OxytocinVasopressinandSocialBehavior-FromNeuralCircuitstoClinicalOpprotunities

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/362188695

Oxytocin, Vasopressin, and Social Behavior: From Neural Circuits to Clinical


Opportunities

Article in Endocrinology · July 2022


DOI: 10.1210/endocr/bqac111

CITATIONS READS

54 784

4 authors, including:

Nicole Rigney Geert J. De Vries


Emory University Georgia State University
12 PUBLICATIONS 225 CITATIONS 175 PUBLICATIONS 17,146 CITATIONS

SEE PROFILE SEE PROFILE

Aras Petrulis
Georgia State University
60 PUBLICATIONS 1,613 CITATIONS

SEE PROFILE

All content following this page was uploaded by Nicole Rigney on 27 January 2023.

The user has requested enhancement of the downloaded file.


Endocrinology, 2022, 163, 1–13
https://doi.org/10.1210/endocr/bqac111
Advance access publication 21 July 2022
Mini-Review

Oxytocin, Vasopressin, and Social Behavior: From Neural


Circuits to Clinical Opportunities

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


Nicole Rigney,1 Geert J. de Vries,1,2 Aras Petrulis,1 and Larry J. Young3,4,5,
1
Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
2
Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA
3
Center for Translational Social Neuroscience, Emory University, Atlanta, Georgia 30329, USA
4
Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329,
USA; and
5
Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
Correspondence: Larry J. Young, PhD, Emory National Primate Center, Emory University, 954 Gatewood Rd, Atlanta, GA 30329, USA. Email: lyoun03@emory.edu.

Abstract
Oxytocin and vasopressin are peptide hormones secreted from the pituitary that are well known for their peripheral endocrine effects on child-
birth/nursing and blood pressure/urine concentration, respectively. However, both peptides are also released in the brain, where they modulate
several aspects of social behaviors. Oxytocin promotes maternal nurturing and bonding, enhances social reward, and increases the salience of
social stimuli. Vasopressin modulates social communication, social investigation, territorial behavior, and aggression, predominantly in males.
Both peptides facilitate social memory and pair bonding behaviors in monogamous species. Here we review the latest research delineating the
neural circuitry of the brain oxytocin and vasopressin systems and summarize recent investigations into the circuit-based mechanisms modu-
lating social behaviors. We highlight research using modern molecular genetic technologies to map, monitor activity of, or manipulate neuro-
peptide circuits. Species diversity in oxytocin and vasopressin effects on social behaviors are also discussed. We conclude with a discussion of
the translational implications of oxytocin and vasopressin for improving social functioning in disorders with social impairments, such as autism
spectrum disorder.
Key Words: autism, neuropeptides, social communication, social behavior, oxytocin, vasopressin
Abbreviations: AON, accessory olfactory nucleus; AMY, amygdala; AVP, arginine vasopressin; ASD, autism spectrum disorder; BNST, bed nucleus of the stria
terminalis; DR, dorsal raphe; HYP, hypothalamus; KO, knockout; LHb, lateral habenula; LS, lateral septum; LC, locus coeruleus; MeA, medial amygdala; MPOA,
medial preoptic area; NAcc, nucleus accumbens; OB, olfactory bulb; OXT, oxytocin; OXTR, oxytocin receptor; PAG, periaqueductal gray; PVN, paraventricular nu-
cleus of the hypothalamus; PMd, premammillary nucleus (dorsal); SON, supraoptic nucleus of the hypothalamus; VP, ventral pallidum; VTA, ventral tegmental area.

Social distancing and isolation during the COVID-19 pan- and empathy-like consoling (8-14), whereas central AVP has
demic have caused major disruptions to our social relation- been shown to regulate social communication, territorial ag-
ships, likely affecting our well-being, as social interactions are gression, pair bonding, and mate guarding, especially in males
fundamental for physical and mental health (1). Dysfunction (9, 15, 16). Some of the behavioral effects of OXT and AVP
in social communication and behavior is also a prominent are functionally linked to their peripheral function, as OXT
aspect of many psychopathologies including autism, schizo- promotes birth and nursing but also parental nurturing, while
phrenia, and social anxiety (2). Considerable insights into the AVP modulates urine production and also territorial behavior,
biological framework regulating functional social behavior, which often involves urine and scent marking (17, 18). Since
and therefore its dysfunction, have been gained through exam- these peptides modulate social cognition and motivation, ma-
ination of oxytocin (OXT) and arginine vasopressin (AVP) nipulations of the OXT and AVP systems offer promising fu-
neuropeptide systems and their nonmammalian homologues. ture therapeutics for individuals with social deficits, such as in
OXT and AVP were first identified as regulators of periph- autism spectrum disorder (ASD) (19-21).
eral physiology, being secreted into the bloodstream through Although the neural mechanisms underlying social behavior
hypothalamic terminals in posterior pituitary and median are highly complex, recent anatomical, genetic, and molecular
eminence to influence parturition and lactation (OXT) and technologies have considerably advanced our understanding
water retention, blood pressure, body temperature, and the of how OXT and AVP act within the brain to regulate so-
hypothalamic-pituitary-adrenal axis (AVP) (3-6). In the 1960s, cial behavior. Most of this recent work has been conducted in
work by de Wied substantially advanced the field of endocrin- laboratory mice because of the availability of powerful gen-
ology by demonstrating that AVP can act in the brain to alter etic tools in this species, such as recombinase-expressing and
behavior, thereby introducing the concept of “neuropeptide” conditional knockout (KO) strains that, when combined with
(7). Since then, OXT acting centrally has been shown to be es- site-specific targeting with viral vectors, allow unprecedented
sential for maternal behavior, mother-infant bonding, sexual access to chemically and spatially defined neural systems.
behavior, and other affiliative behaviors such as pair bonding These advancements have allowed researchers to manipulate
Received: 1 April 2022. Editorial Decision: 11 July 2022. Corrected and Typeset: 27 July 2022
© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
2 Endocrinology, 2022, Vol. 163, No. 9

neurons expressing OXT and AVP and their cognate recep- Oxytocin
tors within the “social behavior neural network” (SBNN), as PVN OXT-expressing cells send fibers to the SON forming
well as in other areas, such as the hippocampus, and thereby the neurohypophyseal tract (44), and classical retro-
determine how these discrete, interlinked systems alter social grade tracing and modern viral tracing strategies in rats
behavior (6, 16, 22). Since laboratory mice have limited be- and mice have shown that the PVN OXT-expressing cells
havioral diversity, and species differences in the distribution also send projections to the thalamus, cortex, amygdala,
of OXT/AVP receptors are well documented (10), it is im- striatum, and hippocampus (Fig. 1A (53-55). The SON
portant to expand these intersectional tools to other species OXT-expressing cells have more restricted projections,
(23). For example, oxytocin and vasotocin modulate social mainly to the posterior pituitary, but also project to the
interactions in many species including teleost fish, lizards,

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


medial and central amygdala (44). In rats and mice, sev-
and birds (24-26) and genome editing approaches are now eral regions provide input to PVN OXT-expressing cells,
being used to examine OXT and AVP circuitry and social be- including the brainstem, thalamic, hypothalamic, and cor-
havioral functions in prairie voles and medaka fish (27-30). tical cell populations (56-58) (see Fig. 1A). Recently, Son
Importantly, the disproportionate focus on OXT compared to et al (59) found no correlation between OXT neuronal
AVP systems suggests there is a need to rebalance research so projections and OXTR expression, consistent with para-
that the role of different AVP circuits in social behavior can crine signaling.
be refined and expanded.
Consequently, this mini-review will highlight recent work Vasopressin
on the behaviorally relevant sources of OXT and AVP, the PVN AVP-expressing cells send projections to the thalamus,
inputs and outputs of these sources, and the receptor systems cortex, amygdala, striatum, and hippocampus (Fig. 1B).
they act on to generate complex social behaviors; see (6, 16, Although the number of PVN AVP neurons do not appear
31-38) for reviews of foundational work that guided recent to differ by sex, central projections from these neurons are
discoveries of OXT/AVP’s role in social behavior. generally denser in female than in male mice (60). PVN/SON
AVP cells receive monosynaptic inputs locally and from lat-
eral septum (LS), bed nucleus of the stria terminalis (BNST),
Neuronal Sources of Oxytocin and Vasopressin MPOA, ventromedial hypothalamus, and suprachiasmatic
Hypothalamus nucleus (Fig. 1B). Some differences between inputs to PVN
The paraventricular (PVN) and supraoptic nuclei (SON) of and SON cells are apparent. For example, PVN AVP cells re-
the hypothalamus release OXT and AVP, both centrally and ceive stronger input from the BNST than does the SON. Few
peripherally. Within these regions, each peptide is stored in of the inputs to PVN AVP cells are AVP immunoreactive,
large, dense core vesicles, which can be released through and may, in fact, be OXT-ergic (61, 62), suggesting func-
somatodendritic, en passant axonal release, as well as tional crosstalk between OXT and AVP PVN cells. Whether
through synaptic terminals (39-41). AVP is also produced release of hypothalamic OXT and AVP is synchronized
in other hypothalamic nuclei such as the suprachiasmatic across all OXT/AVP sources under various social conditions
nucleus, well known for regulating circadian rhythms or has region-specific projections for distinct social behav-
(42), and the accessory nuclei/nucleus circularis region, a iors is still unclear.
likely source of AVP regulating aggression and competitive
communication (43). OXT is also produced in the medial Extended Amygdala
preoptic area (MPOA), where it regulates maternal and Oxytocin
sexual behavior (44). OXT neurons have been reported in the extended medial
Both the PVN and SON contain magnocellular OXT and amygdala (eg, the BNST and medial amygdala proper [MeA])
AVP neurosecretory cells that project to the posterior pi- in rats (63), California mice (64), and marmosets (65). Very
tuitary, releasing each peptide peripherally as a hormone little is known about the neuroanatomical connectivity
(45), and the PVN contains an additional population of of these neurons. However, OXT neurons in the BNST of
nonneurosecretory, parvocellular cells that modulate anterior California mice make local connections within the BNST,
pituitary release of adrenocorticotropin (46), as well as pro- project to the hypothalamus, and are involved in social vigi-
ject to other brain and spinal cord areas (47, 48); for a com- lance (66).
prehensive review see (6, 40, 49). Some reports have shown
that AVP messenger RNA (mRNA) density measurements in Vasopressin
the SON of rats are greater in males (50), while, in mice, OXT The extended medial amygdala contains neurons that ex-
production is greater in females (51); however, other studies press AVP in a steroid-dependent and sexually dimorphic
have shown a similar amount of OXT and AVP in both sexes manner (33). Indeed, the BNST and MeA contribute to the
in the PVN/SON of various rodent species, nonhuman pri- most pronounced sex difference in AVP innervation in the
mates, and humans (49, 52). brain (33, 60). For example, male rodents have 2 to 3 times
To better understand how OXT and AVP modulate neural as many AVP cells as females in the BNST/MeA and pro-
circuits controlling social behavior, it is important to delin- nounced sex-different projections to areas involved in so-
eate the central nervous system projections of these neurons cial behavior such as the ventral pallidum, lateral septum
as well as their inputs. The development of mice with Cre (LS), lateral habenula, dorsal raphe (DR), periaqueductal
recombinase driven by OXT or AVP neurons as well as vir- gray, as well as other forebrain areas (33, 60) (Fig. 1C).
uses with transgenes driven by neuropeptide promoters have The sex difference in AVP expression is driven primarily
considerably enhanced our understanding of OXT and AVP by organizational effects of steroid hormones, although
circuit architecture. hormone-independent influences of sex chromosomes play
Endocrinology, 2022, Vol. 163, No. 9 3

