Você está na página 1de 10

Physiology & Behavior 141 (2015) 154163

Contents lists available at ScienceDirect

Physiology & Behavior


journal homepage: www.elsevier.com/locate/phb

Oxytocin mechanisms of stress response and aggression in a


territorial nch
James L. Goodson, Sara E. Schrock, Marcy A. Kingsbury
Department of Biology, Indiana University, Bloomington, IN 47405 USA

H I G H L I G H T S
Oxytocin receptor antagonism reduces aggression in a territorial nch.
Hypothalamic oxytocin cell groups respond to stressors (subjugation and pursuit).
Oxytocin inhibition of the HPA axis may be permissive for aggression.

a r t i c l e

i n f o

Article history:
Received 16 July 2014
Received in revised form 12 January 2015
Accepted 13 January 2015
Available online 14 January 2015
Keywords:
Oxytocin
Mesotocin
Aggression
Corticosterone
Stress Coping
Defense
Songbird
Violet-eared waxbill

a b s t r a c t
All jawed vertebrates produce a form of oxytocin (OT), and in birds, mammals and sh, OT is strongly associated
with afliation. However, remarkably few data are available on the roles of OT and OT receptors (OTRs) in aggression. Because OT and OTRs exert anxiolytic effects in mammals (although context-specic) and modulate stress
coping, we hypothesized that OTR activation is at least permissive for territorial aggression. Indeed, we nd that
peripheral injections of an OTR antagonist signicantly reduce malemale and femalefemale aggression in a
highly territorial nch. This nding suggests the hypothesis that aggression is accompanied by an increase in
transcriptional (Fos) activity of OT neurons, but contrary to this hypothesis, we nd that dominant male residents
do not elevate OT-Fos colocalization following an aggressive encounter and that OT-Fos colocalization in the
preoptic area and hypothalamus correlates negatively with aggression. Furthermore, OT-Fos colocalization increases dramatically in males that were aggressively subjugated or pursued by a human hand, likely reecting
OT modulation of stress response. Because OT inhibits the hypothalamopituitaryadrenal axis, the antagonist
effects may reect the fact that aggressive birds and mammals tend to be hyporesponsive to stress. If this is correct, then 1) the observed effects of OTR antagonism may reect alterations in corticosterone feedback to the
brain rather than centrally mediated OTR effects, and 2) the negative correlation between OT-Fos colocalization
and aggression may reect the fact that more aggressive, stress hyporesponsive males require less inhibition of
the hypothalamopituitaryadrenal axis than do less aggressive males, despite the requirement of that inhibition
for the normal display of aggression.
2015 Elsevier Inc. All rights reserved.

1. Introduction
The modulation of social cognition, afliation and anxiety by the
neuropeptides oxytocin (OT)1 and vasopressin (VP) has become a

Corresponding author.
E-mail address: mk24@indiana.edu (M.A. Kingsbury).
1
Vertebrates express a variety of OT forms, and although the canonical form in mammals is Leu8-OT, some mammals also express Pro8-OT or Ile8-OT (mesotocin) [13]. This
latter form is expressed in birds and other non-mammalian tetrapods. Similarly, multiple
forms of VP have been identied, including the canonical Arg8-VP form found in most (but
not all) mammals, and Ile3-VP (vasotocin), which is found in non-mammalian vertebrates
[1,3]. Thus for clarity, we will simply refer to all OT forms as OT and all VP forms as VP
(following other recent precedents [3,4]). We will also refer to homologous oxytocic receptors (of which one appears to be present in all vertebrates) as OTRs, following the nomenclature of Ocampo Daza et al. [5], and also Yamaguchi et al. [6].

http://dx.doi.org/10.1016/j.physbeh.2015.01.016
0031-9384/ 2015 Elsevier Inc. All rights reserved.

major eld of inquiry, with more than 100 relevant papers being published annually [7]. This literature supports the general view that OT reduces anxiety and promotes many aspects of afliation in mammals
[810]. OT also facilitates social approach in goldsh (Carassius auratus)
[11], and similarly, we have found that ocking behavior in zebra
nches (Taeniopygia guttata) is promoted by both central OTR activation
[12] and OT production in the paraventricular nucleus of the hypothalamus (PVN; a female-specic effect) [4], suggesting an evolutionarily
deep history of OT effects on sociality. OT immunoreactivity uctuates
in relation to seasonal ocking in sparrows [13], consistent with these
ndings in nches. However, OT and OT receptors (OTRs) also inuence
many other aspects of physiology and behavior, including appetite, immune function, glucocorticoid secretion and thermoregulation [1417].
Of particular relevance here, OT promotes maternal aggression in selected rat lines ([18]; but see [19]), although it is not clear that OT effects

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

in this context will be observed in other contexts of aggression. In fact,


exogenous OT is capable of reducing aggression in male rats, despite
the fact that OTR antagonism is largely without effects [20,21]. However, it must be considered that endogenous OT may modulate behavior
via heterologous binding to V1a receptors (V1aRs), which are known
to mediate at least some of OT's effects (e.g., [22,23]).
In contrast to the voluminous data on the afliation effects of OT, relatively little is known about the role(s) that endogenous OT plays in
same-sex offensive aggression. OT effects on intrasexual aggression
have been primarily examined within a developmental framework
(e.g., using neonatal OT injections in prairie voles, Microtus ochrogaster,
and Mandarin voles, Lasiopodomys mandarinus [24,25] and intranasal
OT administrations in pigs [26]) or using knockout mice that lack OT
or the OTR [22,27]. The data obtained using these methods has provided
consistent evidence that OT facilitates aggression, contrary to the
popular conception of OT as a prosocial peptide [2830]. Regardless,
such manipulations do not directly address the inuence of OT in
adult animals, since these developmental manipulations likely act
neonatally to organize brain and behavior [24,25].
Remarkably, only one study has clearly demonstrated that
endogenous OT modulates resident-intruder aggression in adults. This
experiment shows that OT infusions into the preoptic area-anterior hypothalamus (POA-AH) decrease resident-intruder aggression in female
Syrian hamsters (Mesocricetus auratus), and more importantly, that OTR
antagonism facilitates aggression [31]. However, broad generalization
from this nding to males, and to other brain areas and species is difcult, because 1) AH infusions of VP exert opposing effects on aggression
in male and female hamsters ([32]; and thus OT may likewise exert sexspecic effects, particularly given that OT-VP receptors tend to be promiscuous; e.g., [3335]); 2) OT effects on maternal aggression are mediated in areas of the brain outside of the POA-AH, including the amygdala
[18,19]; and 3) the neural distribution of OTRs is species-specic [12,33,
3639].
Importantly, because OT is important for anxiolysis, fear reduction
and stress coping [4042], and because aggressive birds, sh, and
some rodents tend to be hyporesponsive to stress (dened primarily
in terms of glucocorticoid secretion [4347]), we hypothesized that
OTR activation is at a minimum permissive for territorial aggression,
even if it does not actively promote it (but note that stress reactivity
likely interacts with coping style to modulate aggression, rather than
having unitary effects; [48,49]).
One caveat is that endogenous OT does not exert anxiolytic effects
across all contexts, but rather in specic contexts that are otherwise associated with OT release, such as lactation and parturition [50]. In fact,
recent antisense experiments in zebra nches (conducted after the
completion of the present experiments) demonstrate that OT neurons
of the paraventricular hypothalamus (PVN) do not modulate anxiety
when subjects are tested in a nonsocial context, and actually promote
passive coping behavior [4] (note that although these more recent
data could not inform our hypotheses for the experiments presented
here, they are nonetheless important to consider in relation to the
results, and will be revisited in the Discussion section). A second consideration is that OT inhibits basal and stress-induced activity of the
hypothalamopituitaryadrenal (HPA) axis in a manner that appears
to be uncoupled from the central modulation of anxiety [51], at least
in rodents.
The species under investigation here, the violet-eared waxbill
(Estrildidae: Uraeginthus granatina) is exceptionally territorial [52,53],
and thus we hypothesized that aggressive interactions in this species
will drive endogenous release of OT, which could thus modulate both
the HPA axis and central anxiety processes (presuming socially- and/
or stress-mediated OT release; see [50] for a consideration of contextspecic effects on anxiety), thereby facilitating aggression. Other
known effects of OT may also serve to facilitate aggressive response,
such as enhanced sensory processing of social stimuli, and modulation
of autonomic function (reviews: [7,54]).

