, Jamieson P, Digruccio M,

Lewis K, Vale W. Mouse corticotropin-releasing factor receptortype 2alpha gene: isolation, distribution, pharmacologicalcharacterization and regulation by stress and glucocorticoids.Mol Endocrinol. 2005;19:441e458.

23. Pisarchik A, Slominski A. Molecular and functional character-ization of novel CRFR1 isoforms from the skin. Eur J Biochem.

2004;271:2821e2830.24. Grammatopoulos DK, Dai Y, Randeva HS, Levine MA,

Karteris E, Easton AJ, Hillhouse EW. A novel spliced variant ofthe type 1 corticotropin-releasing hormone receptor witha deletion in the seventh transmembrane domain present in thehuman pregnant term myometrium and fetal membranes. Mol

Endocrinol. 1999;13:2189e2202.25. Van Pett K, Viau V, Bittencourt JC, Chan RK, Li HY, Arias C,

Prins GS, Perrin M, Vale W, Sawchenko PE. Distribution ofmRNAs encoding CRF receptors in brain and pituitary of ratand mouse. J Comp Neurol. 2000;428:191e212.

26. Kostich WA, Chen A, Sperle K, Largent BL. Molecular identi-fication and analysis of a novel human corticotropin-releasingfactor (CRF) receptor: the CRF2gamma receptor. Mol Endocrinol.

1998;12:1077e1085.27. Lovenberg TW, Chalmers DT, Liu C, De Souza EB. CRF2 alpha

and CRF2 beta receptor mRNAs are differentially distributedbetween the rat central nervous system and peripheral tissues.Endocrinology. 1995;136:4139e4142.

28. Potter E, Behan DP, Fischer WH, Linton EA, Lowry PJ,Vale WW. Cloning and characterization of the cDNAs forhuman and rat corticotropin releasing factor-binding proteins.Nature. 1991;349:423e426.

29. Behan DP, De Souza EB, Lowry PJ, Potter E, Sawchenko P,Vale WW. Corticotropin releasing factor (CRF) binding protein:a novel regulator of CRF and related peptides. Front Neuro-

endocrinol. 1995;16:362e382.30. Seasholtz AF, Valverde RA, Denver RJ. Corticotropin-releasing

hormone-binding protein: biochemistry and function fromfishes to mammals. J Endocrinol. 2002;175:89e97.

III. HORMONES, BRAIN FUN

31. Dunn AJ, Berridge CW. Physiological and behavioral responsesto corticotropin-releasing factor administration: is CRF a medi-ator of anxiety or stress responses? Brain Res Brain Res Rev.1990;15:71e100.

32. Steckler T, Holsboer F. Corticotropin-releasing hormone receptorsubtypes and emotion. Biol Psychiatry. 1999;46:1480e1508.

33. Heinrichs SC, Koob GF. Corticotropin-releasing factor in brain:a role in activation, arousal, and affect regulation. J Pharmacol

Exp Ther. 2004;311:427e440.34. Zorrilla EP, Koob GF. Progress in corticotropin-releasing factor-1

antagonist development. Drug Discov Today 15, 371-83.35. Lowry CA, Moore FL. Regulation of behavioral responses by

corticotropin-releasing factor. Gen Comp Endocrinol. 2006;146:19e27.

36. Dunn AJ, Berridge CW, Lai YI, Yachabach TL. CRF-inducedexcessive grooming behavior in rats and mice. Peptides.

1987;8:841e844.37. Sherman JE, Kalin NH. The effects of ICV-CRH on novelty-

induced behavior. Pharmacol Biochem Behav. 1987;26:699e703.38. Jones DN, Kortekaas R, Slade PD, Middlemiss DN, Hagan JJ.

The behavioural effects of corticotropin-releasing factor-relatedpeptides in rats. Psychopharmacology (Berl). 1998;138:124e132.

39. Eaves M, Thatcher-Britton K, Rivier J, Vale W, Koob GF. Effectsof corticotropin releasing factor on locomotor activity inhypophysectomized rats. Peptides. 1985;6:923e926.

40. Britton DR, Varela M, Garcia A, Rosenthal M. Dexamethasonesuppresses pituitary-adrenal but not behavioral effects of cen-trally administered CRF. Life Sci. 1986;38:211e216.

41. Moore FL, Miller LJ. Stress-induced inhibition of sexualbehavior: corticosterone inhibits courtship behaviors of a maleamphibian (Taricha granulosa). Horm Behav. 1984;18:400e410.

42. Lowry CA, Deviche P, Moore FL. Effects of corticotropin-releasing factor (CRF) and opiates on amphibian locomotion.Brain Res. 1990;513:94e100.

43. Lowry CA, Burke KA, Renner KJ, Moore FL, Orchinik M.Rapid changes in monoamine levels following administrationof corticotropin-releasing factor or corticosterone are localizedin the dorsomedial hypothalamus. Horm Behav. 2001;39:195e205.

44. Lowry CA, Moore FL. Corticotropin-releasing factor (CRF)antagonist suppresses stress-induced locomotor activity in anamphibian. Horm Behav. 1991;25:84e96.

45. Clements S, Schreck CB, Larsen DA, Dickhoff WW. Centraladministration of corticotropin-releasing hormone stimulateslocomotor activity in juvenile chinook salmon (Oncorhynchus

tshawytscha). Gen Comp Endocrinol. 2002;125:319e327.46. Clements S, Schreck CB. Central administration of corticotropin-

releasing hormone alters downstream movement in an artificialstream in juvenile chinook salmon (Oncorhynchus tshawytscha).Gen Comp Endocrinol. 2004;137:1e8.

47. Ohgushi A, Bungo T, Shimojo M, Masuda Y, Denbow DM,Furuse M. Relationships between feeding and locomotionbehaviors after central administration of CRF in chicks. PhysiolBehav. 2001;72:287e289.

48. Zhang R, Nakanishi T, Ohgushi A, Ando R, Yoshimatsu T,Denbow DM, Furuse M. Interaction of corticotropin-releasingfactor and glucagon-like peptide-1 on behaviors in chicks. Eur JPharmacol. 2001;430:73e78.

49. Johnson RW, von Borell EH, Anderson LL, Kojic LD,Cunnick JE. Intracerebroventricular injection of corticotropin-releasing hormone in the pig: acute effects on behavior,adrenocorticotropin secretion, and immune suppression. Endo-crinology. 1994;135:642e648.

50. Salak-Johnson JL, McGlone JJ, Whisnant CS, Norman RL,Kraeling RR. Intracerebroventricular porcine corticotropin-

CTION AND BEHAVIOR

Page 15: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

REFERENCES 369

releasing hormone and cortisol effects on pig immune measuresand behavior. Physiol Behav. 1997;61:15e23.

51. Parrott RF, Vellucci SV, Goode JA. Behavioral and hormonaleffects of centrally injected "anxiogenic" neuropeptides ingrowing pigs. Pharmacol Biochem Behav. 2000;65:123e129.

52. Parrott RF, Vellucci SV. Behaviour of pigs given corticotrophin-releasing hormone in combination with flumazenil or diaz-epam. Pharmacol Biochem Behav. 2000;67:465e471.

53. Berridge CW, Dunn AJ. Corticotropin-releasing factor elicitsnaloxone sensitive stress-like alterations in exploratory behaviorin mice. Regul Pept. 1986;16:83e93.

54. Dunn AJ, Berridge CW. Corticotropin-releasing factor admin-istration elicits a stress-like activation of cerebral catechol-aminergic systems. Pharmacol Biochem Behav. 1987;27:685e691.

55. Kalin NH, Shelton SE, Kraemer GW, McKinney WT. Cortico-tropin-releasing factor administered intraventricularly to rhesusmonkeys. Peptides. 1983;4:217e220.