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022

Figure 1. Neural circuits (inputs and outputs) of oxytocin (OXT) and arginine vasopressin (AVP). Blue arrows indicate OXT and AVP neuronal inputs,
while red arrows indicate OXT and AVP neuronal outputs. Combined blue and red arrows indicate regions that share both inputs and outputs. HYP
refers to hypothalamic regions outside the paraventricular nucleus (PVN) and supraoptic nuclei (SON) of the hypothalamus. A, PVN and SON OXT
neurons. B, PVN and SON AVP neurons. C, Sexually dimorphic AVP neurons within the bed nucleus of the stria terminalis (BNST) and medial amygdala
(MeA).

a role (67). It is currently unknown which regions and Collectively, OXT and AVP cells in PVN receive and
neuronal cell types send monosynaptic inputs to BNST or send projections to similar regions, including thalamus,
MeA AVP. hypothalamus, midbrain, and extended amygdala. More
4 Endocrinology, 2022, Vol. 163, No. 9

neuroanatomical experiments are needed to map the con- for long-term neural plasticity after social interactions
nectivity of OXT/AVP cells in the extended amygdala. (10).
Although constitutive KO of OXT does not alter social ap-
proach in mice (79), specific OXT systems do influence so-
Oxytocin and Vasopressin Receptors cial reward, approach, and investigation. For example, Dölen
OXT has one canonical receptor type (ie, OXTR), whereas et al (80), using elegant circuit manipulations, demonstrated
AVP has 3: V1aR, V1bR, and V2. The behavioral effects of that PVN OXT and DR serotonin projections to the nucleus
AVP are mediated primarily by V1aR (or Avpr1a) and V1bR accumbens (NAcc) are required for social reward in mice.
(16, 37, 38, 49, 68), while AVP acts on V2 to regulate fluid Further, PVN OXT cells are responsible for the facilitatory
homeostasis in the kidney (69). Most research investigating effects of 3,4-methylendioxymethamphetamine (MDMA) on

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


OXT and AVP functions do so by manipulating the recep- mouse prosocial behavior and social reward memory (81).
tors, or receptor-expressing neurons, without regard to the PVN OXT can also facilitate social reward through its ac-
source of the peptide. One of the most interesting features tions on brain dopamine systems (82), originating, for ex-
of the OXT/AVP system is the diversity in receptor func- ample, in the ventral tegmental area (VTA), and through
tion in relation to behavior. While there is remarkable con- endocannabinoid-dependent mechanisms (83). In male
sistency in the distribution of OXT and AVP neurons and and female Syrian hamsters, social interaction increases the
their projections across vertebrates (33), there are striking amount of c-Fos expression (a marker for neural activity) in
species differences in the distributions in the distribution OXT-immunoreactive neurons in the PVN and SON, while
of OXT and AVP receptors in the brain, even between OXTRs in the VTA mediate the rewarding properties of these
closely related species (10). Humans, chimpanzees, and social interactions (84).
other nonhuman primate species all show unique distribu- Similarly, in mice, social touch during early life increases
tions of OXTR and V1aR in the brain, which may relate both PVN OXT neuronal firing and social approach later
to species differences in primate social behaviors (70). The in adulthood through interaction with substance P systems
behavioral significance of diversity in receptor distribution, within the periaqueductal gray (58). PVN OXT cells in adults
both within and between species, has been highlighted by are also active during interactions with juveniles (85), and
studies of the neural basis of monogamy in prairie voles stimulation of PVN OXT terminals in the VTA increased
(discussed later). In addition, the distribution of OXTR social investigation through actions on OXTR specifically.
messenger RNA in prairie vole brain is surprisingly wide- Moreover, deletion of OXTR from dopaminergic neurons in
spread, suggesting that OXT may modulate essentially the VTA eliminated preferences for interacting with juveniles
brain-wide circuit activity (71). In California mice, OXTR (82). Monogamous mandarin voles raised without a father
is expressed in different cell types depending on the brain socialize less and have fewer PVN OXT cells, a behavioral
region (72). difference that can be reversed by optogenetic stimulation of
PVN OXT terminals within the prelimbic cortex (86). Taken
together, OXT from the PVN influences social behavior via its
Modulation of Social Behavior
actions on defined DR serotonin– and dopamine system–ex-
Sensory Processing of Social Cues pressing populations as well as others, perhaps serving as a
Olfactory processing is critical for appropriate social be- “gating” mechanism for the flow of information to reward
havior driven by chemosignals produced by a conspecific systems (83, 87).
(73). While there are no known OXT cells in the olfac- Historically OXT is known for having prosocial effects on
tory bulb (OB), PVN OXT can act early in the olfactory behavior, whereas AVP is thought to play more of a role in
processing stream, via OXT receptors in the anterior ol- aggression, stress, and anxiety. However, in California mice,
factory nucleus. This system facilitates social recognition in knockdown of OXT or administration of OXTR antagonist
rats by indirectly inhibiting the primary projection cells in in the anterior BNST increased social approach and decreased
the OB (74, 75), thereby reducing background “noise” and social vigilance, an anxiety-like behavior, which suggests that
increasing signal salience. A similar process may also occur OXT may also drive social anxiety-like behavior depending
via AVP release, from inhibitory OB interneurons, in con- on its source or context (66, 88).
junction with cholinergic mechanisms (76, 77). This modu- AVP can have prosocial effects as well. With site-specific
lation of signal to noise may be a common neural circuit targeting using viral vectors, we have begun to identify which
mechanism to enhance the salience of multimodal social AVP sources are responsible for specific social behaviors in
stimuli (10, 78). mice. AVP cell groups appear to play opposite roles in so-
cial investigation in males and females depending on the
Social Motivation and Investigation source. BNST AVP cell ablation and AVP knockdown both
To respond adaptively to their social environment, ani- reduced male-male social investigation. These are similar to
mals must be motivated to respond to social signals. effects of BNST AVP knockdown in avian species (reviewed
After processing social information (eg, on familiarity, in [89]). PVN AVP cell ablation increased female social in-
sex, age, health), animals must decide how to interact: vestigation, but does not alter investigation in males (52, 90,
communicate, fight/compete, flee, cooperate, reproduce, 91). However, PVN AVP cell stimulation in male mice caused
and, in some species, form pair bonds and raise offspring self-grooming instead of social interactions with female mice
together. OXT and AVP sources in the hypothalamus and (92). Nevertheless, there may be differential involvement of
extended amygdala play pivotal and distinct roles in so- PVN and BNST AVP cells in the control of social/emotional
cial investigation, motivation, and social reward (dis- behavior in each sex (52). There is also a sex-specific role of
cussed later). Further, OXT (and likely AVP) is important AVP in the regulation of social play behavior in juvenile rats,
Endocrinology, 2022, Vol. 163, No. 9 5

as blocking V1aR in the LS increases social play behavior in Social Communication, Territoriality, and
males and decreases it in females (49). The extreme sexually Aggression
dimorphic nature of BNST AVP cells may explain stronger AVP was first shown to influence social communication by
effects in males than in females. The female bias in the effects its ability to induce flank marking in Syrian hamsters in the
of similar manipulations of PVN AVP cells may be related anterior hypothalamus and MPOA (16, 111-113). AVP in
to females having denser PVN AVP projections compared to these areas is also critical for the establishment of domin-
males (60). ance relationships; for a comprehensive review on AVP effects
on aggression and communication, see (114). Mice deposit
Social Recognition urine marks to communicate social status and dominance