155

Despite the potential for the independent modulation of central and


peripheral processes [55], it is also possible that the central and peripheral effects of OT (e.g., sensory and autonomic effects) are coupled to
some extent. This is because the parvocellular OT population of the
PVN that projects to the anterior pituitary also projects centrally, including the autonomic brainstem; and magnocellular OT populations that
project to the posterior pituitary also release peptide centrally from
axons, soma and dendrites (reviews: [7,54,56]).
Thus, in order to examine the combined central and peripheral
effect of oxytocic signaling on aggression, we here quantied residentintruder aggression following peripheral injections of an OTR antagonist
or vehicle in male and female violet-eared waxbills. In a second experiment we then quantied the Fos responses of OT neurons to handling
(control), pursuit by a human hand, aggressive subjugation, or aggressive domination. OT-Fos colocalization was quantied for the two
largest hypothalamic populations, which lie in the PVN and supraoptic
nucleus (SON), as well as within the smaller OT populations of the
medial preoptic nucleus (POM; magnocellular) and AH (embedded in
the hypothalamo-pituitary tract; parvocellular).
2. Materials and methods
2.1. Subjects
Captive-bred violet-eared waxbills (n = 9 females and 24 males)
were individually housed in cages 61 cm W 36 cm D 43 cm H on
a 14L:10D photoperiod and provided nch mix and water ad libitum.
Experiments were conducted under non-breeding conditions and subjects were separated from opposite-sex partners for a minimum of
two weeks prior to testing. Violet-eared waxbills are generally encountered as singletons or as pairs in the wild (unless dependent young are
present) ([53]; J.L. Goodson, pers. obs.), and thus the period of isolation
is not expected to produce behavioral abnormalities. Violet-eared waxbills are aggressive year-round [52]. Experiments were performed in
compliance with federal and institutional guidelines and were approved
by the Institutional Animal Care and Use Committee of Indiana
University.
2.2. OTR antagonist experiment
2.2.1. Behavioral screenings
Same-sex pairs of violet-eared waxbills were screened for aggression in short resident-intruder assays (most less than 1 min; none
more than 2 min; terminated as soon as an obvious dominance relationship appeared) in order to select subordinate (intruder) and dominant
(resident) subjects. Due to the limited number of birds available, most
birds were used as both stimuli and subjects, with at least 1 week between tests. This was possible because most birds could dominate an
intruder in their home cage, but were subordinated as intruders. Only
subjects who dominated same-sex intruders in their own home cage
were used as subjects.
2.2.2. Injections
Thirty minutes prior to testing, subjects (9 females and 10
males) were injected into the inguinal leg fold with either
0.05 cm 3 saline vehicle or vehicle containing 5 g OTR antagonist
(desGly-NH 2 ,d(CH 2 ) 5 [Tyr(Me) 2 ,Thr 4 ] ornithine vasotocin) in a
counterbalanced, repeated-measures design with 2 days allowed between tests. This dose has been used in previous studies and effects
have been replicated using a much lower dose centrally [12]. Importantly, effects of systemic administration of the OTR antagonist do not
produce general behavioral alterations. For instance, subjects do not
decrease approach to conspecics [12], and despite potent effects on
nest-building behavior in female zebra nches, the behavior of males
is not altered [57], suggesting that effects are highly specic. Another
important consideration is that an iodinated form of this same

156

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

antagonist yields binding pattern that is closely matched to OTR (VT3)


mRNA expression in zebra nches and sparrows, but does not match
mRNA expression patterns for other OT-VP receptor types [33,38].
2.2.3. Resident-intruder aggression
A same-sex intruder was introduced into the subject's home cage
and observations were conducted for 7 min or until 40 displacements
were observed, in which case behaviors were prorated. A variety of observations suggest that prorating in this species provides an accurate estimate of behavior [58,59]. We recorded displacements, threats, pecks,
and agonistic calls.
2.3. Fos experiment
2.3.1. Behavior
The Fos experiment was conducted using males only. Pairs of male
violet-eared waxbills were screened for aggression in short residentintruder assays as described above and at least one month was allowed
between screenings and testing. Based on behavior in these screenings,
we were able to create treatment groups that did not differ in their
levels of aggression, based on their behavior as residents during the
screenings. Of the 20 males used for the Fos experiment, 10 had been
used for the OTR antagonist study described above (including all subjects in the Dominant condition) and the remaining 10 had been tested
in an identical paradigm, but following peripheral injection of a dopamine antagonist.
Prior to testing and brain collection, we acclimated subjects to a
sound isolation booth (3 h per day for 2 days), and on the third day
left the subjects in the booth overnight. Tests were conducted the following morning. Subjects were assigned to one of four conditions
(n = 5 each): Control, Subordinate, Dominant or Defense. Dominant
and Subordinate subjects were exposed to a resident-intruder encounter as described above, with a xed test duration of 7 min (i.e., no test
termination or prorating). Because Subordinate subjects were handled
twice (to introduce them to the resident's cage and to remove them),
other subjects were likewise handled twice. Subjects in the Defense
condition were pursued by a human hand 40 times over a period of
7 min. Birds were perfused 90 min after the start of testing.
2.4. Immunouorescent labeling and quantication
Subjects were intracardially perfused with 0.1 M phosphatebuffered saline (PBS; pH 7.4) followed by 4% paraformaldehyde. Brains
were postxed overnight, sunk for two days in 30% sucrose, and
sectioned on a cryostat into three series of 40 m sections. We followed
standard lab protocols for the immunouorescent labeling of OT and Fos
using free-oating sections [4,60]. These antibodies were labeled using
secondaries conjugated to Alexa Fluor 488 (green) and 594 (red),
respectively (Invitrogen, Carlsbad, CA). We used a rabbit anti-Fos antibody (Santa Cruz Biotechnology, Santa Cruz, CA; used at 1:1000) that
has been extensively used and validated in birds (e.g., [61]), and a guinea pig anti-OT antibody (Bachem, Torrance, CA; used at 1:1000) from a
lot that we have previously shown does not cross-react with arginine
vasotocin (Ile3-vasopressin) in sh ([62]; please note that this is a
polyclonal antibody and that we nd that newer lots raised from a different guinea pig do indeed cross-react with Ile3-VP [vasotocin]). In addition, we nd that preabsorption of this antibody with 10 M Ile8-OT
(mesotocin) eliminates labeling, whereas identical preabsorptions
with vasotocin do not.
Photomicrographs of sections containing OT populations in the
POM, SON, PVN and AH were shot on a Zeiss AxioImager microscope
outtted with a z-drive and optical dissector (Apotome; Carl Zeiss Inc.,
Gttingen, Germany). Cell counts were conducted using Adobe
Photoshop CS5 (Adobe Systems, Seattle, WA) and Image J (National
Institutes of Health, Bethesda, MD) as previously described [63,64].