56. Winslow JT, Newman JD, Insel TR. CRH and alpha-helical-CRHmodulate behavioral measures of arousal in monkeys. Pharma-

col Biochem Behav. 1989;32:919e926.57. Takahashi LK, Kalin NH, Vanden Burgt JA, Sherman JE.

Corticotropin-releasing factor modulates defensive-withdrawaland exploratory behavior in rats. Behav Neurosci. 1989;103:648e654.

58. Baldwin HA, Rassnick S, Rivier J, Koob GF, Britton KT. CRFantagonist reverses the "anxiogenic" response to ethanol with-drawal in the rat. Psychopharmacology (Berl). 1991;103:227e232.

59. Momose K, Inui A, Asakawa A, Ueno N, Nakajima M,Fujimiya M, Kasuga M. Intracerebroventricularly administeredcorticotropin-releasing factor inhibits food intake and producesanxiety-like behaviour at very low doses in mice. Diabetes Obes

Metab. 1999;1:281e284.60. Berridge CW, Dunn AJ. CRF and restraint-stress decrease

exploratory behavior in hypophysectomized mice. Pharmacol

Biochem Behav. 1989;34:517e519.61. Spadaro F, Berridge CW, Baldwin HA, Dunn AJ. Corticotropin-

releasing factor acts via a third ventricle site to reduce explor-atory behavior in rats. Pharmacol Biochem Behav. 1990;36:305e309.

62. Sherman JE, Kalin NH. ICV-CRH alters stress-induced freezingbehavior without affecting pain sensitivity. Pharmacol BiochemBehav. 1988;30:801e807.

63. PelleymounterMA, JoppaM, CarmoucheM, CullenMJ, Brown B,Murphy B, Grigoriadis DE, Ling N, Foster AC. Role of cortico-tropin-releasing factor (CRF) receptors in the anorexic syndromeinduced by CRF. J Pharmacol Exp Ther. 2000;293:799e806.

64. Dunn AJ, File SE. Corticotropin-releasing factor has an anxio-genic action in the social interaction test. Horm Behav. 1987;21:193e202.

65. Swerdlow NR, Geyer MA, Vale WW, Koob GF. Corticotropin-releasing factor potentiates acoustic startle in rats: blockade bychlordiazepoxide. Psychopharmacology (Berl). 1986;88:147e152.

66. Liang KC, Melia KR, Miserendino MJ, Falls WA, Campeau S,Davis M. Corticotropin-releasing factor: long-lasting facilitationof the acoustic startle reflex. J Neurosci. 1992;12:2303e2312.

67. Diamant M, Croiset G, de Wied D. The effect of cortico-tropin-releasing factor (CRF) on autonomic and behavioralresponses during shock-prod burying test in rats. Peptides. 1992;13:1149e1158.

68. Arase K, York DA, Shimizu H, Shargill N, Bray GA. Effects ofcorticotropin-releasing factor on food intake and brownadipose tissue thermogenesis in rats. Am J Physiol.

1988;255:E255eE259.69. Krahn DD, Gosnell BA, Grace M, Levine AS. CRF antagonist

partially reverses CRF- and stress-induced effects on feeding.Brain Res Bull. 1986;17:285e289.

III. HORMONES, BRAIN FUN

70. Gosnell BA, Morley JE, Levine AS. A comparison of the effectsof corticotropin releasing factor and sauvagine on food intake.Pharmacol Biochem Behav. 1983;19:771e775.

71. Benoit SC, Thiele TE, Heinrichs SC, Rushing PA, Blake KA,Steeley RJ. Comparison of central administration of cortico-tropin-releasing hormone and urocortin on food intake, condi-tioned taste aversion, and c-Fos expression. Peptides.2000;21:345e351.

72. Pedersen CA, Caldwell JD, McGuire M, Evans DL. Cortico-tropin-releasing hormone inhibits maternal behavior andinduces pup-killing. Life Sci. 1991;48:1537e1546.

73. Sirinathsinghji DJ. Inhibitory influence of corticotropin releasingfactor on components of sexual behaviour in the male rat. BrainRes. 1987;407:185e190.

74. Cador M, Ahmed SH, Koob GF, Le Moal M, Stinus L. Cortico-tropin-releasing factor induces a place aversion independent ofits neuroendocrine role. Brain Res. 1992;597:304e309.

75. Heinrichs SC, Britton KT, Koob GF. Both conditioned tastepreference and aversion induced by corticotropin-releasingfactor. Pharmacol Biochem Behav. 1991;40:717e721.

76. Moreau JL, Kilpatrick G, Jenck F. Urocortin, a novel neuropep-tide with anxiogenic-like properties. NeuroReport. 1997;8:1697e1701.

77. Spina MG, Merlo-Pich E, Akwa Y, Balducci C, Basso AM,Zorrilla EP, Britton KT, Rivier J, Vale WW, Koob GF. Time-dependent induction of anxiogenic-like effects after centralinfusion of urocortin or corticotropin-releasing factor in the rat.Psychopharmacology (Berl). 2002;160:113e121.

78. Regev L, Ezrielev E, Gershon E, Gil S, Chen A. Genetic approachfor intracerebroventricular delivery. Proc Natl Acad Sci USA.

2010;107:4424e4429.79. LeDoux J. The amygdala. Curr Biol. 2007;17:R868eR874.80. Ehrlich I, Humeau Y, Grenier F, Ciocchi S, Herry C, Luthi A.

Amygdala inhibitory circuits and the control of fear memory.Neuron. 2009;62:757e771.

81. Roozendaal B, McEwen BS, Chattarji S. Stress, memory and theamygdala. Nat Rev Neurosci. 2009;10:423e433.

82. Palkovits M, Brownstein MJ, Vale W. Distribution of cortico-tropin-releasing factor in rat brain. Fed Proc. 1985;44:215e219.

83. Swanson LW, Sawchenko PE, Rivier J, Vale WW. Organizationof ovine corticotropin-releasing factor immunoreactive cells andfibers in the rat brain: an immunohistochemical study. Neuro-

endocrinology. 1983;36:165e186.84. De Souza EB, Insel TR, Perrin MH, Rivier J, Vale WW, Kuhar MJ.

Corticotropin-releasing factor receptors are widely distributedwithin the rat central nervous system: an autoradiographicstudy. J Neurosci. 1985;5:3189e3203.

85. Justice NJ, Yuan ZF, Sawchenko PE, Vale W. Type 1 cortico-tropin-releasing factor receptor expression reported in BACtransgenic mice: implications for reconciling ligand-receptormismatch in the central corticotropin-releasing factor system. JComp Neurol. 2008;511:479e496.

86. Tazi A, Swerdlow NR, LeMoal M, Rivier J, Vale W, Koob GF.Behavioral activation by CRF: evidence for the involvement ofthe ventral forebrain. Life Sci. 1987;41:41e49.

87. Wiersma A, Bohus B, Koolhaas JM. Corticotropin-releasinghormone microinfusion in the central amygdala diminishesa cardiac parasympathetic outflow under stress-free conditions.Brain Res. 1993;625:219e227.

88. Liang KC, Lee EH. Intra-amygdala injections of corticotropinreleasing factor facilitate inhibitory avoidance learning andreduce exploratory behavior in rats. Psychopharmacology (Berl).

1988;96:232e236.89. Sajdyk TJ, Schober DA, Gehlert DR, Shekhar A. Role of corti-

cotropin-releasing factor and urocortin within the basolateral

CTION AND BEHAVIOR

Page 16: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY370

amygdala of rats in anxiety and panic responses. Behav Brain

Res. 1999;100:207e215.90. Spiga F, Lightman SL, Shekhar A, Lowry CA. Injections of

urocortin 1 into the basolateral amygdala induce anxiety-likebehavior and c-Fos expression in brainstem serotonergicneurons. Neuroscience. 2006;138:1265e1276.