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


Although social behavior is diverse across species, one (115). Removal of BNST AVP cells in mice increases male
common feature is that the animal must differentiate urine marking toward females. However, AVP knockdown in
between conspecifics, which is critical for the establish- this region did not affect urine marking but did reduce other
ment of relationships between animals (ie, affiliative, ter- aspects of male communication (eg, vocalizations and tail rat-
ritorial). OXT and AVP systems have long been known tles), suggesting that BNST AVP is involved in some aspects of
to play a major role in the formation of social memory male offensive/territorial signaling (91). In contrast, blockade
(93). For example, mice with impairments in either system of V1a receptors in the lateral habenula and DR (downstream
show deficits in distinguishing between familiar and novel BNST structures) reduced, instead of increased, male urine
conspecifics (74, 94, 95). OXTRs in the MeA receive OXT marking toward potential competitor males. More work is
input from the PVN that help discriminate between sex- clearly needed to dissect which AVP sources and output re-
and individual cues (53, 74, 96). Ablating OXTR expres- gions are responsible for urine marking and other forms of
sion in aromatase-positive neurons in the MeA abolishes social communication in mice.
male mouse preferences for females (96). Social memory AVP also plays a role in aggression in prairie voles and
processing also requires the hippocampus. OXT and AVP- Syrian hamsters, sometimes in a sex-specific manner. For ex-
expressing cells in PVN and SON send projections to ample, pharmacological studies revealed that AVP in the an-
the hippocampus, with the highest fiber density seen in terior hypothalamus facilitates intermale aggression both in
the CA2 region for AVP and CA2/CA3 regions for OXT prairie voles and hamsters (16, 43, 116), while the opposite
projections (97-100). OXTRs in the anterior DG-CA2/ effect was found in female hamsters (117). In hamsters, social
CA3 circuit facilitate social, but not object discrimin- isolation increases aggression both in male and females, but
ation (101), and AVP projections from PVN to hippo- V1aR density is higher in males than females (114). Recently,
campus influence recognition of conspecifics as well as constitutive KO of Avpr1a in Syrian hamsters paradoxic-
spatial features in mice (102). For example, Smith et al ally increased flank marking and aggression in both sexes,
(99) demonstrated that optogenetic activation of the PVN highlighting the complex role of Avpr1a in territorial behav-
AVP cell inputs to hippocampal CA2 area enhances social iors and potentially highlighting the role of compensatory
memory, via a V1bR mechanism, in mice. Further, when mechanisms and/or effects on Avpr1a KO in multiple brain
the PVN to CA2 pathway is activated chemogenetically, regions (118). Collectively, these data suggest more work is
nonmonogamous C57Bl6 mice form partner preferences needed using comparative and gene-editing approaches to
in a species-atypical way (103). Projections from PVN to elucidate AVP’s role in social communication and aggression.
other brain regions can also alter aspects of social rec-
ognition. For example, PVN OXT input to the central Sexual Behavior
amygdala mediates the ability of mice to discriminate be- OXT is strongly involved both in male and female sexual be-
tween negative and positive emotional states of conspe- havior through central and peripheral action (38). OXT con-
cifics (104). tributes to sexual arousal and sexual reward in males, while
Taken together, these studies suggest that AVP and OXT OXT influences lordosis and orgasm in females (38). In rats,
cells in the PVN help generate social preferences and influ- PVN OXT neurons are active during sexual behavior in males
ence social recognition by direct hippocampal, amygdala, and females and blocking OXT action in ventromedial hypo-
and cortical synaptic release. One explanation for OXT and thalamus and MPOA reduces female receptivity (38).
AVP effects on social recognition may be that these peptides In mice, BNST and MeA AVP cells are active during male
act as attentional modulators, increasing the salience of so- sexual behavior (101, 119) and knockdown of AVP within
cial information through different circuits (10). Indeed, acti- the BNST reduced the number of intromissions and ejacula-
vated PVN OXT neurons increased not only affiliative, but tions in males, but did not alter sexual behavior in females
also agonistic behaviors (105). For a comprehensive review (91). Hormonal action on this cell population may help drive
on OXT and AVP’s role in social memory via the hippo- aspects of steroid-sensitive sexual behavior since gonadal hor-
campus, see (106). However, it is unlikely that just PVN mones affect both male sexual behavior (120, 121) and AVP
projections contribute to AVP effects on social recognition expression within the extended amygdala (33).
as ablation of AVP cells in PVN and SON did not affect
social recognition (52, 107). Extrahypothalamic AVP pro- Parental Behavior
jections may therefore be an important regulator of social OXT is well known for its influence on social bonding and
recognition. For example, BNST AVP cell ablation impairs maternal behavior (9, 18), and its role in parental behavior
social recognition in males, likely through its projection to highlights the putative function of OXT as an attentional
the LS (94, 108, 109), and restoration of BNST—LS AVP modulator (122). For example, OXT from PVN increases the
signaling in Magel2-KO mice ameliorated social discrimin- salience of mouse pup vocalizations, while a dam’s experience
ation deficits (110). with pups adjusts the balance of excitatory and inhibitory
6 Endocrinology, 2022, Vol. 163, No. 9

tuning in the auditory cortex (54, 123, 124). Further, PVN bonding. Viral-mediated small interfering RNA knockdown
OXT neurons are activated when virgin mice observe pup of OXTR in NAcc of prairie voles inhibits partner preference
retrievals by another mother and optogenetic stimulation of (138), while overexpressing NAcc OXTR enhances partner
PVN OXT efferents to auditory cortex enhances pup retrieval preference formation (139). The OXT fibers in the NAcc ori-
learning in virgin mice (124, 125). This increase in OXT re- ginate from the PVN (140), and OXT is released in females
lease by the PVN may be driven by the posterior intralaminar during mating (141).
complex of the thalamus inputs to the PVN (56). Other loci Robust individual differences in NAcc relate to individual
of PVN OXT action include the VTA and NAcc as PVN OXT variation in alloparental nurturing behavior, and resilience
projections to these areas promote paternal behaviors in man- to early-life adversity with respect to later-life pair bonding
darin voles (126). An animal’s transition to parenthood is (142, 143). This variation in OXTR density in the NAcc is de-

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


known to affect sensory perception of infant cues. How PVN termined by genetic variation in the Oxtr gene (144), which in
OXT modulates this change through various OXT circuits turn predicts how early-life nurturing experience affects later-
remains to be further explored. Additionally, parental/sensory life bonding ability (143).
deprivation or cesarean delivery can lead to disruptions of Recent ex vivo electrophysiological studies provide clues
hypothalamic OXT and maternal behavior (127-130), while, about how one’s social experience can alter OXTR signaling
in rats, enriched environments increase hypothalamic OXT in effect on NAcc activity to influence pair-bond formation and
dams and their male offspring (131). Therefore, OXT, social expression. In virgin female prairie voles, OXTR signaling,
experience, and environment heavily influence functional par- mimicking the first mating, decreases spontaneous activity of
ental behavior. NAcc neurons, effectively reducing the noise in the circuit,
There are far fewer studies examining the role of central perhaps to facilitate synaptic plasticity to make partner cues
AVP on parental behavior. This is not surprising given that rewarding. After pair bonding, OXTR signaling increases
AVP is traditionally associated with territorial behavior and evoked NAcc cell activity from neural inputs, perhaps amp-
anxiety. However, the seminal paper demonstrating the effect lifying the signal of the neural representation of the partner,
of OXT on maternal behavior also showed that AVP stimu- making the partner reinforcing and motivating affiliation
lates maternal behavior, but with a longer latency of action (140). This experience-dependent change in OXTR signaling
(132). Recent work has demonstrated that chemogenetic on physiology is mediated by OXTR-endocannabinoid inter-
inhibition of PVN AVP cell activity increases nest building actions, and blocking CB1 receptor elicits defensive rejection
in male mice (while excitation decreases nest building in fe- by the female of the partner. Interestingly, OXT stimulates the
males), without altering other aspects of parental behavior activation of NAcc activity in men in monogamous relation-
(133). Knockdown of V1aR in the MPOA impairs maternal ships when viewing images of their partner (145).
behavior in rats (134). Although the precise sources of AVP Once the pair bond is formed, withdrawal of PVN OXT
regulating parental behavior is undefined, AVP projections signaling in NAcc mediates “grieving”-like passive coping
from the extended amygdala may also influence maternal be- behavior following the loss of the partner, which is mim-
havior, as AVP in the LS facilitates paternal behavior in prairie icked in the presence of the partner by small interfering RNA
voles (135, 136). While hypothalamic OXT plays a substan- knockdown of NAc OXTR (140, 146). The negative affect
tial role in rodent maternal behavior compared to AVP, the stimulated by the loss of OXT signaling when separated by
sources of AVP responsible for V1aR-mediated changes in the partner may serve to motivate partners to stay together in
parental behavior require further examination. enduring relationships.
Prairie voles also display empathy-based consoling behavior
Pair Bonding and Mate Choice toward their distress partner, sibling, or familiar cage mate,
Studies in monogamous prairie voles have revealed the critical which is mediated by OXTR in the anterior cingulate cortex,
roles of OXT and AVP and their receptors in pair bonding, a region involved in empathetic responses in humans (8). Oxtr
biparental care, and mate guarding. For a detailed review of KO prairie voles display altered social preferences and reduced
this topic, see (9). Bonding with a partner is the culmination helping behavior toward a familiar cagemate (27, 147).
of many of the interdependent processes discussed earlier. To
pair bond, mates need to sense their partner’s social cues, be Vasopressin and monogamy
motivated to interact with them, and remember their partner’s AVP stimulates partner preferences as well as selective ag-
identity. Mate guarding is a form of territoriality, while the gression, or mate guarding, in male prairie voles (15, 148).
pair bond has similarities to the maternal bond. Thus, it is not V1aR signaling in the ventral pallidum and LS promote
surprising that OXT and AVP are integrally involved in pair partner preferences (149), while pair bond-induced aggres-
bonding in monogamous species, and mate choices more gen- sion is mediated by V1aR in the lateral hypothalamus (43,
erally. Here we provide a brief overview of OXT’s and AVP’s 150). Viral-mediated Avpr1a receptor overexpression studies
role in pair bonding and mate choice, highlighting recent ad- show that species differences in Avpr1a expression mediate
vances in the context of more classic studies. species differences in social behavior, and variation in Avpr1a
expression in the brain is related to microsatellite diversity in
Oxytocin and monogamy the gene’s 5′ flanking region (151-154). Voles display remark-
Mating and cohabitation stimulates a partner preference in able individual variation in V1aR density in the retrosplenial
both male and female prairie voles. Blocking brain OXT recep- cortex (155), a region involved in spatial memory, and this
tors in either sex prevents pair bonding, potentially disrupting receptor density is related to Avpr1a polymorphisms, space
coordinated activity across a social salience network (137). use in relation to nesting, and sexual fidelity in males (156).
Pharmacological studies revealed that OXT signaling in the The sources of AVP regulating any of these behaviors have
prefrontal cortex and NAcc facilitate mating-induced pair not been explored.
Endocrinology, 2022, Vol. 163, No. 9 7