2.5. Statistical analyses


Behavioral effects of the OTR antagonist were analyzed using
repeated-measures ANOVA with Sex as a between-subjects factor and
Treatment as the repeated measure. OT-Fos double-labeling was
analyzed using one-way ANOVA with the single factor of Condition
(Control, Subordinate, Dominant, Defense). Other data were analyzed
using linear regressions, as described below. Violet-eared waxbills are
among the most difcult nches to breed in captivity [53] and thus
our n's for these experiments are relatively modest. However, the extraordinary robustness of aggressive behavior in this species (e.g., [59,
65]) allows for excellent behavioral quantication, which should yield
substantial reductions in measurement error.
3. Results
3.1. OTR antagonism decreases aggression in male and female waxbills
As shown in Fig. 1A, displacements of the intruder were signicantly
reduced by OTR antagonist injections (main effect of Treatment:
F(1,17) = 5.38, p = 0.03, repeated-measures ANOVA) as was the total
number of aggressive behaviors (Fig. 1B; main effect of Treatment:
F(1,17) = 4.68, p = 0.04). Other than displacements, only threats were
exhibited with sufcient frequency for separate analysis, but antagonist
treatments did not signicantly alter this behavior (main effect of
Treatment: F(1,17) = 1.23, p N 0.10). Males were signicantly more
aggressive than females (for total aggression, main effect of Sex:
F(1,17) = 5.16, p = 0.04), although females were still very aggressive, averaging approximately 32 displacements and 60 aggressive behaviors
following injections of vehicle (Fig. 1). No Sex*Treatment interactions
were observed, and the latency to displace the intruder was not affected
by OTR antagonism (all p N 0.10).
3.2. OT neurons are Fos-activated by stressors
Representative co-labeling of OT and Fos is shown in Fig. 2. As
shown in Fig. 3A, OT-Fos colocalization in the POM (i.e., the percent of
POM OT-immunoreactive, ir, neurons that are Fos-ir+) is substantially greater in Defense and Subordinate subjects than in Control and
Dominant subjects. A similar pattern is observed in the SON (Fig. 3B),
although this does not reach signicance. In the PVN, only Defense subjects exhibit signicant elevations of OT-Fos colocalization (Fig. 3C).
Finally, OT-Fos colocalization in the AH is signicantly elevated above
Control levels in Defense and Subordinate subjects, as in the POM, but
Dominant subjects do not differ signicantly from any other group
(Fig. 3D).
3.3. OT neuron activation correlates with aggression
Based on the behavioral effects of OTR antagonist injections, we
predicted that Dominant subjects would show the greatest OT-Fos
colocalization. As just described, this is not the case. Rather, only
Defense and Subordinate subjects exhibit signicant increases in
colocalization above control levels. However, the Control baseline
level of colocalization is very high, with comparable levels in Dominant
subjects (~1535%; Fig. 3), and this colocalization may reect constitutive neuromodulation, phasic response to handling (all subjects were
handled twice), and/or phasic response to the testing environment. All
of these factors apply equally to the Control and Dominant subjects,
and thus we hypothesized that aggression is positively correlated with
the ability to cope, either constitutively or in response to phasic
stressors. This led to the prediction that OT neuromodulation would
correlate positively with aggression in Dominant subjects.
In considering such phenotypic variation in OT neuromodulation,
we considered that 50% OT-Fos colocalization would presumably yield
less neuromodulation in subjects that had relatively few OT neurons

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

157

Fig. 1. Aggressive displacements (A) and total numbers of aggressive behaviors (B) exhibited by male and female violet-eared waxbills in 7-min resident-intruder tests. Subjects were
tested in a within-subjects design following injections of saline vehicle or an OTR antagonist (OTA). Data are shown as means S.E.M. *p b 0.05, main effect of Treatment, Treatment*Sex
repeated-measures ANOVA; n = 9 females and 10 males.

as opposed to relatively many OT neurons. We therefore present our


analyses here based upon total numbers of OT-ir neurons expressing
Fos. This yields multiple signicant results (described below), which
comparable analyses based on the percent of OT-ir neurons that express
Fos do not (all p N 0.10).
Contrary to our predictions, numbers of OT-Fos co-labeled neurons
actually correlate negatively with aggression. As with the group data
shown in Fig. 3, all four OT cell groups tend to follow the same pattern,
although the effect is signicant only for the PVN (Fig. 4A). However, because all cell groups tend to follow the same pattern, we conducted an
additional analysis with all cell groups combined, and again nd that
OT-Fos co-labeling correlates negatively with aggression (Fig. 4B).

level of stress entailed in the two experiments was likely very different.
In the antagonist study, animals were handled once for a peripheral injection that was given 30 min prior to resident-intruder testing. In contrast, subjects in the Fos study were acclimated and housed in small
sound booths prior to testing, and were handled both before and after
testing. An examination of displacements exhibited by dominant animals during resident-intruder testing across both experiments does
suggest that the Fos testing environment was likely more stressful, in
that dominant birds exhibited signicantly less displacements when
tested in the sound booths, relative to testing in the home environment
(Fig. 5).
4. Discussion

3.4. Aggression by dominant animals varies between test environment


4.1. Paradoxical effects of OTR antagonism and OT neuronal response
Based on the paradoxical results from the antagonist and Fos studies,
and the observation that the activation of a stress response likely explains the results of the Fos study, we further examined whether the

The present experiments provide novel evidence that endogenous


activation of OTRs promotes or is permissive for resident-intruder

Fig. 2. Representative OT-Fos double-labeling in the PVN (A), SON (B), AH (C) and POM (D) of subjects in the Defense group. Arrows highlight double-labeled neurons. The box in panel A
corresponds to the location of the inset. Scale bars = 50 mm for panels AD; 10 mm for the panel A inset. Abbreviations: AH, anterior hypothalamus; ot, optic tract; POM, medial preoptic
nucleus; PVN, paraventricular nucleus; SON, supraoptic nucleus; vaf, ventral amygdalofugal pathway (occipitomesencephalic tract).

158

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

Fig. 4. Total aggression exhibited by dominant male violet-eared waxbills correlates


negatively with the number of OT-Fos double labeled cells in the PVN (A) and across all
hypothalamic cell groups (B; log-transformed for normality).

Fig. 3. OT-Fos colocalization in male violet-eared waxbills following handled control conditions (Control), 40 pursuits by a human hand (Defense), or a resident-intruder encounter in which the subject was dominant (Dominant) or subordinate (Subordinate). Data are
shown as means S.E.M. for the POM (A), SON (B), PVN (C) and AH (D). Different letters
above the error bars denote signicant pairwise comparisons following signicant ANOVA
(p b 0.05); n = 5 per condition. Abbreviations: AH, anterior hypothalamus; POM, medial
preoptic nucleus; PVN, paraventricular nucleus; SON, supraoptic nucleus.

aggression. We rst show that antagonism of OTRs reduces aggression,


yielding the prediction that aggression is positively associated with the
activation of OT neurons. However, the results for OT-Fos colocalization
are inconsistent with that prediction. Signicant elevations in
colocalization are observed only for Subordinate and Defense subjects,
indicating that OT neurons are strongly activated by stressors. Further
inconsistent with our expectations, we nd that the number of cells
that co-label for OT and Fos correlates negatively with aggression in
Dominant subjects, despite the fact that Dominant subjects did not exhibit an increase in OT-Fos colocalization above the level of Control subjects. This nding may reect a baseline (constitutive) modulation of
aggression-related processes by OT neurons. Alternatively, the test conditions for the Fos experiment (isolation in the sound booths, being handled twice and observed) may have induced a phasic, stress-related
increase in OT-Fos colocalization in all subjects, including Dominant
subjects and Controls.
Indeed, as described below, the Fos testing environment was likely
much more stressful than the subjects' home environment, where the
antagonist experiment was conducted. Thus, it is likely that OT neurons

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

Fig. 5. Evidence that levels of stress varied between the antagonist (OTA) experiment and
the Fos experiment (as assessed in a group of males that participated in both the OTA experiment and the Dominant condition of the Fos experiment). Subjects in the antagonist
experiment had been housed for many months in the same room and were handled
only once for injection 30 min prior to testing. For the Fos experiment, subjects were acclimated for 3 h per day for 2 days in a sound attenuated booth, and then remained in the
booth overnight prior to the Fos experiment. Subjects were handled immediately before
introduction of the intruder and immediately afterwards. As shown here, displacements
were signicantly suppressed in the Fos experiment relative to the OTA experiment (based
on control saline data). Data are shown as means S.E.M. *p b 0.05, paired t-test.