91. Rainnie DG, Bergeron R, Sajdyk TJ, Patil M, Gehlert DR,Shekhar A. Corticotrophin releasing factor-induced synapticplasticity in the amygdala translates stress into emotionaldisorders. J Neurosci. 2004;24:3471e3479.

92. Jochman KA, Newman SM, Kalin NH, Bakshi VP. Cortico-tropin-releasing factor-1 receptors in the basolateral amygdalamediate stress-induced anorexia. Behav Neurosci. 2005;119:1448e1458.

93. Keen-Rhinehart E, Michopoulos V, Toufexis DJ, Martin EI,Nair H, Ressler KJ, Davis M, Owens MJ, Nemeroff CB,Wilson ME. Continuous expression of corticotropin-releasingfactor in the central nucleus of the amygdala emulates thedysregulation of the stress and reproductive axes. Mol Psychi-

atry. 2009;14:37e50.94. Regev L, Neufeld-Cohen A, Tsoory M, Kuperman Y,

Getselter D, Gil S, Chen A. Prolonged and site-specific over-expression of corticotropin-releasing factor reveals differentialroles for extended amygdala nuclei in emotional regulation.Mol

Psychiatry; 2011, in press.95. Heinrichs SC, Pich EM, Miczek KA, Britton KT, Koob GF.

Corticotropin-releasing factor antagonist reduces emotionalityin socially defeated rats via direct neurotropic action. Brain Res.

1992;581:190e197.96. Swiergiel AH, Takahashi LK, Kalin NH. Attenuation of stress-

induced behavior by antagonism of corticotropin-releasingfactor receptors in the central amygdala in the rat. Brain Res.

1993;623:229e234.97. Liebsch G, Landgraf R, Gerstberger R, Probst JC, Wotjak CT,

Engelmann M, Holsboer F, Montkowski A. Chronic infusion ofa CRH1 receptor antisense oligodeoxynucleotide into the centralnucleus of the amygdala reduced anxiety-related behavior insocially defeated rats. Regul Pept. 1995;59:229e239.

98. Sajdyk TJ, Gehlert DR. Astressin, a corticotropin releasing factorantagonist, reverses the anxiogenic effects of urocortin whenadministered into the basolateral amygdala. Brain Res.

2000;877:226e234.99. Robison CL, Meyerhoff JL, Saviolakis GA, Chen WK, Rice KC,

Lumley LA. A CRH1 antagonist into the amygdala of miceprevents defeat-induced defensive behavior. Ann NY Acad Sci.

2004;1032:324e327.100. Sztainberg Y, Kuperman Y, Tsoory M, Lebow M, Chen A. The

anxiolytic effect of environmental enrichment is mediated viaamygdalar CRF receptor type 1.Mol Psychiatry. 2010;15:905e917.

101. Walker DL, Toufexis DJ, Davis M. Role of the bed nucleus of thestria terminalis versus the amygdala in fear, stress, and anxiety.Eur J Pharmacol. 2003;463:199e216.

102. Chalmers DT, Lovenberg TW, De Souza EB. Localization ofnovel corticotropin-releasing factor receptor (CRF2) mRNAexpression to specific subcortical nuclei in rat brain: comparisonwith CRF1 receptor mRNA expression. J Neurosci. 1995;15:6340e6350.

103. Walker DL, Miles LA, Davis M. Selective participation of thebed nucleus of the stria terminalis and CRF in sustainedanxiety-like versus phasic fear-like responses. Prog Neuro-

psychopharmacol Biol Psychiatry. 2009;33:1291e1308.104. Lee Y, Davis M. Role of the hippocampus, the bed nucleus of the

stria terminalis, and the amygdala in the excitatory effect ofcorticotropin-releasing hormone on the acoustic startle reflex. JNeurosci. 1997;17:6434e6446.

III. HORMONES, BRAIN FUN

105. Lee Y, Fitz S, Johnson PL, Shekhar A. Repeated stimulation ofCRF receptors in the BNST of rats selectively induces social butnot panic-like anxiety. Neuropsychopharmacology. 2008;33:2586e2594.

106. Sahuque LL, Kullberg EF, McGeehan AJ, Kinder JR, Hicks MP,Blanton MG, Janak PH, Olive MF. Anxiogenic and aversiveeffects of corticotropin-releasing factor (CRF) in the bed nucleusof the stria terminalis in the rat: role of CRF receptor subtypes.Psychopharmacology (Berl). 2006;186:122e132.

107. Liang KC, Chen HC, Chen DY. Posttraining infusion ofnorepinephrine and corticotropin releasing factor into the bednucleus of the stria terminalis enhanced retention in an inhibi-tory avoidance task. Chin J Physiol. 2001;44:33e43.

108. Ciccocioppo R, Fedeli A, Economidou D, Policani F, Weiss F,Massi M. The bed nucleus is a neuroanatomical substrate for theanorectic effect of corticotropin-releasing factor and for itsreversal by nociceptin/orphanin FQ. J Neurosci. 2003;23:9445e9451.

109. Silverman AJ, Hou-Yu A, Chen WP. Corticotropin-releasingfactor synapses within the paraventricular nucleus of thehypothalamus. Neuroendocrinology. 1989;49:291e299.

110. Gray TS, Magnuson DJ. Peptide immunoreactive neurons in theamygdala and the bed nucleus of the stria terminalis project tothe midbrain central gray in the rat. Peptides. 1992;13:451e460.

111. Monnikes H, Heymann-Monnikes I, Tache Y. CRF in the para-ventricular nucleus of the hypothalamus induces dose-relatedbehavioral profile in rats. Brain Res. 1992;574:70e76.

112. Krahn DD, Gosnell BA, Levine AS, Morley JE. Behavioral effectsof corticotropin-releasing factor: localization and characteriza-tion of central effects. Brain Res. 1988;443:63e69.

113. Heinrichs SC, Menzaghi F, Pich EM, Hauger RL, Koob GF.Corticotropin-releasing factor in the paraventricular nucleusmodulates feeding induced by neuropeptide Y. Brain Res.1993;611:18e24.

114. Elliott E, Ezra-Nevo G, Regev L, Neufeld-Cohen A, Chen A.Resilience to social stress coincides with functional DNAmethylation of the Crf gene in adult mice. Nat Neurosci.2010;13:1351e1353.

115. Phillips RG, LeDoux JE. Differential contribution of amygdalaand hippocampus to cued and contextual fear conditioning.Behav Neurosci. 1992;106:274e285.

116. Maren S, Aharonov G, Fanselow MS. Neurotoxic lesions of thedorsal hippocampus and Pavlovian fear conditioning in rats.Behav Brain Res. 1997;88:261e274.

117. Radulovic J, Ruhmann A, Liepold T, Spiess J. Modulation oflearning and anxiety by corticotropin-releasing factor (CRF) andstress: differential roles of CRF receptors 1 and 2. J Neurosci.

1999;19:5016e5025.118. Todorovic C, Radulovic J, Jahn O, Radulovic M, Sherrin T,

Hippel C, Spiess J. Differential activation of CRF receptorsubtypes removes stress-induced memory deficit and anxiety.Eur J Neurosci. 2007;25:3385e3397.

119. Bertoglio LJ, Joca SR, Guimaraes FS. Further evidence thatanxiety and memory are regionally dissociated within thehippocampus. Behav Brain Res. 2006;175:183e188.

120. Pentkowski NS, Blanchard DC, Lever C, Litvin Y, Blanchard RJ.Effects of lesions to the dorsal and ventral hippocampuson defensive behaviors in rats. Eur J Neurosci. 2006;23:2185e2196.