Mate choice compromised. OXT has been more intensively investigated in


Medaka fish are promiscuous breeders of which females this regard, and its therapeutic potential is related to its modu-
prefer to mate with familiar males that manage to maintain lation of social motivation and the salience of social stimuli
proximity to the female despite efforts of competing males, (19-21). Single-cell transcriptomics studies show that PVN
while males mate indiscriminately. Females with targeted Oxt OXT neurons projecting within the brain that mediate social
or Oxtr mutations lose preference for familiar males and reward behaviors are disrupted in a mouse model of ASD,
mate indiscriminately, while Oxt or Oxtr mutant males pref- and those neurons are enriched for ASD-risk genes (159).
erentially mate with familiar females. Thus, OXTR signaling Further transcriptomic work exploring the function of sub-
promotes the adaptive mate preference of particular species, types of OXT and AVP-expressing cells (160) would be useful
in delineating relevant subcircuits driving social reward. In

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


which in promiscuous species may be sexually dimorphic
(28). In zebrafish, OXT and OXTR drive social behavior in a another ASD mouse model, OXTR signaling in VTA dopa-
context-dependent manner, suggesting a broad role of OXT/ mine neurons, which mediate social reward, is disrupted, and
OXTR in social grouping (157, 158). Like voles, both AVP a kinase inhibitor that restores OXTR signaling also restores
and V1aR homologues in medaka fish mediate mate guarding disrupted ASD-relevant social behaviors (161). Prairie vole
behavior, which is related to territorial behaviors. Thus, both studies suggest that pharmacologically evoking OXT using
OXT and AVP mediate adaptive sociosexual behaviors across melanocortin agonists may be another strategy for enhancing
vertebrate taxa, although the behavior that is adaptive may social functioning (162). Clinically, the efficacy of intranasally
differ across species. delivered OXT has been mixed, and there is a critical need for
clinical trials to be designed based on our understanding of
Translational Opportunities OXT’s function in the brain, such as enhancing the salience of
Because of the effects of OXT and AVP on social behaviors, social stimuli (20).
both peptide systems are potential therapeutic targets for Both AVP and V1aR antagonists have been explored as
treating disorders such as ASD, in which social functioning is potential ASD, schizophrenia, and drug-abuse therapies

Figure 2. Oxytocin (OXT) and arginine vasopressin (AVP) circuits that regulate social behavior. The paraventricular nucleus (PVN) and bed nucleus of the
stria terminalis (BNST) contain OXT and AVP cell bodies that directly influence specific social behaviors: social recognition, social approach and reward,
social communication, sexual behavior, parental behavior, and pair bonding and mate choice. A, PVN and BNST OXT circuits. B, PVN and BNST AVP
circuits. Some contributions of AVP sources and sites of action are unknown, indicated by a short black arrow and “?”. All systems are not depicted for
clarity.
8 Endocrinology, 2022, Vol. 163, No. 9

(163). Based on a subset of preclinical studies demonstrating Data Availability


anxiogenic and agonistic effects of AVP, the V1aR antagonist
Data sharing is not applicable to this article because no data
balovaptan has been tested in clinical trials in ASD indi-
sets were generated or analyzed during the present study.
viduals with mixed effects on social end points (164, 165).
Similarly, male rhesus macaques in large groups that have
naturally impaired social functioning have decreased AVP in References
cerebrospinal fluid (166). This led to investigations in pedi- 1. Leigh-Hunt N, Bagguley D, Bash K, et al. An overview of system-
atric ASD and control samples that also found decreased atic reviews on the public health consequences of social isolation
cerebrospinal fluid AVP (167, 168). Finally, intranasal AVP and loneliness. Public Health. 2017;152:157-171. doi:10.1016/j.
treatment considerably improved social abilities in children

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


puhe.2017.07.035
with ASD (169). 2. Kennedy DP, Adolphs R. The social brain in psychiatric and
Translational opportunities for OXT and AVP may extend neurological disorders. Trends Cogn Sci. 2012;16(11):559-572.
beyond mental health as polymorphisms in V1aR and OXTR doi:10.1016/j.tics.2012.09.006
have been linked to variations in normal as well as disordered 3. Bilezikjian LM, Vale WW. Regulation of ACTH secretion from
corticotrophs: the interaction of vasopressin and CRF. Ann N
human behavior (170). Social relationships are important for
Y Acad Sci. 1987;512:85-96. doi:10.1111/j.1749-6632.1987.
physical well-being as well (171). A recent lung cancer study
tb24952.x
found that a factor in serum samples of bonded monogamous 4. Lee HJ, Macbeth AH, Pagani JH, Young WS III. Oxytocin: the
California mice decreased oncogenic potential of tumor cells great facilitator of life. Prog Neurobiol. 2009;88(2):127-151.
when compared to serum samples from bond-disrupted ani- doi:10.1016/j.pneurobio.2009.04.001
mals (172). Identifying the molecular mechanisms by which 5. Stoop R. Neuromodulation by oxytocin and vasopressin. Neuron.
positive social relationships are transduced to improved 2012;76(1):142-159. doi:10.1016/j.neuron.2012.09.025
health and reduce social stress could lead to novel approaches 6. Johnson ZV, Young LJ. Oxytocin and vasopressin neural net-
for improving well-being, and as OXT and AVP are inextric- works: implications for social behavioral diversity and transla-
ably involved in social relationships, they may have unfore- tional neuroscience. Neurosci Biobehav Rev. 2017;76(Pt A):87-98.
doi:10.1016/j.neubiorev.2017.01.034
seen translational opportunities based on their function in the
7. de Wied D. The influence of the posterior and intermediate lobe
brain (173).
of the pituitary and pituitary peptides on the maintenance of a
conditioned avoidance response in rats. Int J Neuropharmacol.
1965;4:157-167. doi:10.1016/0028-3908(65)90005-5
Concluding Remarks 8. Burkett JP, Andari E, Johnson ZV, Curry DC, de Waal FBM,
Modern molecular approaches are greatly enhancing our Young LJ. Oxytocin-dependent consolation behavior in rodents.
understanding of how and where OXT and AVP act to regu- Science. 2016;351(6271):375-378. doi:10.1126/science.aac4785
late social processes. Despite this progress, considerably more 9. Walum H, Young LJ. The neural mechanisms and circuitry of the
pair bond. Nat Rev Neurosci. 2018;19(11):643-654. doi:10.1038/
attention has been directed toward the OXT system than the
s41583-018-0072-6
AVP system in the past decade because of its perceived trans-
10. Froemke RC, Young LJ. Oxytocin, neural plasticity, and social
lational applications. However, more recent translational re- behavior. Annu Rev Neurosci. 2021;44:359-381. doi:10.1146/
search suggests that both peptides have important clinical annurev-neuro-102320-102847
potential. Consequently, it will be important to rebalance 11. Carter CS. Oxytocin and sexual behavior. Neurosci Biobehav Rev.
OXT/AVP research in behavioral neuroscience. Specifically, 1992;16(2):131-144. doi:10.1016/s0149-7634(05)80176-9
more attention needs to be devoted to understanding the con- 12. Carter CS, DeVries AC, Getz LL. Physiological substrates of mam-
tribution of different peptide sources and their specific targets, malian monogamy: the prairie vole model. Neurosci Biobehav Rev.
particularly with respect to AVP (Fig. 2). Finally, the incorp- 1995;19(2):303-314. doi:10.1016/0149-7634(94)00070-h
oration of modern molecular genetic approaches into studies 13. Kendrick KM, Da Costa AP, Broad KD, et al. Neural control of ma-
ternal behaviour and olfactory recognition of offspring. Brain Res
of alternative model organisms with diverse social systems is
Bull. 1997;44(4):383-395. doi:10.1016/s0361-9230(97)00218-9
essential for gaining a complete understanding of OXT and
14. Grinevich V, Knobloch-Bollmann HS, Eliava M, Busnelli M,
AVP function in the brain, and thereby maximizing transla- Chini B. Assembling the puzzle: pathways of oxytocin signaling
tional opportunities (19, 174). in the brain. Biol Psychiatry. 2016;79(3):155-164. doi:10.1016/j.
biopsych.2015.04.013
15. Nair HP, Young LJ. Vasopressin and pair-bond formation: genes
Acknowledgments to brain to behavior. Physiology (Bethesda). 2006;21:146-152.
We thank Dr Elliott Albers for helpful comments on the doi:10.1152/physiol.00049.2005
16. Albers HE. Species, sex and individual differences in the vasotocin/
manuscript. Figures were created with BioRender.com.
vasopressin system: relationship to neurochemical signaling in
the social behavior neural network. Front Neuroendocrinol.
2015;36:49-71. doi:10.1016/j.yfrne.2014.07.001
Financial Support 17. Arakawa H, Higuchi Y. Exocrine scent marking: coordinative
This work was supported by the National Institutes of role of arginine vasopressin in the systemic regulation of social
Health (grant Nos. R01MH115831, P50MH100023, signaling behaviors. Neurosci Biobehav Rev. 2022;136:104597.
and R01MH112788 to L.J.Y.; R01 MH121603 and R03 doi:10.1016/j.neubiorev.2022.104597
MH120549 to A.P. and G.J.D.; and F31 MH125659 to N.R.). 18. Rilling JK, Young LJ. The biology of mammalian parenting and its ef-
fect on offspring social development. Science. 2014;345(6198):771-
776. doi:10.1126/science.1252723
19. Ford CL, Young LJ. Translational opportunities for circuit-based
Disclosures social neuroscience: advancing 21st century psychiatry. Curr Opin
The authors have nothing to disclose. Neurobiol. 2021;68:1-8. doi:10.1016/j.conb.2020.11.007
Endocrinology, 2022, Vol. 163, No. 9 9