in all subjects exhibited some amount of stress-induced Fos response. If


this is correct, and if more aggressive violet-eared waxbills are hyporesponsive to stress (as in other species [4347]), then stress-induced
activation of OT neurons should correlate negatively with aggression,
which is what we found. And importantly, this relationship can be expected even if OT inhibition of the HPA axis is permissive for aggression.
Thus, this interpretation of results suggests that endogenous activation
of OTRs is necessary for normal levels of aggression, even if the most aggressive animals exhibit less stress-induced activation of OT neurons.
As expounded upon below, we believe that this interpretation provides the most parsimonious explanation of our results. However, we
must consider that the avian OTR binds both the avian OT and VP
forms (mesotocin and vasotocin) with high afnity [33], and thus it is
possible that the behavioral effects of OTR antagonism reect endogenous actions of VP and not OT neurons. Thus, we rst address this
possibility before focusing specically on OT.
4.2. Are OTR-mediated effects driven by endogenous OT or VP?
The present experiments were designed to examine the relationships between the behavioral effects of OTR antagonism and OT
neuronal activation. However, because avian OTRs bind VP with a high
afnity ([33]; also see [38]), we must also consider the possibility that
VP is mediating some of the observed effects. If this were the case, the
decreased aggression observed here following OTR antagonism would
suggest that VP (via binding to OTRs) increases aggression. However,
virtually all lines of evidence suggest that VP circuits inhibit territorial
aggression in songbirds. For instance, in both violet-eared waxbills
and territorial sparrows, intraseptal VP infusions decrease residentintruder aggression [58,66]. Conversely, central administrations of a
V1aR antagonist increase aggression in male zebra nches after they
have pair bonded and are defending nests [67]. In addition, Fos activation of PVN VP neurons is negatively related to aggression in sparrows
and mice [68,69], and VP neurons in the medial bed nucleus of the
stria terminalis (BSTm) show no response to socially aversive and/or
aggressive interactions in nches, chickens, or mice [6870]. In fact,
knockdown of VP production in the BSTm reduces afliation and profoundly increases aggression in male zebra nches [71]. Consistent
with this observation, the density of VP-ir elements in the BSTm
correlates negatively with aggression in territorial sparrows, and differentiates species that are more or less aggressive [13]. Relative to

159

aggressive (short attack latency) mice, less aggressive mice also exhibit
more VP neurons in the BSTm and greater VP ber densities in the lateral septum [72].
However, endogenous VP does act within the AH to promote aggression in male Syrian hamsters (M. auratus) that have been isolated and
trained as ghters ([73]; for discussion see [74]), and also in male prairie
voles following mating [75]. Both of these effects are associated with
experience-dependent up-regulation of V1aRs, and notably, no such effects are observed in male hamsters that have been socially housed [76].
Furthermore, endogenous VP inhibits aggression in the AH of female
hamsters, and VP-Fos colocalization in the AH and other VP cell groups
correlates positively with bites received by subordinate male mice [69].
In fact, no VP cell group in dominant male mice exhibits a positive Fos
response to aggressive interactions [69].
To a great extent, the negative relationship between aggression and
the Fos activation of PVN VP neurons in sparrows and mice [68,69] is
consistent with the idea that more aggressive males show lower socially
induced activation of the HPA than do less aggressive males, at least in
some species of birds and rodents (review: [49]). This is because VP
released into the anterior pituitary (presumably derived from the
PVN) is a potent secretagogue for adrenocorticotropic hormone
(ACTH), and in fact, VP may be a more potent ACTH secretagogue in
birds than is corticotropin-releasing hormone [77].

4.3. OT modulation of behavioral and hormonal stress responses: a


resolution to the paradox?
Although we have focused primarily on the relationship of OT to
aggression, the most obvious effect obtained here is the robust activation of OT neurons following exposure to stressors such as social subjugation or pursuit by a potential human predator. Consistent with this
observation, OT reduces HPA responses to a wide variety of social and
physical stressors in rodents [51,78], and comparable ndings are
obtained in primates, including humans [40,7981]. In general, OT
dampens autonomic responses to stress, as well (rodents: [82,83]),
and modulates behavioral aspects of stress coping. The behavioral
effects on stress coping are complex. For instance, intraventricular infusions of OT or OT agonist decrease immobility in the mouse tail suspension test [84] and forced swim test [85,86], with some evidence that
these effects are mediated via the OTR [85]; however site-specic OT
manipulations in the central amygdala of rats produce an opposite pattern of results in the forced swim assay [87], suggesting that there are
site-specic, species-specic, or other complex OT effects on stress coping that are not yet fully understood. PVN OT neurons likewise promote
passive stress coping in zebra nches [4]. Interestingly, whereas OT effects on stress coping and physiological stress responses are observed
in a diversity of nonsocial paradigms, such as forced swimming and restraint [4,87], effects on anxiety-like behaviors tend to be restricted to
social and reproductive contexts [50]. Moreover, it is important to
note that most anxiety research involves behavioral tests of animals in
a non-social context, which may represent a confound to conclusions
drawn about the role of OT in anxiety, particularly when this research
is compared to research in humans or non-human primates focusing
on social anxiety [40,7981].
Our hypotheses for the present experiments were focused largely on
the central, behavioral functions of OT. Thus, we hypothesized that OT
would be at least permissive for aggression, based on ndings that
socially induced OT release in mammals is anxiolytic [88] and that OT
promotes active stress coping, at least in some paradigms [89]. However, whereas the behavioral effects of OTR antagonism are consistent
with those hypotheses (i.e., antagonism reduces aggression), the results
of the OT-Fos experiment are not. Specically, if the antagonist results
reect OT effects on anxiolysis and/or stress coping, then OT-Fos coexpression should correlate positively with aggression, which is not
the case.

160

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

The most parsimonious resolution to this apparent paradox is complex. First, if OT exerts tonic inhibitory effects on the HPA axis in waxbills, as shown in rats [51], then peripheral OTR antagonism should
increase basal HPA activity. Given that corticosterone (CORT) inhibits
territorial behavior in songbirds [90,91], OTR antagonism could thereby
reduce aggression via increases in CORT release. Notably, in contrast to
the Fos experiment, the antagonist experiment was conducted in the
home cage environment and entailed only a single handling 30 min
prior to testing. Hence, ndings from the antagonist experiment may
reect tonic inhibitory OT effects on the HPA axis, not stress-induced actions of OT. This idea is consistent with our antagonist data, but we
would also expect OT-Fos co-labeling to correlate positively with aggression (based on the OT inhibition of CORT secretion), which is not
the case (Fig. 4). However, the level of stress entailed in the two experiments was likely very different, given that subjects were housed in
small sound booths for the Fos experiment and were handled both before and after testing. Indeed, the behavioral data do suggest that the
Fos testing environment was stressful, given that the dominant birds
were signicantly less aggressive (by ~80%) when tested in the sound
booths, relative to testing in the home environment (Fig. 5). Whether
the acclimation, housing and handling conditions in the Fos experiment
may have represented an acute, chronic or acute short-term stressor is
currently unclear. It is also worth noting that VEWs may be more territorial in their home cage (which was also the cage they occupied with
their mate) used for the antagonist experiment in contrast to the testing
cage used for the Fos experiment that the animals had occupied the
night before testing.
If the testing conditions for the Fos experiment induced a stress response, and hence induced OT-Fos co-labeling across all subjects, we
would expect that the more stress-responsive birds would exhibit
greater OT-Fos co-labeling. Thus, it may be that aggressive male waxbills are hyporesponsive to stress, in terms of CORT secretion, as
shown in other species [4347]. This does not mean that OT neurons
(particularly parvocellular PVN neurons, which project to the anterior
pituitary) are not still exerting inhibitory effects on the HPA axis. Rather,
we hypothesize that the more aggressive birds simply require less of a
brake on that axis in order to exhibit aggression, since they are less
stress responsive. In this scenario, the stress-induced OT-Fos response
is effectively masking the underlying positive association between OT
and aggression, which we hypothesize is mediated via OT's inhibition
of the HPA. An alternative explanation for the negative correlation between OT-Fos co-labeling and aggression in dominate animals is that
more aggressive animals may acclimate faster to testing conditions
rather than being hyporesponsive to stress. However, we would still
hypothesize that the more aggressive birds require less of a brake on
the HPA axis to display aggression due to less OT-Fos co-labeling as
compared to less aggressive birds.
Both interpretations yield the testable hypothesis that baseline CORT
will correlate negatively with aggression. Because we no longer maintain a lab population of violet-eared waxbills, and that they are not commercially available, we cannot address this hypothesis directly (note
that our long-term population of waxbills was originally derived from
African birds that we collected in the wild). Nonetheless, our data do
suggest that the Fos testing environment was stressful, as addressed
above.
An important implication of this hypothesis is that it might explain
the inconsistent results obtained following central OTR antagonist
infusions in male rats, in which no effects on aggression are observed
[20,21], and those obtained here following peripheral antagonist infusions in male and female violet-eared waxbills, in which aggression is
reduced. In contrast to peripheral injections, central antagonist infusions likely have little or no access to the anterior pituitary, and thus
the seemingly inconsistent results may reect differences in the sites
of potential action.
We must also consider that OT cell groups of the SON, MPO and AH
may each play a unique role in stress response that is somewhat