121. Pentkowski NS, Litvin Y, Blanchard DC, Vasconcellos A,King LB, Blanchard RJ. Effects of acidic-astressin and ovine-CRFmicroinfusions into the ventral hippocampus on defensivebehaviors in rats. Horm Behav. 2009;56:35e43.

122. Cassens G, Roffman M, Kuruc A, Orsulak PJ, Schildkraut JJ.Alterations in brain norepinephrine metabolism induced by

CTION AND BEHAVIOR

Page 17: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

REFERENCES 371

environmental stimuli previously paired with inescapableshock. Science. 1980;209:1138e1140.

123. Korf J, Aghajanian GK, Roth RH. Increased turnover ofnorepinephrine in the rat cerebral cortex during stress: role ofthe locus coeruleus. Neuropharmacology. 1973;12:933e938.

124. Cedarbaum JM, Aghajanian GK. Activation of locus coeruleusneurons by peripheral stimuli: modulation by a collateralinhibitory mechanism. Life Sci. 1978;23:1383e1392.

125. Grzanna R, Molliver ME. The locus coeruleus in the rat: animmunohistochemical delineation. Neuroscience. 1980;5:21e40.

126. Swanson LW. The locus coeruleus: a cytoarchitectonic, Golgiand immunohistochemical study in the albino rat. Brain Res.

1976;110:39e56.127. Swanson LW, Hartman BK. The central adrenergic system.

An immunofluorescence study of the location of cell bodiesand their efferent connections in the rat utilizing dopamine-beta-hydroxylase as a marker. J Comp Neurol. 1975;163:467e505.

128. Butler PD, Weiss JM, Stout JC, Nemeroff CB. Corticotropin-releasing factor produces fear-enhancing and behavioral acti-vating effects following infusion into the locus coeruleus. J

Neurosci. 1990;10:176e183.129. Swiergiel AH, Takahashi LK, Rubin WW, Kalin NH. Antago-

nism of corticotropin-releasing factor receptors in the locuscoeruleus attenuates shock-induced freezing in rats. Brain Res.

1992;587:263e268.130. Smagin GN, Harris RB, Ryan DH. Corticotropin-releasing factor

receptor antagonist infused into the locus coeruleus attenuatesimmobilization stress-induced defensive withdrawal in rats.Neurosci Lett. 1996;220:167e170.

131. Chen MF, Chiu TH, Lee EH. Noradrenergic mediation of thememory-enhancing effect of corticotropin-releasing factor in thelocus coeruleus of rats. Psychoneuroendocrinology. 1992;17:113e124.

132. Fekete EM, Zorrilla EP. Physiology, pharmacology, and thera-peutic relevance of urocortins in mammals: ancient CRF paral-ogs. Front Neuroendocrinol. 2007;28:1e27.

133. Kuperman Y, Chen A. Urocortins: emerging metabolic andenergy homeostasis perspectives. Trends Endocrinol Metab.

2008;19:122e129.134. Joels M, Baram TZ. The neuro-symphony of stress. Nat Rev

Neurosci. 2009;10:459e466.135. Nemeroff CB. The role of corticotropin-releasing factor in the

pathogenesis of major depression. Pharmacopsychiatry. 1988;21:76e82.

136. Nemeroff CB. New vistas in neuropeptide research in neuro-psychiatry: focus on corticotropin-releasing factor. Neuro-

psychopharmacology. 1992;6:69e75.137. Arborelius L, Owens MJ, Plotsky PM, Nemeroff CB. The role of

corticotropin-releasing factor in depression and anxiety disor-ders. J Endocrinol. 1999;160:1e12.

138. Reul JM, Holsboer F. Corticotropin-releasing factor receptors 1and 2 in anxiety and depression. Curr Opin Pharmacol.

2002;2:23e33.139. Valdez GR, Inoue K, Koob GF, Rivier J, Vale W, Zorrilla EP.

Human urocortin II: mild locomotor suppressive and delayedanxiolytic-like effects of a novel corticotropin-releasing factorrelated peptide. Brain Res. 2002;943:142e150.

140. Valdez GR, Zorrilla EP, Rivier J, Vale WW, Koob GF. Locomotorsuppressive and anxiolytic-like effects of urocortin 3, a highlyselective type 2 corticotropin-releasing factor agonist. Brain Res.

2003;980:206e212.141. Henry B, Vale W, Markou A. The effect of lateral septum

corticotropin-releasing factor receptor 2 activation on anxiety ismodulated by stress. J Neurosci. 2006;26:9142e9152.

III. HORMONES, BRAIN FUN

142. Zhao Y, Valdez GR, Fekete EM, Rivier JE, Vale WW, Rice KC,Weiss F, Zorrilla EP. Subtype-selective corticotropin-releasingfactor receptor agonists exert contrasting, but not opposite,effects on anxiety-related behavior in rats. J Pharmacol Exp Ther.

2007;323:846e854.143. Heinrichs SC, Lapsansky J, Lovenberg TW, De Souza EB,

Chalmers DT. Corticotropin-releasing factor CRF1, but notCRF2, receptors mediate anxiogenic-like behavior. Regul Pept.1997;71:15e21.

144. Liebsch G, Landgraf R, Engelmann M, Lorscher P, Holsboer F.Differential behavioural effects of chronic infusion of CRH 1and CRH 2 receptor antisense oligonucleotides into the ratbrain. J Psychiatr Res. 1999;33:153e163.

145. Ruhmann A, Bonk I, Lin CR, Rosenfeld MG, Spiess J. Structuralrequirements for peptidic antagonists of the corticotropin-releasing factor receptor (CRFR): development of CRFR2beta-selective antisauvagine-30. Proc Natl Acad Sci USA.

1998;95:15264e15269.146. Takahashi LK, Ho SP, Livanov V, Graciani N, Arneric SP.

Antagonism of CRF(2) receptors produces anxiolyticbehavior in animal models of anxiety. Brain Res. 2001;902:135e142.

147. Ohata H, Shibasaki T. Effects of urocortin 2 and 3 on motoractivity and food intake in rats. Peptides. 2004;25:1703e1709.

148. Zorrilla EP, Reinhardt LE, Valdez GR, Inoue K, Rivier JE,Vale WW, Koob GF. Human urocortin 2, a corticotropin-releasing factor (CRF)2 agonist, and ovine CRF, a CRF1 agonist,differentially alter feeding and motor activity. J Pharmacol Exp

Ther. 2004;310:1027e1034.149. Venihaki M, Sakihara S, Subramanian S, Dikkes P, Weninger SC,

Liapakis G, Graf T, Majzoub JA. Urocortin III, a brain neuro-peptide of the corticotropin-releasing hormone family: modu-lation by stress and attenuation of some anxiety-like behaviours.J Neuroendocrinol. 2004;16:411e422.

150. Bakshi VP, Smith-Roe S, Newman SM, Grigoriadis DE,Kalin NH. Reduction of stress-induced behavior by antagonismof corticotropin-releasing hormone 2 (CRH2) receptors in lateralseptum or CRH1 receptors in amygdala. J Neurosci. 2002;22:2926e2935.

151. Hammack SE, Schmid MJ, LoPresti ML, Der-Avakian A,Pellymounter MA, Foster AC, Watkins LR, Maier SF. Cortico-tropin releasing hormone type 2 receptors in the dorsal raphenucleus mediate the behavioral consequences of uncontrollablestress. J Neurosci. 2003;23:1019e1025.

152. Ho SP, Takahashi LK, Livanov V, Spencer K, Lesher T, Maciag C,Smith MA, Rohrbach KW, Hartig PR, Arneric SP. Attenuation offear conditioning by antisense inhibition of brain corticotropinreleasing factor-2 receptor. Brain Res Mol Brain Res. 2001;89:29e40.