20. Ford CL, Young LJ. Refining oxytocin therapy for autism: con- 39. Grinevich V, Ludwig M. The multiple faces of the oxytocin
text is key. Nat Rev Neurol. 2022;18(2):67-68. doi:10.1038/ and vasopressin systems in the brain. J Neuroendocrinol.
s41582-021-00602-9 2021;33(11):e13004.
21. DeMayo MM, Young LJ, Hickie IB, Song YJC, Guastella AJ. 40. Grinevich V, Neumann ID. Brain oxytocin: how puzzle stones
Circuits for social learning: a unified model and application to from animal studies translate into psychiatry. Mol Psychiatry.
autism spectrum disorder. Neurosci Biobehav Rev. 2019;107:388- 2021;26(1):265-279. doi:10.1038/s41380-020-0802-9
398. doi:10.1016/j.neubiorev.2019.09.034 41. Oti T, Satoh K, Uta D, et al. Oxytocin influences male sexual ac-
22. Kelly AA. consideration of brain networks modulating so- tivity via non-synaptic axonal release in the spinal cord. Curr Biol.
cial behavior. Horm Behav. 2022;141:105138. doi:10.1016/j. 2021;31(1):103-114.e5. doi:10.1016/j.cub.2020.09.089
yhbeh.2022.105138 42. Rohr KE, Telega A, Savaglio A, Evans JA. Vasopressin regulates

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


23. Boender AJ, Young LJ. Oxytocin, vasopressin and social behavior daily rhythms and circadian clock circuits in a manner influenced
in the age of genome editing: a comparative perspective. Horm by sex. Horm Behav. 2021;127:104888. doi:10.1016/j.
Behav. 2020;124:104780. doi:10.1016/j.yhbeh.2020.104780 yhbeh.2020.104888
24. Silva AC, Pandolfi M. Vasotocinergic control of agonistic behavior 43. Gobrogge KL, Liu Y, Young LJ, Wang Z. Anterior hypotha-
told by neotropical fishes. Gen Comp Endocrinol. 2019;273:67- lamic vasopressin regulates pair-bonding and drug-induced ag-
72. doi:10.1016/j.ygcen.2018.04.025 gression in a monogamous rodent. Proc Natl Acad Sci U S A.
25. Campos SM, Belkasim SS. Chemical communication in lizards and 2009;106(45):19144-19149. doi:10.1073/pnas.0908620106
a potential role for vasotocin in modulating social interactions. 44. Jirikowski GF. Diversity of central oxytocinergic projections.
Integr Comp Biol. 2021;61(1):205-220. doi:10.1093/icb/icab044 Cell Tissue Res. 2019;375(1):41-48. doi:10.1007/s00441-018-
26. Goodson JL, Schrock SE, Klatt JD, Kabelik D, Kingsbury MA. 2960-5
Mesotocin and nonapeptide receptors promote estrildid flocking 45. Scott Young W III, Gainer H. Transgenesis and the study of ex-
behavior. Science. 2009;325(5942):862-866. doi:10.1126/ pression, cellular targeting and function of oxytocin, vasopressin
science.1174929 and their receptors. Neuroendocrinology. 2003;78(4):185-203.
27. Horie K, Inoue K, Suzuki S, et al. Oxytocin receptor knockout doi:10.1159/000073702
prairie voles generated by CRISPR/Cas9 editing show reduced 46. Gillies GE, Linton EA, Lowry PJ. Corticotropin releasing activity of
preference for social novelty and exaggerated repetitive behaviors. the new CRF is potentiated several times by vasopressin. Nature.
Horm Behav. 2019;111:60-69. doi:10.1016/j.yhbeh.2018.10.011 1982;299(5881):355-357. doi:10.1038/299355a0
28. Yokoi S, Naruse K, Kamei Y, et al. Sexually dimorphic role 47. Castel M, Morris JF. The neurophysin-containing innervation of
of oxytocin in medaka mate choice. Proc Natl Acad Sci U S A. the forebrain of the mouse. Neuroscience. 1988;24(3):937-966.
2020;117(9):4802-4808. doi:10.1073/pnas.1921446117 doi:10.1016/0306-4522(88)90078-4
29. Yokoi S, Okuyama T, Kamei Y, et al. An essential role of the ar- 48. Ludwig M, Leng G. Dendritic peptide release and peptide-
ginine vasotocin system in mate-guarding behaviors in triadic dependent behaviours. Nat Rev Neurosci. 2006;7(2):126-136.
relationships of medaka fish (Oryzias latipes). PLoS Genet. doi:10.1038/nrn1845
2015;11(2):e1005009. doi:10.1371/journal.pgen.1005009 49. Dumais KM, Veenema AH. Vasopressin and oxytocin receptor sys-
30. Horie K, Inoue K, Nishimori K, Young LJ. Investigation of Oxtr- tems in the brain: sex differences and sex-specific regulation of so-
expressing neurons projecting to nucleus accumbens using Oxtr-ires- cial behavior. Front Neuroendocrinol. 2016;40:1-23. doi:10.1016/j.
Cre knock-in prairie voles (Microtus ochrogaster). Neuroscience. yfrne.2015.04.003
2020;448:312-324. doi:10.1016/j.neuroscience.2020.08.023 50. Taylor PV, Veenema AH, Paul MJ, Bredewold R, Isaacs S,
31. Goodson JL, Bass AH. Social behavior functions and related an- de Vries GJ. Sexually dimorphic effects of a prenatal immune chal-
atomical characteristics of vasotocin/vasopressin systems in lenge on social play and vasopressin expression in juvenile rats.
vertebrates. Brain Res Rev. 2001;35(3):246-265. doi:10.1016/ Biol Sex Differ. 2012;3(1):15. doi:10.1186/2042-6410-3-15
s0165-0173(01)00043-1 51. Häussler HU, Jirikowski GF, Caldwell JD. Sex differences among
32. Landgraf R, Neumann ID. Vasopressin and oxytocin release within oxytocin-immunoreactive neuronal systems in the mouse hypothal-
the brain: a dynamic concept of multiple and variable modes of amus. J Chem Neuroanat. 1990;3(4):271-276.
neuropeptide communication. Front Neuroendocrinol. 2004;25(3- 52. Rigney N, Whylings J, de Vries GJ, Petrulis A. Sex differences in the
4):150-176. doi:10.1016/j.yfrne.2004.05.001 control of social investigation and anxiety by vasopressin cells of the
33. De Vries GJ, Panzica GC. Sexual differentiation of central vaso- paraventricular nucleus of the hypothalamus. Neuroendocrinology.
pressin and vasotocin systems in vertebrates: different mechanisms, 2021;111(6):521-535. doi:10.1159/000509421
similar endpoints. Neuroscience. 2006;138(3):947-955. 53. Knobloch HS, Charlet A, Hoffmann LC, et al. Evoked axonal ox-
doi:10.1016/j.neuroscience.2005.07.050 ytocin release in the central amygdala attenuates fear response.
34. de Vries GJ. Sex differences in vasopressin and oxytocin innerva- Neuron. 2012;73(3):553-566. doi:10.1016/j.neuron.2011.11.030
tion of the brain. Prog Brain Res. 2008;170:17-27. doi:10.1016/ 54. Mitre M, Marlin BJ, Schiavo JK, et al. A distributed network
S0079-6123(08)00402-0 for social cognition enriched for oxytocin receptors. J Neurosci.
35. Donaldson ZR, Young LJ. Oxytocin, vasopressin, and the 2016;36(8):2517-2535. doi:10.1523/JNEUROSCI.2409-15.2016
neurogenetics of sociality. Science. 2008;322(5903):900-904. 55. Zhang B, Qiu L, Xiao W, et al. Reconstruction of the hypothalamo-
doi:10.1126/science.1158668 neurohypophysial system and functional dissection of magnocellular
36. Bredewold R, Veenema AH. Sex differences in the regulation of oxytocin neurons in the brain. Neuron. 2021;109(2):331-346.e7.
social and anxiety-related behaviors: insights from vasopressin and doi:10.1016/j.neuron.2020.10.032
oxytocin brain systems. Curr Opin Neurobiol. 2018;49:132-140. 56. Dobolyi A, Cservenák M, Young LJ. Thalamic integration of
doi:10.1016/j.conb.2018.02.011 social stimuli regulating parental behavior and the oxytocin
37. Smith CJW, DiBenedictis BT, Veenema AH. Comparing vaso- system. Front Neuroendocrinol. 2018;51:102-115. doi:10.1016/j.
pressin and oxytocin fiber and receptor density patterns in the yfrne.2018.05.002
social behavior neural network: implications for cross-system 57. Tang Y, Benusiglio D, Lefevre A, et al. Social touch promotes
signaling. Front Neuroendocrinol. 2019;53:100737. doi:10.1016/j. interfemale communication via activation of parvocellular oxy-
yfrne.2019.02.001 tocin neurons. Nat Neurosci. 2020;23(9):1125-1137. doi:10.1038/
38. Jurek B, Neumann ID. The oxytocin receptor: from intracel- s41593-020-0674-y
lular signaling to behavior. Physiol Rev. 2018;98(3):1805-1908. 58. Yu H, Miao W, Ji E, et al. Social touch-like tactile stimulation
doi:10.1152/physrev.00031.2017 activates a tachykinin 1-oxytocin pathway to promote social
10 Endocrinology, 2022, Vol. 163, No. 9

interactions. Neuron. 2022;110(6):1051-1067.e7. doi:10.1016/j. 76. Tobin VA, Hashimoto H, Wacker DW, et al. An intrinsic vas-
neuron.2021.12.022 opressin system in the olfactory bulb is involved in social rec-
59. Son S, Manjila SB, Newmaster KT, et al. Whole-brain wiring diagram ognition. Nature. 2010;464(7287):413-417. doi:10.1038/
of oxytocin system in adult mice. J Neurosci. 2022;42(25):5021- nature08826
5033. doi:10.1523/JNEUROSCI.0307-22.2022 77. Suyama H, Egger V, Lukas M. Top-down acetylcholine signaling
60. Rood BD, Stott RT, You S, Smith CJW, Woodbury ME, De Vries GJ. via olfactory bulb vasopressin cells contributes to social dis-
Site of origin of and sex differences in the vasopressin inner- crimination in rats. Commun Biol. 2021;4(1):603. doi:10.1038/
vation of the mouse (Mus musculus) brain. J Comp Neurol. s42003-021-02129-7
2013;521(10):2321-2358. doi:10.1002/cne.23288 78. Namba T, Taniguchi M, Murata Y, et al. Activation of arginine vas-
61. Wei HH, Yuan XS, Chen ZK, et al. Presynaptic inputs to vaso- opressin receptor 1a facilitates the induction of long-term potenti-