uncoupled from the functions of the PVN and other OT neuronal populations. The present data suggest that this is unlikely, given that the patterns of neuronal activation for each cell group are generally similar, as
are their relationships to behavior. This is not to say that each cell group
does not modulate different aspects of the stress response (autonomic,
hormonal and behavioral), but rather that those activities appear to be
coordinated. In fact, recent experiments in zebra nches demonstrate
that complex dimensions of behavioral phenotype (social competence/dominance, gregariousness, and anxiety) are all signicantly predicted by functional interactions across multiple OT-VP cell groups [60].
Nonetheless, if OT's relationship to aggression is primarily mediated via
effects on CORT, we should expect that the OT neurons of the PVN
(which project to the anterior pituitary) will show the strongest coupling to behavior. As shown in Fig. 4, this is indeed the case.
4.4. Evolving perspectives on a prosocial peptide
A wide variety of work shows that OT promotes afliation behaviors
across the vertebrate taxa, including maternal care, trust, and pair bonding [7,9,54]. This has led to a popular view of OT as a cuddle hormone
and it is increasingly common for behavioral scientists to present OT as a
molecule that selectively promotes prosocial behavior [29,9294]. The
tendency to view OT in this light is somewhat understandable, because
the vast majority of relevant literature does show that OT promotes afliation. Problematically, however, other possible functions have not
been examined in detail, despite the very broad effects that OT exerts
on autonomic, sensory, and anxiety processes [7,95].
This lack of experimental breadth is particularly of concern with
regard to offensive aggression. Only a single study has examined the
effects of site-specic OTR antagonism on offensive aggression in adult
animals. That study demonstrates that OT acts within the POA-AH to
reduce resident-intruder aggression in female Syrian hamsters [31],
although endogenous OT acts within the central amygdala and
paraventricular hypothalamus to facilitate maternal aggression in rats
[18]. Whether these different effects in hamsters and rats relate to the
differences in social context and/or specic brain region remains to be
determined. Although two additional studies have shown that intraventricular infusions of exogenous OT reduce male-male aggression in rats,
no facilitation of aggression is observed following OTR antagonism [20,
21]. In light of these results and those presented in Fig. 5, particular attention should be paid to the species, sex, phenotype and experimental
context in which the behavior is being measured. For instance, in
another study examining aggression in male VEWs, a V1aR antagonist
reduced mate competition aggression but had no effect on residentintruder aggression or male/male aggression in a novel cage, highlighting the importance of experimental context when measuring aggressive
behavior [59]. Furthermore, the V1aR antagonist actually increased
aggression in subordinate, less aggressive males within the resident intruder paradigm (with no effect in dominant males), demonstrating
phenotypic-specic effects, in addition to context-specic effects [59].
We now show that an OTR antagonist signicantly reduces aggression in both male and female violet-eared waxbills, indicating that
endogenous activation of the avian OTR (VT3) is at least permissive
for aggression. The magnitude of the effect is particularly noteworthy
in females, in which displacements are reduced by an average of 71%.
Importantly, an iodinated form of the OTR antagonist used here exhibits
a pattern of binding that is closely matched to the distribution of OTR
(VT3) mRNA [33,38]. Hence, we are condent that the pharmacological
results reect endogenous effects of OTR activation. Furthermore, previous experiments using peripheral and central delivery of this OTR antagonist have shown that the different routes of administration yield
similar results [12], suggesting that the antagonist does gain entrance
to the brain, although we propose that the present ndings reect actions at the level of the pituitary (see previous section).
Importantly, although our ndings our novel with respect to aggression, they are certainly not the rst to show that OT can promote

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

negative behavior and perceptions, as is increasingly well documented


in humans. For instance, intranasal OT administration reduces trust
and cooperation in borderline personality patients [96], and promotes
parochial altruism, ethnocentrism, and out-group derogation in healthy
men [97,98]. Recent work also shows that, in contrast to acute OT
administrations [99], chronic intranasal OT administrations actually impair pair bonding in male prairie voles (as dened by a reduction in
preference for the familiar partner [100]; but see [101]) and promote
aggression in pigs, as well as dysregulate the HPA axis [26]. Furthermore, although not an antisocial effect, intranasal OT administrations
reduce activity in the nucleus accumbens of men viewing pictures of
their own children relative to viewing pictures of other children, suggesting that OT does not increase the social reward associated with
father-offspring bonds [102]. The effects of a single OT administration
during development on the display of adult social behaviors are equally
complex. Female prairie voles that received a low-dose of OT took longer to approach their pups but developed a partner preference like controls in contrast to females that received a high OT dose which retrieved
their pups more often but failed to develop a partner preference [103].
Finally, after a single OT administration at birth, female prairie voles
(but not males) displayed increased aggression and less social behavior
compared to control females or females injected with an OTA antagonist
at birth [24]. Thus, the present dataset is but one piece of a larger body of
evidence that undermines the simplistic notion that OT is a prosocial
neuropeptide [29,94].
Whether the present ndings are predictive for other taxa remains
to be determined, but this seems likely, given the extensive similarities
in OT anatomy and function across vertebrate classes. For instance, OTRs
in both birds and mammals promote maternal care [9,104], facilitate
various aspects of same-sex afliation (e.g., ocking in gregarious
nches, [7]), and are necessary for pair bonding in prairie voles and
zebra nches [54,105].
5. Conclusions
Although afliation-related effects of oxytocic signaling have been
addressed in hundreds of studies, only three prior experiments have examined the relationships between oxytocic signaling and same-sex aggression using OT manipulations in adult animals [20,21,31]. Of these,
only one has demonstrated a function for endogenous OTR-mediated
signaling, reecting site-specic actions in the POA-AH. Using peripheral injections of an OTR antagonist that is known to bind to selectively to
the avian OTR, we have here demonstrated that endogenous activation
of OTRs is required for the normal display of both male and female
aggression in the highly territorial violet-eared waxbill. Importantly,
this experiment was conducted in the home cage environment with
minimal handling. We further demonstrate that multiple OT cell groups
exhibit signicant Fos responses to the stressors of subjugation and pursuit by a human hand. However, evidence suggests that all subjects
were stressed by the experimental environment (which included
isolation in a sound booth and multiple handlings), and we nd that
OT-Fos co-labeling actually correlates negatively with aggression. This
is observed despite the fact that Dominant animals do not differ from
Controls in OT-Fos co-labeling. After considering and discarding various
alternative hypotheses, we propose that this negative relationship reects a greater stress response in the less aggressive animals, and
hence greater OT neuronal activation and HPA response. Considering
that the more aggressive animals are likely hyporesponsive to stress,
as shown in other species [4347], we hypothesize that the more
aggressive animals require less OT-mediated inhibition of the HPA
axis, although HPA inhibition is nonetheless required for the normal
display of aggression. The present results suggest the importance of
considering peripherally-mediated effects of OTR antagonism, and
yield multiple testable hypotheses regarding the interrelationships between behavioral phenotype, OT signaling, CORT and aggression that
should be addressed in future experiments.

161

Acknowledgments
Funding for this work was provided by Indiana University. This
paper is dedicated in loving memory to Jim Goodson (November 17,
1965August, 14, 2014), who advocated for a comprehensive understanding of nonapeptide signaling via the examination of site-specic
effects in the brain, neuronal modulation throughout the social behavior
network, and the consideration of sex, species, phenotype and behavioral context when measuring social behaviors.