153. Risbrough VB, Hauger RL, Pelleymounter MA, Geyer MA. Roleof corticotropin releasing factor (CRF) receptors 1 and 2 in CRF-potentiated acoustic startle in mice. Psychopharmacology (Berl).2003;170:178e187.

154. Pelleymounter MA, Joppa M, Ling N, Foster AC. Pharmaco-logical evidence supporting a role for central corticotropin-releasing factor(2) receptors in behavioral, but not endocrine,response to environmental stress. J Pharmacol Exp Ther.

2002;302:145e152.155. Pelleymounter MA, Joppa M, Ling N, Foster AC. Behavioral

and neuroendocrine effects of the selective CRF2 receptoragonists urocortin II and urocortin III. Peptides. 2004;25:659e666.

156. Neufeld-Cohen A, Evans AK, Getselter D, Spyroglou A, Hill A,Gil S, Tsoory M, Beuschlein F, Lowry CA, Vale W, Chen A.Urocortin-1 and -2 double-deficient mice show robust anxiolytic

CTION AND BEHAVIOR

Page 18: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY372

phenotype and modified serotonergic activity in anxietycircuits. Mol Psychiatry. 15, 426-41, 339.

157. Staub DR, Evans AK, Lowry CA. Evidence supporting a role forcorticotropin-releasing factor type 2 (CRF2) receptors in theregulation of subpopulations of serotonergic neurons. Brain Res.

2006;1070:77e89.158. Staub DR, Spiga F, Lowry CA. Urocortin 2 increases c-Fos

expression in topographically organized subpopulations ofserotonergic neurons in the rat dorsal raphe nucleus. Brain Res.

2005;1044:176e189.159. Amat J, Tamblyn JP, Paul ED, Bland ST, Amat P, Foster AC,

Watkins LR, Maier SF. Microinjection of urocortin 2 into thedorsal raphe nucleus activates serotonergic neurons andincreases extracellular serotonin in the basolateral amygdala.Neuroscience. 2004;129:509e519.

160. Maier SF, Watkins LR. Stressor controllability and learnedhelplessness: the roles of the dorsal raphe nucleus, serotonin,and corticotropin-releasing factor. Neurosci Biobehav Rev.2005;29:829e841.

161. Bakshi VP, Kalin NH. Corticotropin-releasing hormone andanimal models of anxiety: gene-environment interactions. BiolPsychiatry. 2000;48:1175e1198.

162. Bale TL, Vale WW. CRF and CRF receptors: role in stressresponsivity and other behaviors. Annu Rev Pharmacol Toxicol.

2004;44:525e557.163. Keck ME, Ohl F, Holsboer F, Muller MB. Listening to mutant

mice: a spotlight on the role of CRF/CRF receptor systemsin affective disorders. Neurosci Biobehav Rev. 2005;29:867e889.

164. Stenzel-Poore MP, Cameron VA, Vaughan J, Sawchenko PE,Vale W. Development of Cushing’s syndrome in corticotropin-releasing factor transgenic mice. Endocrinology. 1992;130:3378e3386.

165. Stenzel-Poore MP, Heinrichs SC, Rivest S, Koob GF, Vale WW.Overproduction of corticotropin-releasing factor in transgenicmice: a genetic model of anxiogenic behavior. J Neurosci.

1994;14:2579e2584.166. van Gaalen MM, Stenzel-Poore MP, Holsboer F, Steckler T.

Effects of transgenic overproduction of CRH on anxiety-likebehaviour. Eur J Neurosci. 2002;15:2007e2015.

167. Heinrichs SC, Min H, Tamraz S, Carmouche M, Boehme SA,Vale WW. Anti-sexual and anxiogenic behavioral consequencesof corticotropin-releasing factor overexpression are centrallymediated. Psychoneuroendocrinology. 1997;22:215e224.

168. Heinrichs SC, Stenzel-Poore MP, Gold LH, Battenberg E,Bloom FE, Koob GF, Vale WW, Pich EM. Learning impairmentin transgenic mice with central overexpression of corticotropin-releasing factor. Neuroscience. 1996;74:303e311.

169. van Gaalen MM, Stenzel-Poore M, Holsboer F, Steckler T.Reduced attention in mice overproducing corticotropin-releasing hormone. Behav Brain Res. 2003;142:69e79.

170. Dirks A, Groenink L, Bouwknecht JA, Hijzen TH, Van DerGugten J, Ronken E, Verbeek JS, Veening JG, Dederen PJ,Korosi A, Schoolderman LF, Roubos EW, Olivier B. Over-expression of corticotropin-releasing hormone in transgenicmice and chronic stress-like autonomic and physiologicalalterations. Eur J Neurosci. 2002;16:1751e1760.

171. Dirks A, Groenink L, Schipholt MI, van der Gugten J,Hijzen TH, Geyer MA, Olivier B. Reduced startle reactivity andplasticity in transgenic mice overexpressing corticotropin-releasing hormone. Biol Psychiatry. 2002;51:583e590.

172. Kasahara M, Groenink L, Breuer M, Olivier B, Sarnyai Z.Altered behavioural adaptation in mice with neural corticotro-phin-releasing factor overexpression. Genes Brain Behav.

2007;6:598e607.

III. HORMONES, BRAIN FUN

173. Lu A, Steiner MA, Whittle N, Vogl AM, Walser SM, Ableitner M,Refojo D, Ekker M, Rubenstein JL, Stalla GK, Singewald N,Holsboer F, Wotjak CT, Wurst W, Deussing JM. Conditionalmouse mutants highlight mechanisms of corticotropin-releasinghormone effects on stress-coping behavior. Mol Psychiatry.

2008;13:1028e1042.174. Venihaki M, Majzoub JA. Animal models of CRH deficiency.

Front Neuroendocrinol. 1999;20:122e145.175. Dunn AJ, Swiergiel AH. Behavioral responses to stress are intact

in CRF-deficient mice. Brain Res. 1999;845:14e20.176. Weninger SC, Dunn AJ, Muglia LJ, Dikkes P, Miczek KA,

Swiergiel AH, Berridge CW, Majzoub JA. Stress-inducedbehaviors require the corticotropin-releasing hormone (CRH)receptor, but not CRH. Proc Natl Acad Sci USA. 1999;96:8283e8288.

177. Smith GW, Aubry JM, Dellu F, Contarino A, Bilezikjian LM,Gold LH, Chen R, Marchuk Y, Hauser C, Bentley CA,Sawchenko PE, Koob GF, Vale W, Lee KF. Corticotropinreleasing factor receptor 1-deficient mice display decreasedanxiety, impaired stress response, and aberrant neuroendocrinedevelopment. Neuron. 1998;20:1093e1102.

178. Timpl P, Spanagel R, Sillaber I, Kresse A, Reul JM, Stalla GK,Blanquet V, Steckler T, Holsboer F, Wurst W. Impaired stressresponse and reduced anxiety in mice lacking a functionalcorticotropin-releasing hormone receptor 1. Nat Genet. 1998;19:162e166.

179. Turnbull AV, Smith GW, Lee S, Vale WW, Lee KF, Rivier C. CRFtype I receptor-deficient mice exhibit a pronounced pituitary-adrenal response to local inflammation. Endocrinology.1999;140:1013e1017.

180. Contarino A, Dellu F, Koob GF, Smith GW, Lee KF, Vale W,Gold LH. Reduced anxiety-like and cognitive performance inmice lacking the corticotropin-releasing factor receptor 1. BrainRes. 1999;835:1e9.

181. Gammie SC, Stevenson SA. Intermale aggression in cortico-tropin-releasing factor receptor 1 deficient mice. Behav Brain Res.