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


pressin neurons in the hypothalamic supraoptic nucleus and par- ation in the accessory olfactory bulb of male mice. Neurosci Lett.
aventricular nucleus in mice. Exp Neurol. 2021;343:113784. 2016;634:107-113. doi:10.1016/j.neulet.2016.09.056
doi:10.1016/j.expneurol.2021.113784 79. Crawley JN, Chen T, Puri A, et al. Social approach behaviors in
62. Kim A, Madara JC, Wu C, Andermann ML, Lowell BB. Neural basis oxytocin knockout mice: comparison of two independent lines
for regulation of vasopressin secretion by anticipated disturbances tested in different laboratory environments. Neuropeptides.
in osmolality. Elife 2021;10:e66609. doi:10.7554/eLife.66609 2007;41(3):145-163. doi:10.1016/j.npep.2007.02.002
63. DiBenedictis BT, Nussbaum ER, Cheung HK, Veenema AH. 80. Dölen G, Darvishzadeh A, Huang KW, Malenka RC. Social re-
Quantitative mapping reveals age and sex differences in vaso- ward requires coordinated activity of nucleus accumbens oxytocin
pressin, but not oxytocin, immunoreactivity in the rat social be- and serotonin. Nature. 2013;501(7466):179-184. doi:10.1038/
havior neural network. J Comp Neurol. 2017;525(11):2549-2570. nature12518
doi:10.1002/cne.24216 81. Nardou R, Lewis EM, Rothhaas R, et al. Oxytocin-dependent
64. Steinman MQ, Duque-Wilckens N, Greenberg GD, et al. Sex- reopening of a social reward learning critical period with
specific effects of stress on oxytocin neurons correspond with MDMA. Nature. 2019;569(7754):116-120. doi:10.1038/
responses to intranasal oxytocin. Biol Psychiatry. 2016;80(5):406- s41586-019-1075-9
414. doi:10.1016/j.biopsych.2015.10.007 82. Hung LW, Neuner S, Polepalli JS, et al. Gating of social re-
65. Wang Z, Moody K, Newman JD, Insel TR. Vasopressin and ward by oxytocin in the ventral tegmental area. Science.
oxytocin immunoreactive neurons and fibers in the fore- 2017;357(6358):1406-1411. doi:10.1126/science.aan4994
brain of male and female common marmosets (Callithrix 83. Xiao L, Priest MF, Kozorovitskiy Y. Oxytocin functions as a spa-
jacchus). Synapse. 1997;27(1):14-25. doi:10.1002/ tiotemporal filter for excitatory synaptic inputs to VTA dopamine
(SICI)1098-2396(199709)27:1<14::AID-SYN2>3.0.CO;2-G neurons. Elife. 2018;7:e33892. doi:10.7554/eLife.33892
66. Duque-Wilckens N, Torres LY, Yokoyama S, et al. Extrahypothalamic 84. Borland JM, Aiani LM, Norvelle A, et al. Sex-dependent regu-
oxytocin neurons drive stress-induced social vigilance and avoid- lation of social reward by oxytocin receptors in the ventral teg-
ance. Proc Natl Acad Sci U S A. 2020;117(42):26406-26413. mental area. Neuropsychopharmacology. 2019;44(4):785-792.
doi:10.1073/pnas.2011890117 doi:10.1038/s41386-018-0262-y
67. De Vries GJ, Rissman EF, Simerly RB, et al. A model system for study 85. Resendez SL, Namboodiri VMK, Otis JM, et al. Social stimuli
of sex chromosome effects on sexually dimorphic neural and be- induce activation of oxytocin neurons within the paraventric-
havioral traits. J Neurosci. 2002;22(20):9005-9014. doi:10.1523/ ular nucleus of the hypothalamus to promote social behavior in
JNEUROSCI.22-20-09005.2002 male mice. J Neurosci. 2020;40(11):2282-2295. doi:10.1523/
68. French JA, Taylor JH, Mustoe AC, Cavanaugh J. Neuropeptide JNEUROSCI.1515-18.2020
diversity and the regulation of social behavior in New World 86. He Z, Young L, Ma XM, et al. Increased anxiety and decreased
primates. Front Neuroendocrinol. 2016;42:18-39. doi:10.1016/j. sociability induced by paternal deprivation involve the PVN-
yfrne.2016.03.004 PrL OTergic pathway. Elife. 2019;8:e44026. doi:10.7554/
69. Juul KV, Bichet DG, Nielsen S, Nørgaard JP. The physiological and eLife.44026
pathophysiological functions of renal and extrarenal vasopressin 87. Peris J, MacFadyen K, Smith JA, de Kloet AD, Wang L, Krause EG.
V2 receptors. Am J Physiol Renal Physiol. 2014;306(9):F931 Oxytocin receptors are expressed on dopamine and glutamate
-F940. doi:10.1152/ajprenal.00604.2013 neurons in the mouse ventral tegmental area that project to nu-
70. Rogers Flattery CN, Coppeto DJ, Inoue K, Rilling JK, Preuss TM, cleus accumbens and other mesolimbic targets. J Comp Neurol.
Young LJ. Distribution of brain oxytocin and vasopressin 2017;525(5):1094-1108. doi:10.1002/cne.24116
V1a receptors in chimpanzees (Pan troglodytes): comparison 88. Duque-Wilckens N, Steinman MQ, Busnelli M, et al. Oxytocin
with humans and other primate species. Brain Struct Funct. receptors in the anteromedial bed nucleus of the stria terminalis
2022;227(5):1907-1919. doi:10.1007/s00429-021-02369-7 promote stress-induced social avoidance in female California
71. Inoue K, Ford CL, Horie K, Young LJ. Oxytocin receptors are mice. Biol Psychiatry. 2018;83(3):203-213. doi:10.1016/j.
widely distributed in prairie vole (Microtus ochrogaster) brain: biopsych.2017.08.024
relation to social behavior, genetic polymorphisms, and the do- 89. Kelly AM, Goodson JL. Social functions of individual vasopressin-
pamine system. J Comp Neurol. Published online June 28, 2022. oxytocin cell groups in vertebrates: what do we really know?
doi:10.1002/cne.25382 Front Neuroendocrinol. 2014;35(4):512-529. doi:10.1016/j.
72. Luo PX, Zakharenkov HC, Torres LY, et al. Oxytocin receptor yfrne.2014.04.005
behavioral effects and cell types in the bed nucleus of the stria 90. Rigney N, Whylings J, Mieda M, de Vries G, Petrulis A. Sexually
terminalis. Horm Behav. 2022;143:105203. doi:10.1016/j. dimorphic vasopressin cells modulate social investigation and
yhbeh.2022.105203 communication in sex-specific ways. eNeuro. 2019;6(1):ENE
73. Wyatt TD. Pheromones and Animal Behaviour. Cambridge URO.0415-18.2019. doi:10.1523/ENEURO.0415-18.2019
University Press; 2003. 91. Rigney N, Zbib A, Vries GJ, Petrulis A. Knockdown of sexually
74. Ferguson JN, Aldag JM, Insel TR, Young LJ. Oxytocin in the me- differentiated vasopressin expression in the bed nucleus of the stria
dial amygdala is essential for social recognition in the mouse. J terminalis reduces social and sexual behavior in male, but not fe-
Neurosci. 2001;21(20):8278-8285. male, mice. J Neuroendocrinol. Published online December 22,
75. Oettl LL, Ravi N, Schneider M, et al. Oxytocin enhances so- 2021. doi:10.1111/jne.13083
cial recognition by modulating cortical control of early olfac- 92. Islam MT, Maejima T, Matsui A, Mieda M. Paraventricular hypo-
tory processing. Neuron. 2016;90(3):609-621. doi:10.1016/j. thalamic vasopressin neurons induce self-grooming in mice. Mol
neuron.2016.03.033 Brain. 2022;15(1):47. doi:10.1186/s13041-022-00932-9
Endocrinology, 2022, Vol. 163, No. 9 11

93. Aspesi D, Choleris E. Neuroendocrine underpinning of so- behavior in golden hamsters. Science. 1984;224(4648):521-523.
cial recognition in males and females. J Neuroendocrinol. doi:10.1126/science.6538700
2021;34(2):e13070. doi:10.1111/jne.13070 112. Albers HE, Hennessey AC, Whitman DC. Vasopressin and
94. Bielsky IF, Hu SB, Ren X, Terwilliger EF, Young LJ. The V1a vas- the regulation of hamster social behavior. Ann N Y Acad Sci.
opressin receptor is necessary and sufficient for normal social rec- 1992;652:227-242. doi:10.1111/j.1749-6632.1992.tb34358.x
ognition: a gene replacement study. Neuron. 2005;47(4):503-513. 113. Ferris CF, Albers HE, Wesolowski SM, Goldman BD, Luman SE.
doi:10.1016/j.neuron.2005.06.031 Vasopressin injected into the hypothalamus triggers a stereotypic
95. Wersinger SR, Temple JL, Caldwell HK, Young WS III. behavior in golden hamsters. Science. 1984;224(4648):521-523.
Inactivation of the oxytocin and the vasopressin (Avp) 1b receptor doi:10.1126/science.6538700
genes, but not the Avp 1a receptor gene, differentially impairs the 114. Terranova JI, Ferris CF, Albers HE. Sex differences in the reg-