References
[1] R. Acher, J. Chauvet, The neurohypophysial endocrine regulatory cascade: precursors, mediators, receptors, and effectors, Front. Neuroendocrinol. 16 (1995)
237289.
[2] A.G. Lee, D.R. Cool, W.C. Grunwald Jr., D.E. Neal, C.L. Buckmaster, M.Y. Cheng, et al.,
A novel form of oxytocin in New World monkeys, Biol. Lett. 7 (2011) 584587.
[3] A.M. Kelly, J.L. Goodson, Social functions of individual vasopressin-oxytocin cell
groups in vertebrates: what do we really know? Front. Neuroendocrinol. 35
(2014) 512529.
[4] A.M. Kelly, J.L. Goodson, Hypothalamic oxytocin and vasopressin neurons exert
sex-specic effects on pair bonding, gregariousness and aggression in nches,
Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 60696074.
[5] D. Ocampo Daza, M. Lewicka, D. Larhammar, The oxytocin/vasopressin receptor
family has at least ve members in the gnathostome lineage, including two distinct
V2 subtypes, Gen. Comp. Endocrinol. 175 (2012) 135143.
[6] Y. Yamaguchi, H. Kaiya, N. Konno, E. Iwata, M. Miyazato, M. Uchiyama, et al., The
fth neurohypophysial hormone receptor is structurally related to the V2-type
receptor but functionally similar to V1-type receptors, Gen. Comp. Endocrinol.
178 (2012) 519528.
[7] J.L. Goodson, R.R. Thompson, Nonapeptide mechanisms of social cognition, behavior and species-specic social systems, Curr. Opin. Neurobiol. 20 (2010) 784794.
[8] C.S. Carter, A.J. Grippo, H. Pournaja-Nazarloo, M.G. Ruscio, S.W. Porges, Oxytocin,
vasopressin and sociality, Prog. Brain Res. 170 (2008) 331336.
[9] I.D. Neumann, The advantage of social living: brain neuropeptides mediate the
benecial consequences of sex and motherhood, Front. Neuroendocrinol. 30
(2009) 483496.
[10] A.H. Veenema, I.D. Neumann, Central vasopressin and oxytocin release: regulation
of complex social behaviours, Prog. Brain Res. 170 (2008) 261276.
[11] R.R. Thompson, J.C. Walton, Peptide effects on social behavior: effects of vasotocin
and isotocin on social approach behavior in male goldsh (Carassius auratus),
Behav. Neurosci. 118 (2004) 620626.
[12] J.L. Goodson, S.E. Schrock, J.D. Klatt, D. Kabelik, M.A. Kingsbury, Mesotocin and
nonapeptide receptors promote estrildid ocking behavior, Science 325 (2009)
862866.
[13] J.L. Goodson, L.C. Wilson, S.E. Schrock, To ock or ght: neurochemical signatures
of divergent life histories in sparrows, Proc. Natl. Acad. Sci. U. S. A. 109 (Suppl. 1)
(2012) 1068510692.
[14] J.P. Herman, J. Flak, R. Jankord, Chronic stress plasticity in the hypothalamic
paraventricular nucleus, Prog. Brain Res. 170 (2008) 353364.
[15] B. Robinzon, T.I. Koike, H.L. Neldon, S.L. Kinzler, I.R. Hendry, M.E. el Halawani, Physiological effects of arginine vasotocin and mesotocin in cockerels, Br. Poult. Sci. 29
(1988) 639652.
[16] G. Leng, T. Onaka, C. Caquineau, N. Sabatier, V.A. Tobin, Y. Takayanagi, Oxytocin and
appetite, Prog. Brain Res. 170 (2008) 137151.
[17] D. Atasoy, J.N. Betley, H.H. Su, S.M. Sternson, Deconstruction of a neural circuit for
hunger, Nature 488 (2012) 172177.
[18] O.J. Bosch, S.L. Meddle, D.I. Beiderbeck, A.J. Douglas, I.D. Neumann, Brain oxytocin
correlates with maternal aggression: link to anxiety, J. Neurosci. 25 (2005)
68076815.
[19] D.A. Lubin, J.C. Elliott, M.C. Black, J.M. Johns, An oxytocin antagonist infused into the
central nucleus of the amygdala increases maternal aggressive behavior, Behav.
Neurosci. 117 (2003) 195201.
[20] F. Calcagnoli, S.F. de Boer, M. Althaus, J.A. den Boer, J.M. Koolhaas, Antiaggressive
activity of central oxytocin in male rats, Psychopharmacology (Berlin) 229
(2013) 639651.
[21] F. Calcagnoli, N. Meyer, S.F. de Boer, M. Althaus, J.M. Koolhaas, Chronic enhancement of brain oxytocin levels causes enduring anti-aggressive and pro-social
explorative behavioral effects in male rats, Horm. Behav. 65 (2014) 427433.
[22] M. Sala, D. Braida, D. Lentini, M. Busnelli, E. Bulgheroni, V. Capurro, et al., Pharmacologic rescue of impaired cognitive exibility, social decits, increased aggression,
and seizure susceptibility in oxytocin receptor null mice: a neurobehavioral model
of autism, Biol. Psychiatry 69 (2011) 875882.
[23] A. Schorscher-Petcu, S. Sotocinal, S. Ciura, A. Dupre, J. Ritchie, R.E. Sorge, et al.,
Oxytocin-induced analgesia and scratching are mediated by the vasopressin-1A
receptor in the mouse, J. Neurosci. 30 (2010) 82748284.
[24] K.L. Bales, C.S. Carter, Sex differences and developmental effects of oxytocin on aggression and social behavior in prairie voles (Microtus ochrogaster), Horm. Behav.
44 (2003) 178184.
[25] R. Jia, F.D. Tai, S.C. An, H. Broders, X.L. Ding, Q. Kong, et al., Effects of neonatal
oxytocin treatment on aggression and neural activities in mandarin voles, Physiol.
Behav. 95 (2008) 5662.