2006;171:63e69.182. Gammie SC, Bethea ED, Stevenson SA. Altered maternal

profiles in corticotropin-releasing factor receptor 1 deficientmice. BMC Neurosci. 2007;8:17.

183. Bale TL, Contarino A, Smith GW, Chan R, Gold LH,Sawchenko PE, Koob GF, Vale WW, Lee KF. Mice deficient forcorticotropin-releasing hormone receptor-2 display anxiety-likebehaviour and are hypersensitive to stress. Nat Genet.2000;24:410e414.

184. Coste SC, Kesterson RA, Heldwein KA, Stevens SL, Heard AD,Hollis JH, Murray SE, Hill JK, Pantely GA, Hohimer AR,Hatton DC, Phillips TJ, Finn DA, Low MJ, Rittenberg MB,Stenzel P, Stenzel-Poore MP. Abnormal adaptations to stressand impaired cardiovascular function in mice lacking corti-cotropin-releasing hormone receptor-2. Nat Genet. 2000;24:403e409.

185. Kishimoto T, Radulovic J, Radulovic M, Lin CR, Schrick C,Hooshmand F, Hermanson O, Rosenfeld MG, Spiess J. Deletionof crhr2 reveals an anxiolytic role for corticotropin-releasinghormone receptor-2. Nat Genet. 2000;24:415e419.

186. Bale TL, Vale WW. Increased depression-like behaviors incorticotropin-releasing factor receptor-2-deficient mice: sexuallydichotomous responses. J Neurosci. 2003;23:5295e5301.

187. Todorovic C, Sherrin T, Pitts M, Hippel C, Rayner M, Spiess J.Suppression of the MEK/ERK signaling pathway reversesdepression-like behaviors of CRF2-deficient mice. Neuro-

psychopharmacology. 2009;34:1416e1426.188. Burrows HL, Nakajima M, Lesh JS, Goosens KA, Samuelson LC,

Inui A, Camper SA, Seasholtz AF. Excess corticotropin releasing

CTION AND BEHAVIOR

Page 19: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

REFERENCES 373

hormone-binding protein in the hypothalamic-pituitary-adrenalaxis in transgenic mice. J Clin Invest. 1998;101:1439e1447.

189. Lovejoy DA, Aubry JM, Turnbull A, Sutton S, Potter E,Yehling J, Rivier C, Vale WW. Ectopic expression of the CRF-binding protein: minor impact on HPA axis regulation butinduction of sexually dimorphic weight gain. J Neuroendocrinol.

1998;10:483e491.190. Karolyi IJ, Burrows HL, Ramesh TM, Nakajima M, Lesh JS,

Seong E, Camper SA, Seasholtz AF. Altered anxiety andweight gain in corticotropin-releasing hormone-bindingprotein-deficient mice. Proc Natl Acad Sci USA. 1999;96:11595e11600.

191. Gammie SC, Seasholtz AF, Stevenson SA. Deletion of cortico-tropin-releasing factor binding protein selectively impairsmaternal, but not intermale aggression. Neuroscience. 2008;157:502e512.

192. Bale TL, Picetti R, Contarino A, Koob GF, Vale WW, Lee KF.Mice deficient for both corticotropin-releasing factor receptor 1(CRFR1) and CRFR2 have an impaired stress response anddisplay sexually dichotomous anxiety-like behavior. J Neurosci.

2002;22:193e199.193. Preil J, Muller MB, Gesing A, Reul JM, Sillaber I, van

Gaalen MM, Landgrebe J, Holsboer F, Stenzel-Poore M,Wurst W. Regulation of the hypothalamic-pituitary-adrenocortical system in mice deficient for CRH receptors 1 and2. Endocrinology. 2001;142:4946e4955.

194. Korte SM, Korte-Bouws GA, Koob GF, De Kloet ER, Bohus B.Mineralocorticoid and glucocorticoid receptor antagonists inanimal models of anxiety. Pharmacol Biochem Behav.1996;54:261e267.

195. Tronche F, Kellendonk C, Kretz O, Gass P, Anlag K, Orban PC,Bock R, Klein R, Schutz G. Disruption of the glucocorticoidreceptor gene in the nervous system results in reduced anxiety.Nat Genet. 1999;23:99e103.

196. Muller MB, Zimmermann S, Sillaber I, Hagemeyer TP,Deussing JM, Timpl P, Kormann MS, Droste SK, Kuhn R,Reul JM, Holsboer F, Wurst W. Limbic corticotropin-releasinghormone receptor 1 mediates anxiety-related behavior andhormonal adaptation to stress. Nat Neurosci. 2003;6:1100e1107.

197. Vetter DE, Li C, Zhao L, Contarino A, Liberman MC, Smith GW,Marchuk Y, Koob GF, Heinemann SF, Vale W, Lee KF. Urocortin-deficient mice show hearing impairment and increased anxiety-like behavior. Nat Genet. 2002;31:363e369.

198. Wang X, Su H, Copenhagen LD, Vaishnav S, Pieri F, Shope CD,Brownell WE, De Biasi M, Paylor R, Bradley A. Urocortin-deficient mice display normal stress-induced anxiety behaviorand autonomic control but an impaired acoustic startleresponse. Mol Cell Biol. 2002;22:6605e6610.

199. Chen A, Zorrilla E, Smith S, Rousso D, Levy C, Vaughan J,Donaldson C, Roberts A, Lee KF, Vale W. Urocortin 2-deficientmice exhibit gender-specific alterations in circadian hypothal-amus-pituitary-adrenal axis and depressive-like behavior. J

Neurosci. 2006;26:5500e5510.200. Li C, Chen P, Vaughan J, Lee KF, Vale W. Urocortin 3 regulates

glucose-stimulated insulin secretion and energy homeostasis.Proc Natl Acad Sci USA. 2007;104:4206e4211.

201. Deussing JM, Breu J, Kuhne C, Kallnik M, Bunck M, Glasl L, YenYC, Schmidt MV, Zurmuhlen R, Vogl AM, Gailus-Durner V,Fuchs H, Holter SM, Wotjak CT, Landgraf R, de Angelis MH,Holsboer F, Wurst W. Urocortin 3 modulates social discrimi-nation abilities via corticotropin-releasing hormone receptortype 2. J Neurosci. 30, 9103-9116.

202. Neufeld-Cohen A, Tsoory MM, Evans AK, Getselter D, Gil S,Lowry CA, Vale WW, Chen A. A triple urocortin knockout

III. HORMONES, BRAIN FUN

mouse model reveals an essential role for urocortins in stressrecovery. Proc Natl Acad Sci USA. 1902;107:0e5.

203. Holsboer F, Ising M. Central CRH system in depression andanxietyeevidence from clinical studies with CRH1 receptorantagonists. Eur J Pharmacol. 2008;583:350e357.

204. Binder EB, Nemeroff CB. The CRF system, stress, depressionand anxiety-insights from human genetic studies. MolPsychiatry. 15, 574-88.

205. Nemeroff CB, Widerlov E, Bissette G, Walleus H, Karlsson I,Eklund K, Kilts CD, Loosen PT, Vale W. Elevated concentrationsof CSF corticotropin-releasing factor-like immunoreactivity indepressed patients. Science. 1984;226:1342e1344.

206. Banki CM, Bissette G, Arato M, O’Connor L, Nemeroff CB. CSFcorticotropin-releasing factor-like immunoreactivity in depres-sion and schizophrenia. Am J Psychiatry. 1987;144:873e877.

207. Hartline KM, Owens MJ, Nemeroff CB. Postmortem and cere-brospinal fluid studies of corticotropin-releasing factor inhumans. Ann NY Acad Sci. 1996;780:96e105.