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


Bruce effect in laboratory mice (Mus musculus). Endocrinology. ulation of offensive aggression and dominance by arginine-
2008;149(1):116-121. doi:10.1210/en.2007-1056 vasopressin. Front Endocrinol. 2017;8:308.
96. Yao S, Bergan J, Lanjuin A, Dulac C. Oxytocin signaling in the 115. Hurst JL, Beynon RJ. Scent wars: the chemobiology of com-
medial amygdala is required for sex discrimination of social cues. petitive signalling in mice. Bioessays. 2004;26(12):1288-1298.
Elife. 2017;6:e31373. doi:10.7554/eLife.31373 doi:10.1002/bies.20147
97. Zhang L, Hernández VS. Synaptic innervation to rat hippo- 116. Ferris CF, Melloni RH Jr, Koppel G, Perry KW, Fuller RW,
campus by vasopressin-immuno-positive fibres from the hypo- Delville Y. Vasopressin/serotonin interactions in the anterior hy-
thalamic supraoptic and paraventricular nuclei. Neuroscience. pothalamus control aggressive behavior in golden hamsters. J
2013;228:139-162. doi:10.1016/j.neuroscience.2012.10.010 Neurosci. 1997;17(11):4331-4340.
98. Cui Z, Gerfen CR, Young WS III. Hypothalamic and other 117. Gutzler SJ, Karom M, Erwin WD, Albers HE. Arginine-vasopressin
connections with dorsal CA2 area of the mouse hippocampus. J and the regulation of aggression in female Syrian hamsters
Comp Neurol. 2013;521(8):1844-1866. doi:10.1002/cne.23263 (Mesocricetus auratus). Eur J Neurosci. 2010;31(9):1655-1663.
99. Smith AS, Williams Avram SK, Cymerblit-Sabba A, Song J, doi:10.1111/j.1460-9568.2010.07190.x
Young WS. Targeted activation of the hippocampal CA2 118. Taylor JH, Walton JC, McCann KE, et al. CRISPR-Cas9 editing
area strongly enhances social memory. Mol Psychiatry. of the arginine-vasopressin V1a receptor produces paradoxical
2016;21(8):1137-1144. doi:10.1038/mp.2015.189 changes in social behavior in Syrian hamsters. Proc Natl Acad Sci
100. Tirko NN, Eyring KW, Carcea I, et al. Oxytocin transforms firing U S A. 2022;119(19):e2121037119.
mode of CA2 hippocampal neurons. Neuron. 2018;100(3):593- 119. Ho JM, Murray JH, Demas GE, Goodson JL. Vasopressin cell
608.e3. doi:10.1016/j.neuron.2018.09.008 groups exhibit strongly divergent responses to copulation and
101. Raam T, McAvoy KM, Besnard A, Veenema AH, Sahay A. male-male interactions in mice. Horm Behav. 2010;58(3):368-
Hippocampal oxytocin receptors are necessary for discrimination 377. doi:10.1016/j.yhbeh.2010.03.021
of social stimuli. Nat Commun. 2017;8(1):2001. doi:10.1038/ 120. Baum MJ, Vreeburg JT. Copulation in castrated male rats fol-
s41467-017-02173-0 Erratum in: Nat Commun. 2018;9(1):552. lowing combined treatment with estradiol and dihydrotes-
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5722862/ tosterone. Science. 1973;182(4109):283-285. doi:10.1126/
102. Boyle L, Posani L, Irfan S, Siegelbaum SA, Fusi S. The ge- science.182.4109.283
ometry of hippocampal CA2 representations enables ab- 121. de Vries GJ, Buijs RM, Sluiter AA. Gonadal hormone
stract coding of social familiarity and identity. bioRXiv. actions on the morphology of the vasopressinergic innerva-
doi:10.1101/2022.01.24.477361. tion of the adult rat brain. Brain Res. 1984;298(1):141-145.
103. Cymerblit-Sabba A, Smith AS, Williams Avram SK, et al. Inducing doi:10.1016/0006-8993(84)91157-0
partner preference in mice by chemogenetic stimulation of 122. Valtcheva S, Froemke RC. Neuromodulation of maternal circuits
CA2 hippocampal subfield. Front Mol Neurosci. 2020;13:61. by oxytocin. Cell Tissue Res. 2019;375(1):57-68. doi:10.1007/
doi:10.3389/fnmol.2020.00061 s00441-018-2883-1
104. Ferretti V, Maltese F, Contarini G, et al. Oxytocin signaling in the 123. Marlin BJ, Mitre M, D’amour JA, Chao MV, Froemke RC.
central amygdala modulates emotion discrimination in mice. Curr Oxytocin enables maternal behaviour by balancing cortical
Biol. 2019;29(12):1938-1953.e6. doi:10.1016/j.cub.2019.04.070 inhibition. Nature. 2015;520(7548):499-504. doi:10.1038/
105. Anpilov S, Shemesh Y, Eren N, et al. Wireless optogenetic stim- nature14402
ulation of oxytocin neurons in a semi-natural setup dynami- 124. Schiavo JK, Valtcheva S, Bair-Marshall CJ, Song SC, Martin KA,
cally elevates both pro-social and agonistic behaviors. Neuron. Froemke RC. Innate and plastic mechanisms for maternal
2020;107(4):644-655.e7. doi:10.1016/j.neuron.2020.05.028 behaviour in auditory cortex. Nature. 2020;587(7834):426-431.
106. Cilz NI, Cymerblit-Sabba A, Young WS. Oxytocin and vas- doi:10.1038/s41586-020-2807-6
opressin in the rodent hippocampus. Genes Brain Behav. 125. Carcea I, Caraballo NL, Marlin BJ, et al. Oxytocin neurons
2019;18(1):e12535. doi:10.1111/gbb.12535 enable social transmission of maternal behaviour. Nature.
107. Watanabe J, Takayanagi Y, Yoshida M, et al. Conditional abla- 2021;596(7873):553-557. doi:10.1038/s41586-021-03814-7
tion of vasopressin-synthesizing neurons in transgenic rats. J 126. He Z, Zhang L, Hou W, et al. Paraventricular nucleus oxy-
Neuroendocrinol. 2021;33(12):e13057. tocin subsystems promote active paternal behaviors in man-
108. Whylings J, Rigney N, Peters NV, de Vries GJ, Petrulis A. Sexually darin voles. J Neurosci. 2021;41(31):6699-6713. doi:10.1523/
dimorphic role of BNST vasopressin cells in sickness and so- JNEUROSCI.2864-20.2021
cial behavior in male and female mice. Brain Behav Immun. 127. Zheng JJ, Li SJ, Zhang XD, et al. Oxytocin mediates early
2020;83:68-77. doi:10.1016/j.bbi.2019.09.015 experience-dependent cross-modal plasticity in the sensory cor-
109. Veenema AH, Bredewold R, De Vries GJ. Vasopressin regulates tices. Nat Neurosci. 2014;17(3):391-399. doi:10.1038/nn.3634
social recognition in juvenile and adult rats of both sexes, but 128. Feng T, An S, Kinden R, Zhang X, Jia R, Tai F. Alteration in oxy-
in sex- and age-specific ways. Horm Behav. 2012;61(1):50-56. tocin levels induced by early social environment affects maternal
doi:10.1016/j.yhbeh.2011.10.002 behavior and estrogen receptor alpha in mandarin voles (Microtus
110. Borie AM, Dromard Y, Guillon G, et al. Correction of vasopressin mandarinus). Behav Brain Res. 2019;365:36-47. doi:10.1016/j.
deficit in the lateral septum ameliorates social deficits of mouse bbr.2019.02.038
autism model. J Clin Invest. 2021;131(2):e144450. doi:10.1172/ 129. Li T, Jia SW, Hou D, et al. Intranasal oxytocin restores maternal
JCI144450 behavior and oxytocin neuronal activity in the supraoptic nucleus
111. Ferris CF, Albers HE, Wesolowski SM, Goldman BD, Luman SE. in rat dams with cesarean delivery. Neuroscience. 2021;468:235-
Vasopressin injected into the hypothalamus triggers a stereotypic 246. doi:10.1016/j.neuroscience.2021.06.020
12 Endocrinology, 2022, Vol. 163, No. 9

130. Baracz SJ, Everett NA, Robinson KJ, Campbell GR, Cornish JL. 147. Kitano K, Yamagishi A, Horie K, Nishimori K, Sato N. Helping
Maternal separation changes maternal care, anxiety‐like behavior in prairie voles: a model of empathy and the impor-
behaviour and expression of paraventricular oxytocin and tance of oxytocin. iScience. 2022;25(4):103991. doi:10.1016/j.
corticotrophin‐releasing factor immunoreactivity in lactating rats. isci.2022.103991
J Neuroendocrinol. 2020;32(6):e12861. doi:10.1111/jne.12861 148. Donaldson ZR, Spiegel L, Young LJ. Central vasopressin V1a
131. Cutuli D, Berretta E, Caporali P, et al. Effects of pre-reproductive receptor activation is independently necessary for both partner
maternal enrichment on maternal care, offspring’s play be- preference formation and expression in socially monogamous
havior and oxytocinergic neurons. Neuropharmacology. male prairie voles. Behav Neurosci. 2010;124(1):159-163.
2019;145(Pt A):99-113. doi:10.1016/j.neuropharm.2018.02.015 doi:10.1037/a0018094
132. Pedersen CA, Ascher JA, Monroe YL, Prange AJ Jr. Oxytocin 149. Young LJ, Wang Z. The neurobiology of pair bonding. Nat