162

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163

[26] J.L. Rault, C.S. Carter, J.P. Garner, J.N. Marchant-Forde, B.T. Richert, D.C. Lay Jr.,
Repeated intranasal oxytocin administration in early life dysregulates the HPA
axis and alters social behavior, Physiol. Behav. 112113 (2013) 4048.
[27] A.C. DeVries, W.S. Young III, R.J. Nelson, Reduced aggressive behaviour in mice with
targeted disruption of the oxytocin gene, J. Neuroendocrinol. 9 (1997) 363368.
[28] K. Macdonald, T.M. Macdonald, The peptide that binds: a systematic review of
oxytocin and its prosocial effects in humans, Harv. Rev. Psychiatry 18 (2010) 121.
[29] A. Meyer-Lindenberg, Impact of prosocial neuropeptides on human brain function,
Prog. Brain Res. 170 (2008) 463470.
[30] N. Striepens, K.M. Kendrick, W. Maier, R. Hurlemann, Prosocial effects of oxytocin
and clinical evidence for its therapeutic potential, Front. Neuroendocrinol. 32
(2011) 426450.
[31] A.C. Harmon, K.L. Huhman, T.O. Moore, H.E. Albers, Oxytocin inhibits aggression in
female Syrian hamsters, J. Neuroendocrinol. 14 (2002) 963969.
[32] S.J. Gutzler, M. Karom, W.D. Erwin, H.E. Albers, Arginine-vasopressin and the
regulation of aggression in female Syrian hamsters (Mesocricetus auratus), Eur. J.
Neurosci. 31 (2010) 16551663.
[33] C.H. Leung, C.T. Goode, L.J. Young, D.L. Maney, Neural distribution of nonapeptide
binding sites in two species of songbird, J. Comp. Neurol. 513 (2009) 197208.
[34] Y. Liu, J.T. Curtis, Z. Wang, Vasopressin in the lateral septum regulates pair bond
formation in male prairie voles (Microtus ochrogaster), Behav. Neurosci. 115
(2001) 910919.
[35] B.T. Searcy, C.S. Bradford, R.R. Thompson, T.M. Filtz, F.L. Moore, Identication and
characterization of mesotocin and V1a-like vasotocin receptors in a urodele
amphibian, Taricha granulosa, Gen. Comp. Endocrinol. 170 (2011) 131143.
[36] T.R. Insel, R. Gelhard, L.E. Shapiro, The comparative distribution of forebrain
receptors for neurohypophyseal peptides in monogamous and polygamous mice,
Neuroscience 43 (1991) 623630.
[37] T.R. Insel, L.E. Shapiro, Oxytocin receptor distribution reects social organization in
monogamous and polygamous voles, Proc. Natl. Acad. Sci. U. S. A. 89 (1992)
59815985.
[38] C.H. Leung, D.F. Abebe, S.E. Earp, C.T. Goode, A.V. Grozhik, P. Mididoddi, et al.,
Neural distribution of vasotocin receptor mRNA in two species of songbird,
Endocrinology 152 (2011) 48654881.
[39] A.K. Beery, E.A. Lacey, D.D. Francis, Oxytocin and vasopressin receptor distributions
in a solitary and a social species of tuco-tuco (Ctenomys haigi and Ctenomys
sociabilis), J. Comp. Neurol. 507 (2008) 18471859.
[40] M. Quirin, J. Kuhl, R. Dusing, Oxytocin buffers cortisol responses to stress in individuals with impaired emotion regulation abilities, Psychoneuroendocrinology 36
(2011) 898904.
[41] H.S. Knobloch, A. Charlet, L.C. Hoffmann, M. Eliava, S. Khrulev, A.H. Cetin, et al.,
Evoked axonal oxytocin release in the central amygdala attenuates fear response,
Neuron 73 (2012) 553566.
[42] L.D. Kubzansky, W.B. Mendes, A.A. Appleton, J. Block, G.K. Adler, A heartfelt response: oxytocin effects on response to social stress in men and women, Biol.
Psychol. 90 (2012) 19.
[43] K. Ebner, C.T. Wotjak, R. Landgraf, M. Engelmann, Neuroendocrine and behavioral
response to social confrontation: residents versus intruders, active versus passive
coping styles, Horm. Behav. 47 (2005) 1421.
[44] C. Carere, T.G. Groothuis, E. Mostl, S. Daan, J.M. Koolhaas, Fecal corticosteroids in a
territorial bird selected for different personalities: daily rhythm and the response
to social stress, Horm. Behav. 43 (2003) 540548.
[45] S. Kralj-Fiser, I.B. Scheiber, A. Blejec, E. Moestl, K. Kotrschal, Individualities in a ock
of free-roaming greylag geese: behavioral and physiological consistency over time
and across situations, Horm. Behav. 51 (2007) 239248.
[46] A.H. Veenema, J.M. Koolhaas, E.R. de Kloet, Basal and stress-induced differences in
HPA axis, 5-HT responsiveness, and hippocampal cell proliferation in two mouse
lines, Ann. N. Y. Acad. Sci. 1018 (2004) 255265.
[47] O. Overli, C. Sorensen, K.G.T. Pulman, T.G. Pottinger, W. Korzan, C.H. Summers,
et al., Evolutionary background for stress-coping styles: relationships between
physiological, behavioral, and cognitive traits in non-mammalian vertebrates,
Neurosci. Biobehav. Rev. 31 (2007) 396412.
[48] J.M. Koolhaas, S.F. de Boer, B. Buwalda, K. van Reenen, Individual variation in coping with stress: a multidimensional approach of ultimate and proximate mechanisms, Brain Behav. Evol. 70 (2007) 218226.
[49] J.M. Koolhaas, S.F. de Boer, C.M. Coppens, B. Buwalda, Neuroendocrinology of coping styles: towards understanding the biology of individual variation, Front.
Neuroendocrinol. 31 (2010) 307321.
[50] I.D. Neumann, L. Torner, A. Wigger, Brain oxytocin: differential inhibition of neuroendocrine stress responses and anxiety-related behaviour in virgin, pregnant and
lactating rats, Neuroscience 95 (2000) 567575.
[51] I.D. Neumann, A. Wigger, L. Torner, F. Holsboer, R. Landgraf, Brain oxytocin inhibits
basal and stress-induced activity of the hypothalamopituitaryadrenal axis in
male and female rats: partial action within the paraventricular nucleus, J.
Neuroendocrinol. 12 (2000) 235243.
[52] J.L. Goodson, M.A. Kingsbury, Nonapeptides and the evolution of social group sizes
in birds, Front. Neuroanat. 5 (2011) 13.
[53] D. Goodwin, Estrildid Finches of the World, Cornell University Press, Ithaca, NY,
1982.
[54] H.E. Ross, L.J. Young, Oxytocin and the neural mechanisms regulating social cognition and afliative behavior, Front. Neuroendocrinol. 30 (2009) 534547.
[55] M. Engelmann, C.T. Wotjak, K. Ebner, R. Landgraf, Behavioural impact of
intraseptally released vasopressin and oxytocin in rats, Exp. Physiol. 85 (2000)
(Spec No:125S-30S).
[56] M. Ludwig, G. Leng, Dendritic peptide release and peptide-dependent behaviours,
Nat. Rev. Neurosci. 7 (2006) 126136.

[57] J.D. Klatt, J.L. Goodson, Sex-specic activity and function of hypothalamic
nonapeptide neurons during nest-building in zebra nches, Horm. Behav. 64
(2013) 818824.
[58] J.L. Goodson, Vasotocin and vasoactive intestinal polypeptide modulate aggression
in a territorial songbird, the violet-eared waxbill (Estrildidae: Uraeginthus
granatina), Gen. Comp. Endocrinol. 111 (1998) 233244.
[59] J.L. Goodson, D. Kabelik, S.E. Schrock, Dynamic neuromodulation of aggression by
vasotocin: inuence of social context and social phenotype in territorial songbirds,
Biol. Lett. 5 (2009) 554556.
[60] A.M. Kelly, J.L. Goodson, Personality is tightly coupled to vasopressinoxytocin
neuron activity in a gregarious nch, Front. Behav. Neurosci. 8 (2014) 55.
[61] S.J. Alger, S.N. Maasch, L.V. Riters, Lesions to the medial preoptic nucleus affect
immediate early gene immunolabeling in brain regions involved in song control
and social behavior in male European starlings, Eur. J. Neurosci. 29 (2009)
970982.
[62] J.L. Goodson, A.K. Evans, A.H. Bass, Putative isotocin distributions in sonic sh:
relation to vasotocin and vocal-acoustic circuitry, J. Comp. Neurol. 462 (2003)
114.
[63] J.L. Goodson, D. Kabelik, A.M. Kelly, J. Rinaldi, J.D. Klatt, Midbrain dopamine
neurons reect afliation phenotypes in nches and are tightly coupled to courtship, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 87378742.
[64] J.L. Goodson, Y. Wang, Valence-sensitive neurons exhibit divergent functional
proles in gregarious and asocial species, Proc. Natl. Acad. Sci. U. S. A. 103
(2006) 1701317017.
[65] J.L. Goodson, A.K. Kelly, M.A. Kingsbury, R.R. Thompson, An aggression-specic cell
type in the hypothalamus of nches, Proc. Natl. Acad. Sci. U. S. A. 109 (2012)
1384713852.
[66] J.L. Goodson, Territorial aggression and dawn song are modulated by septal vasotocin and vasoactive intestinal polypeptide in male eld sparrows (Spizella pusilla),
Horm. Behav. 34 (1998) 6777.
[67] D. Kabelik, J.D. Klatt, M.A. Kingsbury, J.L. Goodson, Endogenous vasotocin exerts
context-dependent behavioral effects in a semi-naturalistic colony environment,
Horm. Behav. 56 (2009) 101107.
[68] J.L. Goodson, D. Kabelik, Dynamic limbic networks and social diversity in vertebrates: from neural context to neuromodulatory patterning, Front. Neuroendocrinol. 30 (2009) 429441.
[69] J.M. Ho, J.H. Murray, G.E. Demas, J.L. Goodson, Vasopressin cell groups exhibit
strongly divergent responses to copulation and malemale interactions in mice,
Horm. Behav. 58 (2010) 368377.
[70] J. Xie, W.J. Kuenzel, P.J. Sharp, A. Jurkevich, Appetitive and consummatory
sexual and agonistic behaviour elicits FOS expression in aromatase and
vasotocin neurones within the preoptic area and bed nucleus of the
stria terminalis of male domestic chickens, J. Neuroendocrinol. 23 (2011)
232243.
[71] A.M. Kelly, J.L. Goodson, Functional signicance of a phylogenetically widespread
sexual dimorphism in vasotocin/vasopressin production, Horm. Behav. 64 (2013)
840846.
[72] J.C. Compaan, R.M. Buijs, C.W. Pool, A.J.H. Deruiter, J.M. Koolhaas, Differential lateral
septal vasopressin innervation in aggressive and nonaggressive male mice, Brain
Res. Bull. 30 (1993) 16.
[73] C.F. Ferris, R.H. Melloni, G. Koppel, K.W. Perry, R.W. Fuller, Y. Delville, Vasopressin/
serotonin interactions in the anterior hypothalamus control aggressive behavior in
golden hamsters, J. Neurosci. 17 (1997) 43314340.
[74] H.E. Albers, The regulation of social recognition, social communication and
aggression: vasopressin in the social behavior neural network, Horm. Behav. 61
(2012) 283292.
[75] K.L. Gobrogge, Y. Liu, L.J. Young, Z. Wang, Anterior hypothalamic vasopressin
regulates pair-bonding and drug-induced aggression in a monogamous rodent,
Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 1914419149.
[76] H.E. Albers, A. Dean, M.C. Karom, D. Smith, K.L. Huhman, Role of V1a vasopressin
receptors in the control of aggression in Syrian hamsters, Brain Res. 10731074
(2006) 425430.
[77] W.J. Kuenzel, A. Jurkevich, Molecular neuroendocrine events during stress in poultry, Poult. Sci. 89 (2010) 832840.
[78] R.J. Windle, N. Shanks, S.L. Lightman, C.D. Ingram, Central oxytocin administration
reduces stress-induced corticosterone release and anxiety behavior in rats,
Endocrinology 138 (1997) 28292834.
[79] S.S. Hall, A.A. Lightbody, B.E. McCarthy, K.J. Parker, A.L. Reiss, Effects of intranasal oxytocin on social anxiety in males with fragile X syndrome, Psychoneuroendocrinology
37 (2012) 509518.
[80] M. Heinrichs, T. Baumgartner, C. Kirschbaum, U. Ehlert, Social support and oxytocin
interact to suppress cortisol and subjective responses to psychosocial stress, Biol.
Psychiatry 54 (2003) 13891398.
[81] K.J. Parker, C.L. Buckmaster, A.F. Schatzberg, D.M. Lyons, Intranasal oxytocin
administration attenuates the ACTH stress response in monkeys,
Psychoneuroendocrinology 30 (2005) 924929.
[82] A.J. Grippo, D.M. Trahanas, R.R. Zimmerman II, S.W. Porges, C.S. Carter, Oxytocin
protects against negative behavioral and autonomic consequences of long-term
social isolation, Psychoneuroendocrinology 34 (2009) 15421553.
[83] A.J. Grippo, H. Pournaja-Nazarloo, L. Sanzenbacher, D.M. Trahanas, N. McNeal,
D.A. Clarke, et al., Peripheral oxytocin administration buffers autonomic but not
behavioral responses to environmental stressors in isolated prairie voles, Stress
15 (2012) 149161.
[84] R.H. Ring, L.E. Schechter, S.K. Leonard, J.M. Dwyer, B.J. Platt, R. Graf, et al., Receptor
and behavioral pharmacology of WAY-267464, a non-peptide oxytocin receptor
agonist, Neuropharmacology 58 (2010) 6977.