208. Arato M, Banki CM, Bissette G, Nemeroff CB. Elevated CSF CRFin suicide victims. Biol Psychiatry. 1989;25:355e359.

209. Nemeroff CB, Bissette G, Akil H, Fink M. Neuropeptide concen-trations in the cerebrospinal fluid of depressed patients treatedwith electroconvulsive therapy. Corticotrophin-releasingfactor, beta-endorphin and somatostatin.Br J Psychiatry. 1991;158:59e63.

210. De Bellis MD, Gold PW, Geracioti Jr TD, Listwak SJ, Kling MA.Association of fluoxetine treatment with reductions in CSFconcentrations of corticotropin-releasing hormone and argininevasopressin in patients with major depression. Am J Psychiatry.1993;150:656e657.

211. Veith RC, Lewis N, Langohr JI, Murburg MM, Ashleigh EA,Castillo S, Peskind ER, Pascualy M, Bissette G, Nemeroff CB,et al. Effect of desipramine on cerebrospinal fluid concentrationsof corticotropin-releasing factor in human subjects. PsychiatryRes. 1993;46:1e8.

212. Heuser I, Bissette G, Dettling M, Schweiger U, Gotthardt U,Schmider J, Lammers CH, Nemeroff CB, Holsboer F. Cerebro-spinal fluid concentrations of corticotropin-releasing hormone,vasopressin, and somatostatin in depressed patients andhealthy controls: response to amitriptyline treatment. DepressAnxiety. 1998;8:71e79.

213. Banki CM, Karmacsi L, Bissette G, Nemeroff CB. CSF cortico-tropin-releasing hormone and somatostatin in major depression:response to antidepressant treatment and relapse. Eur Neuro-psychopharmacol. 1992;2:107e113.

214. Gold PW, Chrousos GP. Clinical studies with corticotropinreleasing factor: implications for the diagnosis and pathophys-iology of depression, Cushing’s disease, and adrenal insuffi-ciency. Psychoneuroendocrinology. 1985;10:401e419.

215. Holsboer F, Gerken A, Stalla GK, Muller OA. Blunted aldoste-rone and ACTH release after human CRH administration indepressed patients. Am J Psychiatry. 1987;144:229e231.

216. Amsterdam JD, Maislin G, Winokur A, Kling M, Gold P. Pitui-tary and adrenocortical responses to the ovine corticotropinreleasing hormone in depressed patients and healthy volun-teers. Arch Gen Psychiatry. 1987;44:775e781.

217. Merali Z, Du L, Hrdina P, Palkovits M, Faludi G, Poulter MO,Anisman H. Dysregulation in the suicide brain: mRNAexpression of corticotropin-releasing hormone receptors andGABA(A) receptor subunits in frontal cortical brain region. JNeurosci. 2004;24:1478e1485.

218. Nemeroff CB, Owens MJ, Bissette G, Andorn AC, Stanley M.Reduced corticotropin releasing factor binding sites in thefrontal cortex of suicide victims. Arch Gen Psychiatry. 1988;45:577e579.

CTION AND BEHAVIOR

Page 20: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

15. NEUROPEPTIDE REGULATION OF STRESS-INDUCED BEHAVIOR: INSIGHTS FROM THE CRF/UROCORTIN FAMILY374

219. Austin MC, Janosky JE, Murphy HA. Increased corticotropin-releasing hormone immunoreactivity in monoamine-containingpontine nuclei of depressed suicide men. Mol Psychiatry.2003;8:324e332.

220. Merali Z, Kent P, Du L, Hrdina P, Palkovits M, Faludi G,Poulter MO, Bedard T, Anisman H. Corticotropin-releasinghormone, arginine vasopressin, gastrin-releasing peptide, andneuromedin B alterations in stress-relevant brain regions ofsuicides and control subjects. Biol Psychiatry. 2006;59:594e602.

221. Raadsheer FC, Hoogendijk WJ, Stam FC, Tilders FJ, Swaab DF.Increased numbers of corticotropin-releasing hormone express-ing neurons in the hypothalamic paraventricular nucleus ofdepressed patients. Neuroendocrinology. 1994;60:436e444.

222. Bissette G, Klimek V, Pan J, Stockmeier C, Ordway G. Elevatedconcentrations of CRF in the locus coeruleus of depressedsubjects. Neuropsychopharmacology. 2003;28:1328e1335.

223. Wang SS, Kamphuis W, Huitinga I, Zhou JN, Swaab DF. Geneexpression analysis in the human hypothalamus in depressionby laser microdissection and real-time PCR: the presence ofmultiple receptor imbalances. Mol Psychiatry. 2008;13:786e799.741.

224. Kozicz T, Tilburg-Ouwens D, Faludi G, Palkovits M, Roubos E.Gender-related urocortin 1 and brain-derived neurotrophicfactor expression in the adult humanmidbrain of suicide victimswith major depression. Neuroscience. 2008;152:1015e1023.

225. Kang HJ, Adams DH, Simen A, Simen BB, Rajkowska G,Stockmeier CA, Overholser JC, Meltzer HY, Jurjus GJ,Konick LC, Newton SS, Duman RS. Gene expression profiling inpostmortem prefrontal cortex of major depressive disorder. JNeurosci. 2007;27:13329e13340.

226. Binder EB, Owens MJ, Liu W, Deveau TC, Rush AJ, Trivedi MH,Fava M, Bradley B, Ressler KJ, Nemeroff CB. Association ofpolymorphisms in genes regulating the corticotropin-releasingfactor system with antidepressant treatment response. Arch Gen

Psychiatry. 67, 369-379.227. Wasserman D, Wasserman J, Sokolowski M. Genetics of HPA-

axis, depression and suicidality. Eur Psychiatry. 25, 278-280.228. Licinio J, O’Kirwan F, Irizarry K, Merriman B, Thakur S,

Jepson R, Lake S, Tantisira KG, Weiss ST, Wong ML. Associationof a corticotropin-releasing hormone receptor 1 haplotype andantidepressant treatment response in Mexican-Americans. Mol

Psychiatry. 2004;9:1075e1082.229. Liu Z, Zhu F, Wang G, Xiao Z, Tang J, Liu W, Wang H, Liu H,

Wang X, Wu Y, Cao Z, Li W. Association study of corticotropin-releasing hormone receptor1 gene polymorphisms and antide-pressant response in major depressive disorders. Neurosci Lett.

2007;414:155e158.230. Liu Z, Zhu F, Wang G, Xiao Z, Wang H, Tang J, Wang X, Qiu D,

Liu W, Cao Z, Li W. Association of corticotropin-releasinghormone receptor1 gene SNP and haplotype with majordepression. Neurosci Lett. 2006;404:358e362.

231. Wasserman D, Sokolowski M, Rozanov V, Wasserman J. TheCRHR1 gene: a marker for suicidality in depressed malesexposed to low stress. Genes Brain Behav. 2008;7:14e19.

232. Wasserman D, Wasserman J, Rozanov V, Sokolowski M.Depression in suicidal males: genetic risk variants in the CRHR1gene. Genes Brain Behav. 2009;8:72e79.

233. Papiol S, Arias B, Gasto C, Gutierrez B, Catalan R, Fananas L.Genetic variability at HPA axis in major depression and clinicalresponse to antidepressant treatment. J Affect Disord. 2007;104:83e90.

234. Bradley RG, Binder EB, Epstein MP, Tang Y, Nair HP, Liu W,Gillespie CF, Berg T, Evces M, Newport DJ, Stowe ZN,Heim CM, Nemeroff CB, Schwartz A, Cubells JF, Ressler KJ.Influence of child abuse on adult depression: moderation by the

III. HORMONES, BRAIN FUN

corticotropin-releasing hormone receptor gene. Arch Gen

Psychiatry. 2008;65:190e200.235. De Luca V, Tharmalingam S, Kennedy JL. Association study

between the corticotropin-releasing hormone receptor 2 geneand suicidality in bipolar disorder. Eur Psychiatry. 2007;22:282e287.