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


induces maternal behavior in virgin female rats. Science. Neurosci. 2004;7(10):1048-1054. doi:10.1038/nn1327
1982;216(4546):648-650. doi:10.1126/science.7071605 150. Barrett CE, Keebaugh AC, Ahern TH, Bass CE, Terwilliger EF,
133. Bendesky A, Kwon YM, Lassance JM, et al. The genetic basis Young LJ. Variation in vasopressin receptor (Avpr1a) expres-
of parental care evolution in monogamous mice. Nature. sion creates diversity in behaviors related to monogamy in
2017;544(7651):434-439. doi:10.1038/nature22074 prairie voles. Horm Behav. 2013;63(3):518-526. doi:10.1016/j.
134. Bayerl DS, Bosch OJ. Brain vasopressin signaling modulates yhbeh.2013.01.005
aspects of maternal behavior in lactating rats. Genes Brain Behav. 151. Donaldson ZR, Young LJ. The relative contribution of prox-
2019;18(1):e12517. doi:10.1111/gbb.12517 imal 5′ flanking sequence and microsatellite variation on brain
135. Wang Z, Ferris CF, De Vries GJ. Role of septal vasopressin innerva- vasopressin 1a receptor (Avpr1a) gene expression and be-
tion in paternal behavior in prairie voles (Microtus ochrogaster). havior. PLoS Genet. 2013;9(8):e1003729. doi:10.1371/journal.
Proc Natl Acad Sci U S A. 1994;91(1):400-404. doi:10.1073/ pgen.1003729
pnas.91.1.400 152. Lim MM, Wang Z, Olazábal DE, Ren X, Terwilliger EF,
136. Bosch OJ, Neumann ID. Brain vasopressin is an important reg- Young LJ. Enhanced partner preference in a promiscuous spe-
ulator of maternal behavior independent of dams’ trait anx- cies by manipulating the expression of a single gene. Nature.
iety. Proc Natl Acad Sci U S A. 2008;105(44):17139-17144. 2004;429(6993):754-757. doi:10.1038/nature02539
doi:10.1073/pnas.0807412105 153. Hammock EAD, Young LJ. Microsatellite instability generates
137. Johnson ZV, Walum H, Jamal YA, et al. Central oxytocin receptors diversity in brain and sociobehavioral traits. Science.
mediate mating-induced partner preferences and enhance 2005;308(5728):1630-1634. doi:10.1126/science.1111427
correlated activation across forebrain nuclei in male prairie voles. 154. Young LJ, Nilsen R, Waymire KG, MacGregor GR, Insel TR.
Horm Behav. 2016;79:8-17. doi:10.1016/j.yhbeh.2015.11.011 Increased affiliative response to vasopressin in mice expressing
138. Keebaugh AC, Barrett CE, Laprairie JL, Jenkins JJ, Young LJ. the V1a receptor from a monogamous vole. Nature.
RNAi knockdown of oxytocin receptor in the nucleus accumbens 1999;400(6746):766-768. doi:10.1038/23475
inhibits social attachment and parental care in monogamous fe- 155. Phelps SM, Young LJ. Extraordinary diversity in vasopressin
male prairie voles. Soc Neurosci. 2015;10(5):561-570. doi:10.10 (V1a) receptor distributions among wild prairie voles (Microtus
80/17470919.2015.1040893 ochrogaster): patterns of variation and covariation. J Comp
139. Keebaugh AC, Young LJ. Increasing oxytocin receptor expres- Neurol. 2003;466(4):564-576. doi:10.1002/cne.10902
sion in the nucleus accumbens of pre-pubertal female prairie 156. Okhovat M, Berrio A, Wallace G, Ophir AG, Phelps SM. Sexual
voles enhances alloparental responsiveness and partner prefer- fidelity trade-offs promote regulatory variation in the prairie vole
ence formation as adults. Horm Behav. 2011;60(5):498-504. brain. Science. 2015;350(6266):1371-1374. doi:10.1126/science.
doi:10.1016/j.yhbeh.2011.07.018 aac5791
140. Borie AM, Agezo S, Lunsford P, et al. Social experience alters 157. Gemmer A, Mirkes K, Anneser L, et al. Oxytocin receptors in-
oxytocinergic modulation in the nucleus accumbens of female fluence the development and maintenance of social behavior in
prairie voles. Curr Biol. 2022;32(5):1026-1037.e4. doi:10.1016/j. zebrafish (Danio rerio). Sci Rep. 2022;12(1):4322. doi:10.1038/
cub.2022.01.014 s41598-022-07990-y
141. Ross HE, Cole CD, Smith Y, et al. Characterization of the ox- 158. Nunes AR, Gliksberg M, Varela SAM, et al. Developmental effects
ytocin system regulating affiliative behavior in female prairie of oxytocin neurons on social affiliation and processing of social
voles. Neuroscience. 2009;162(4):892-903. doi:10.1016/j. information. J Neurosci. 2021;41(42):8742-8760. doi:10.1523/
neuroscience.2009.05.055 JNEUROSCI.2939-20.2021
142. Barrett CE, Arambula SE, Young LJ. The oxytocin system 159. Lewis EM, Stein-O’Brien GL, Patino AV, et al. Parallel so-
promotes resilience to the effects of neonatal isolation on adult cial information processing circuits are differentially impacted
social attachment in female prairie voles. Transl Psychiatry. in autism. Neuron. 2020;108(4):659-675.e6. doi:10.1016/j.
2015;5:e606. doi:10.1038/tp.2015.73 neuron.2020.10.002
143. Ahern TH, Olsen S, Tudino R, Beery AK. Natural variation in the 160. Romanov RA, Zeisel A, Bakker J, et al. Molecular interrogation
oxytocin receptor gene and rearing interact to influence reproduc- of hypothalamic organization reveals distinct dopamine neu-
tive and nonreproductive social behavior and receptor binding. ronal subtypes. Nat Neurosci. 2017;20(2):176-188. doi:10.1038/
Psychoneuroendocrinology. 2021;128:105209. doi:10.1016/j. nn.4462
psyneuen.2021.105209 161. Hörnberg H, Pérez-Garci E, Schreiner D, et al. Rescue of oxytocin
144. King LB, Walum H, Inoue K, Eyrich NW, Young LJ. Variation in response and social behaviour in a mouse model of autism. Nature.
the oxytocin receptor gene predicts brain region-specific expres- 2020;584(7820):252-256. doi:10.1038/s41586-020-2563-7
sion and social attachment. Biol Psychiatry. 2016;80(2):160-169. 162. Modi ME, Inoue K, Barrett CE, et al. Melanocortin receptor
doi:10.1016/j.biopsych.2015.12.008 agonists facilitate oxytocin-dependent partner preference
145. Scheele D, Wille A, Kendrick KM, et al. Oxytocin enhances brain formation in the prairie vole. Neuropsychopharmacology.
reward system responses in men viewing the face of their female 2015;40(8):1856-1865. doi:10.1038/npp.2015.35
partner. Proc Natl Acad Sci U S A. 2013;110(50):20308-20313. 163. Rae M, Lemos Duarte M, Gomes I, Camarini R, Devi LA. Oxytocin
doi:10.1073/pnas.1314190110 and vasopressin: signalling, behavioural modulation and poten-
146. Pohl TT, Young LJ, Bosch OJ. Lost connections: oxytocin and the tial therapeutic effects. Br J Pharmacol. 2022;179(8):1544-1564.
neural, physiological, and behavioral consequences of disrupted doi:10.1111/bph.15481
relationships. Int J Psychophysiol. 2019;136:54-63. doi:10.1016/j. 164. Jacob S, Veenstra-VanderWeele J, Murphy D, et al. Efficacy
ijpsycho.2017.12.011 and safety of balovaptan for socialisation and communication
Endocrinology, 2022, Vol. 163, No. 9 13

difficulties in autistic adults in North America and Europe: a improves social deficits in children with autism. Sci Transl
phase 3, randomised, placebo-controlled trial. Lancet Psychiatry. Med. 2019;11(491):eaau7356. doi:10.1126/scitranslmed.
2022;9(3):199-210. doi:10.1016/S2215-0366(21)00429-6 aau7356
165. Bolognani F, Del Valle Rubido M, Squassante L, et al. A phase 2 clin- 170. Aspé-Sánchez M, Moreno M, Rivera MI, Rossi A, Ewer J.
ical trial of a vasopressin V1a receptor antagonist shows improved Oxytocin and vasopressin receptor gene polymorphisms: role
adaptive behaviors in men with autism spectrum disorder. Sci Transl in social and psychiatric traits. Front Neurosci. 2015;9:510.
Med. 2019;11(491):eaat7838. doi:10.1126/scitranslmed.aat7838 doi:10.3389/fnins.2015.00510
166. Parker KJ, Garner JP, Oztan O, et al. Arginine vasopressin in cere- 171. Bosch OJ, Young LJ. Oxytocin and social relationships: from
brospinal fluid is a marker of sociality in nonhuman primates. Sci attachment to bond disruption. Curr Top Behav Neurosci.
Transl Med. 2018;10(439):eaam9100. doi:10.1126/scitranslmed. 2018;35:97-117. doi:10.1007/7854_2017_10

Downloaded from https://academic.oup.com/endo/article/163/9/bqac111/6648172 by Georgia State University user on 12 October 2022


aam9100 172. Naderi A, Soltanmaohammadi E, Kaza V, Barlow S,
167. Oztan O, Garner JP, Constantino JN, Parker KJ. Neonatal Chatzistamou I, Kiaris H. Persistent effects of pair bonding in
CSF vasopressin concentration predicts later medical record lung cancer cell growth in monogamous Peromyscus californicus.
diagnoses of autism spectrum disorder. Proc Natl Acad Sci U S A. Elife. 2021;10:e64711. doi:10.7554/eLife.64711
2020;117(19):10609-10613. doi:10.1073/pnas.1919050117 173. Ford CL, Young LJ. Harnessing the healing power of love.
168. Oztan O, Garner JP, Partap S, et al. Cerebrospinal fluid vas- Trends Mol Med. 2021;27(9):833-834. doi:10.1016/j.
opressin and symptom severity in children with autism. Ann molmed.2021.07.010
Neurol. 2018;84(4):611-615. doi:10.1002/ana.25314 174. Johnson ZV, Young LJ. Evolutionary diversity as a catalyst
169. Parker KJ, Oztan O, Libove RA, et al. A randomized placebo- for biological discovery. Integr Zool. 2018;13(6):616-633.
controlled pilot trial shows that intranasal vasopressin doi:10.1111/1749-4877.12339

View publication stats

You might also like