J.L. Goodson et al. / Physiology & Behavior 141 (2015) 154163


[85] S. Chaviaras, P. Mak, D. Ralph, L. Krishnan, J.H. Broadbear, Assessing the
antidepressant-like effects of carbetocin, an oxytocin agonist, using a modication
of the forced swimming test, Psychopharmacology (Berlin) 210 (2010) 3543.
[86] E. Loyens, D. De Bundel, H. Demaegdt, S.Y. Chai, P. Vanderheyden, Y. Michotte,
et al., Antidepressant-like effects of oxytocin in mice are dependent on the presence of insulin-regulated aminopeptidase, Int. J. Neuropsychopharmacol. (2012)
111.
[87] K. Ebner, O.J. Bosch, S.A. Kromer, N. Singewald, I.D. Neumann, Release of oxytocin
in the rat central amygdala modulates stress-coping behavior and the release of
excitatory amino acids, Neuropsychopharmacology 30 (2005) 223230.
[88] A.S. Smith, Z. Wang, Salubrious effects of oxytocin on social stress-induced decits,
Horm. Behav. 61 (2012) 320330.
[89] I.D. Neumann, R. Landgraf, Balance of brain oxytocin and vasopressin: implications
for anxiety, depression, and social behaviors, Trends Neurosci. 35 (2012) 649659.
[90] S.L. Meddle, L.M. Romero, L.B. Astheimer, W.A. Buttemer, I.T. Moore, J.C. Wingeld,
Steroid hormone interrelationships with territorial aggression in an Arcticbreeding songbird, Gambel's white-crowned sparrow, Zonotrichia leucophrys
gambelii, Horm. Behav. 42 (2002) 212221.
[91] L.M. Romero, S.C. Dean, J.C. Wingeld, Neurally active stress peptide inhibits territorial defense in wild birds, Horm. Behav. 34 (1998) 239247.
[92] S.M. van Anders, K.L. Goldey, P.X. Kuo, The steroid/peptide theory of social bonds:
integrating testosterone and peptide responses for classifying social behavioral
contexts, Psychoneuroendocrinology 36 (2011) 13651375.
[93] P.J. Zak, The physiology of moral sentiments, J. Econ. Behav. Organ. 77 (2011)
5365.
[94] A. Meyer-Lindenberg, G. Domes, P. Kirsch, M. Heinrichs, Oxytocin and vasopressin
in the human brain: social neuropeptides for translational medicine, Nat. Rev.
Neurosci. 12 (2011) 524538.

163

[95] J.L. Goodson, Deconstructing sociality, social evolution, and relevant nonapeptide
functions, Psychoneuroendocrinology 38 (2013) 465478.
[96] J.A. Bartz, J. Zaki, N. Bolger, K.N. Ochsner, Social effects of oxytocin in humans: context and person matter, Trends Cogn. Sci. 15 (2011) 301309.
[97] C.K. De Dreu, L.L. Greer, G.A. Van Kleef, S. Shalvi, M.J. Handgraaf, Oxytocin promotes
human ethnocentrism, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 12621266.
[98] C.K. De Dreu, L.L. Greer, M.J. Handgraaf, S. Shalvi, G.A. Van Kleef, M. Baas, et al., The
neuropeptide oxytocin regulates parochial altruism in intergroup conict among
humans, Science 328 (2010) 14081411.
[99] K.L. Bales, C.S. Carter, Developmental exposure to oxytocin facilitates partner
preferences in male prairie voles (Microtus ochrogaster), Behav. Neurosci. 117
(2003) 854859.
[100] K.L. Bales, A.M. Perkeybile, O.G. Conley, M.H. Lee, C.D. Guoynes, G.M. Downing,
et al., Chronic intranasal oxytocin causes long-term impairments in partner preference formation in male prairie voles, Biol. Psychiatry 74 (2012) 180188.
[101] L.J. Young, When too much of a good thing is bad: chronic oxytocin, development,
and social impairments, Biol. Psychiatry 74 (2013) 160161.
[102] D. Wittfoth-Schardt, J. Grunding, M. Wittfoth, H. Lanfermann, M. Heinrichs, G.
Domes, et al., Oxytocin modulates neural reactivity to children's faces as a function
of social salience, Neuropsychopharmacology 37 (2012) 17991807.
[103] K.L. Bales, J.A. van Westerhuyzen, A.D. Lewis-Reese, N.D. Grotte, J.A. Lanter, C.S.
Carter, Oxytocin has dose-dependent developmental effects on pair-bonding and
alloparental care in female prairie voles, Horm. Behav. 52 (2007) 274279.
[104] A. Thayananuphat, O.M. Youngren, S.W. Kang, T. Bakken, S. Kosonsiriluk, Y.
Chaiseha, et al., Dopamine and mesotocin neurotransmission during the transition
from incubation to brooding in the turkey, Horm. Behav. 60 (2011) 327335.
[105] J.D. Klatt, J.L. Goodson, Oxytocin-like receptors mediate pair bonding in a socially
monogamous songbird, Proc. R. Soc. Lond. B Biol. Sci. 280 (2013) 20122396.

Você também pode gostar