236. Held K, Kunzel H, Ising M, Schmid DA, Zobel A, Murck H,Holsboer F, Steiger A. Treatment with the CRH1-receptor-antagonist R121919 improves sleep-EEG in patients withdepression. J Psychiatr Res. 2004;38:129e136.

237. Kunzel HE, Zobel AW, Nickel T, Ackl N, Uhr M, Sonntag A,Ising M, Holsboer F. Treatment of depression with the CRH-1-receptor antagonist R121919: endocrine changes and sideeffects. J Psychiatr Res. 2003;37:525e533.

238. Zobel AW, Nickel T, Kunzel HE, Ackl N, Sonntag A, Ising M,Holsboer F. Effects of the high-affinity corticotropin-releasinghormone receptor 1 antagonist R121919 in major depression: thefirst 20 patients treated. J Psychiatr Res. 2000;34:171e181.

239. Mathew SJ, Price RB, Charney DS. Recent advances in theneurobiology of anxiety disorders: implications for novel ther-apeutics. Am J Med Genet C Semin Med Genet. 2008;148C:89e98.

240. Yehuda R, Giller EL, Southwick SM, Lowy MT, Mason JW.Hypothalamicepituitaryeadrenal dysfunction in posttraumaticstress disorder. Biol Psychiatry. 1991;30:1031e1048.

241. de Kloet CS, Vermetten E, Geuze E, Kavelaars A, Heijnen CJ,Westenberg HG. Assessment of HPA-axis function in post-traumatic stress disorder: pharmacological and non-pharmaco-logical challenge tests, a review. J Psychiatr Res. 2006;40:550e567.

242. Erhardt A, Ising M, Unschuld PG, Kern N, Lucae S, Putz B,Uhr M, Binder EB, Holsboer F, Keck ME. Regulation of thehypothalamic-pituitary-adrenocortical system in patients withpanic disorder. Neuropsychopharmacology. 2006;31:2515e2522.

243. Schreiber W, Lauer CJ, Krumrey K, Holsboer F, Krieg JC. Dys-regulation of the hypothalamic-pituitary-adrenocortical systemin panic disorder. Neuropsychopharmacology. 1996;15:7e15.

244. Bremner JD, Licinio J, Darnell A, Krystal JH, Owens MJ,Southwick SM, Nemeroff CB, Charney DS. Elevated CSF corti-cotropin-releasing factor concentrations in posttraumatic stressdisorder. Am J Psychiatry. 1997;154:624e629.

245. Baker DG, West SA, Nicholson WE, Ekhator NN, Kasckow JW,Hill KK, Bruce AB, Orth DN, Geracioti Jr TD. Serial CSF corti-cotropin-releasing hormone levels and adrenocortical activity incombat veterans with posttraumatic stress disorder. Am J

Psychiatry. 1999;156:585e588.246. Sautter FJ, Bissette G, Wiley J, Manguno-Mire G,

Schoenbachler B, Myers L, Johnson JE, Cerbone A, Malaspina D.Corticotropin-releasing factor in posttraumatic stress disorder(PTSD) with secondary psychotic symptoms, nonpsychoticPTSD, and healthy control subjects. Biol Psychiatry. 2003;54:1382e1388.

247. Banki CM, Karmacsi L, Bissette G, Nemeroff CB. Cerebrospinalfluid neuropeptides in mood disorder and dementia. J AffectDisord. 1992;25:39e45.

248. Fossey MD, Lydiard RB, Ballenger JC, Laraia MT, Bissette G,Nemeroff CB. Cerebrospinal fluid corticotropin-releasing factorconcentrations in patients with anxiety disorders and normalcomparison subjects. Biol Psychiatry. 1996;39:703e707.

249. Jolkkonen J, Lepola U, Bissette G, Nemeroff C, Riekkinen P. CSFcorticotropin-releasing factor is not affected in panic disorder.Biol Psychiatry. 1993;33:136e138.

250. Chappell P, Leckman J, Goodman W, Bissette G, Pauls D,Anderson G, Riddle M, Scahill L, McDougle C, Cohen D.Elevated cerebrospinal fluid corticotropin-releasing factor inTourette’s syndrome: comparison to obsessive compulsivedisorder and normal controls. Biol Psychiatry. 1996;39:776e783.

CTION AND BEHAVIOR

Page 21: Chapter 15 - Neuropeptide Regulation of Stress-Induced Behavior: … · 2015. 8. 25. · related emotional disorders such as anxiety, anorexia nervosa and melancholic depression.8e10

REFERENCES 375

251. Smoller JW, Rosenbaum JF, Biederman J, Kennedy J, Dai D,Racette SR, Laird NM, Kagan J, Snidman N, Hirshfeld-Becker D,Tsuang MT, Sklar PB, Slaugenhaupt SA. Association of a geneticmarker at the corticotropin-releasing hormone locus withbehavioral inhibition. Biol Psychiatry. 2003;54:1376e1381.

252. Smoller JW, Yamaki LH, Fagerness JA, Biederman J, Racette S,Laird NM, Kagan J, Snidman N, Faraone SV, Hirshfeld-Becker D, Tsuang MT, Slaugenhaupt SA, Rosenbaum JF,Sklar PB. The corticotropin-releasing hormone gene andbehavioral inhibition in children at risk for panic disorder. BiolPsychiatry. 2005;57:1485e1492.

253. Keck ME, Kern N, Erhardt A, Unschuld PG, Ising M,Salyakina D, Muller MB, Knorr CC, Lieb R, Hohoff C,Krakowitzky P, Maier W, Bandelow B, Fritze J, Deckert J,Holsboer F, Muller-Myhsok B, Binder EB. Combined effects ofexonic polymorphisms in CRHR1 and AVPR1B genes in a case/control study for panic disorder. Am J Med Genet B Neuro-

psychiatr Genet. 2008;147B:1196e1204.254. Cryan JF, Holmes A. The ascent of mouse: advances in model-

ling human depression and anxiety. Nat Rev Drug Discov.

2005;4:775e790.255. Wong ML, Licinio J. From monoamines to genomic targets:

a paradigm shift for drug discovery in depression. Nat Rev

Drug. Discov. 2004;3:136e151.

III. HORMONES, BRAIN FUN

256. Krishnan V, Nestler EJ. The molecular neurobiology of depres-sion. Nature. 2008;455:894e902.

257. Francis DD, Szegda K, Campbell G, Martin WD, Insel TR.Epigenetic sources of behavioral differences in mice. Nat Neu-

rosci. 2003;6:445e446.258. McEwen BS. Physiology and neurobiology of stress and

adaptation: central role of the brain. Physiol Rev. 2007;87:873e904.

259. Ulrich-Lai YM, Herman JP. Neural regulation of endocrineand autonomic stress responses. Nat Rev Neurosci. 2009;10:397e409.

260. Lupien SJ, McEwen BS, Gunnar MR, Heim C. Effects of stressthroughout the lifespan on the brain, behaviour and cognition.Nat Rev Neurosci. 2009;10:434e445.

261. McEwen BS. Stress, sex, and neural adaptation to a changingenvironment: mechanisms of neuronal remodeling. Ann NY

Acad Sci. 1204 Suppl, E38-59.262. Holsboer F, Ising M. Stress hormone regulation: biological role

and translation into therapy. Annu Rev Psychol. 61, 81-109, C1-11.263. Bale TL. Sensitivity to stress: dysregulation of CRF pathways

and disease development. Horm Behav. 2005;48:1e10.264. Zorrilla EP, Koob GF. The therapeutic potential of CRF1

antagonists for anxiety. Expert Opin Investig Drugs. 2004;13:799e828.

CTION AND BEHAVIOR