ArticlePDF AvailableLiterature Review

Oxytocin receptor dynamics in the brain across development and species: OXT Receptor Dynamics in the Brain

Authors:

Abstract and Figures

Oxytocin (OXT) signaling through the OXT receptor plays a significant role in a variety of physiological processes throughout the lifespan. OXT's effects depend on the tissue distribution of the receptor. This tissue specificity is dynamic and changes across development, and also varies with sex, experience, and species. The purpose of this review is to highlight these themes with examples from several life stages and several species. Important knowledge gaps will also be emphasized. Understanding the effective sites of action for OXT via its receptor will help refine hypotheses about the roles of this important neuropeptide in the experience-dependent development and expression of species-typical social behavior. © 2016 Wiley Periodicals, Inc. Develop Neurobiol, 2016.
No caption available
… 
Content may be subject to copyright.
Oxytocin Receptor Dynamics in the Brain across
Development and Species
Radhika Vaidyanathan,
1,2
Elizabeth A.D. Hammock
1,2
1
Department of Psychology, Florida State University, Tallahassee, FL
2
Program in Neuroscience, Florida State University, Tallahassee, FL
Received 28 March 2016; revised 19 May 2016; accepted 27 May 2016
ABSTRACT: Oxytocin (OXT) signaling through
the OXT receptor plays a significant role in a variety of
physiological processes throughout the lifespan. OXT’s
effects depend on the tissue distribution of the receptor.
This tissue specificity is dynamic and changes across
development, and also varies with sex, experience, and
species. The purpose of this review is to highlight these
themes with examples from several life stages and several
species. Important knowledge gaps will also be empha-
sized. Understanding the effective sites of action for OXT
via its receptor will help refine hypotheses about the roles
of this important neuropeptide in the experience-
dependent development and expression of species-typical
social behavior. V
C2016 Wiley Periodicals, Inc. Develop Neurobiol
00: 000–000, 2016
Keywords: ontogeny; ornithine vasotocin; oxytocin; OT;
OXT; oxytocin receptor; OTR; OXTR; OT-R; OXT-R
INTRODUCTION
Oxytocin (OXT), a nine amino acid neuropeptide and
hormone, has a range of central and peripheral
effects, demonstrated by numerous studies in humans
and other animals. First appreciated for its well-
known role in uterine contractions and milk ejection,
OXT was hypothesized (Klopfer, 1971) and then
demonstrated (Pedersen and Prange, 1979; Pedersen
et al., 1982) to play a role in the transformation of the
na
ıve to maternal brain, priming the brain to attend to
and care for infants. In addition to maternal care,
OXT modulates sexual behavior, social recognition,
and affiliative behaviors (Caldwell et al., 1986; Insel
and Winslow, 1991; Williams et al., 1992; Ferguson
et al., 2001; Neumann and Landgraf, 2012). Within
the context of adult social behavior, OXT appears to
have an effect through modulation of the salience of
and/or reinforcement by social stimuli (Ferguson
et al., 2001; Young et al., 2001; Dolen et al., 2013;
Marlin et al,. 2015). In the past several decades, it
has become clear that the OXT system (the peptide
and its receptor, OXTR) plays a significant role in the
modulation of species-typical social behavior, which
is diverse: some species provide abundant parental
care, some are monogamous, some are highly aggres-
sive to conspecifics, and so on. How could this single
peptide system regulate such diverse behaviors across
species? The answer to this question is complicated,
but at its core, seems to be driven by species differen-
ces in OXTR location throughout the brain (Young,
1999). This perspective is heavily informed by social
behavior differences in adulthood either associated
with neuropeptide receptor distribution or caused by
direct manipulation of OXTR function (e.g., Bale
et al., 2001; Ferguson et al., 2001) or OXTR levels
(e.g. Lee et al., 2008a, 2008b; Ross et al., 2009).
OXTR is a seven trans-membrane domain G
protein-coupled receptor (GPCR) and its expression
undergoes dramatic upregulation and downregulation
Correspond ence to: E.A.D. Hammock (ehammock@fsu.edu)
Contract grant sponsor: The Florida State University.
Ó2016 Wiley Periodicals, Inc.
Published online 00 Month 2016 in Wiley Online Library
(wileyonlinelibrary.com).
DOI 10.1002/dneu.22403
1
in specific tissues. It is important to remember that
the functions of OXT/OXTR are pleiotropic and that
the Oxtr gene is not only expressed in the brain but
only in several other tissues such as uterus, kidney,
ovary, testis, thymus, heart, vascular endothelium,
osteoclasts, and myoblasts (Gimpl and Fahrenholz,
2001; Zingg and Laporte, 2003). For example, OXTR
is downregulated in uterine tissue after parturition as
demonstrated by a rapid decrease in the levels of
ligand-binding while it is upregulated in the mam-
mary glands during the entire period of lactation
(Soloff et al., 1979). As described above, there is a
great degree of diversity in the anatomical distribu-
tion of this receptor across species, which is espe-
cially well demonstrated within the brain (Insel et al.,
1991; Dubois-Dauphin et al., 1992; Insel and
Shapiro, 1992; Insel et al., 1993; Young et al., 1996;
Wang and Young, 1997; Rabhi et al., 1999; Olazabal
and Young, 2006; Beery et al., 2008; Campbell et al.,
2009; Freeman et al., 2014a, 2014b). Even among
closely related species there is dramatic variation in
the distribution of OXTR density which influences
species-typical social organization (Insel and Sha-
piro, 1992; Young et al., 1996; Young, 1999; Olaza-
bal and Young, 2006; Beery et al., 2008; Campbell
et al., 2009). For example, in voles, the binding
profile of the monogamous prairie (Microtus
ochrogaster) and pine (Microtus pinetorum) voles
show significant levels of OXTR in the nucleus
accumbens, but the closely related polygamous mon-
tane (Microtus montanus) and meadow (Microtus
pennsylvanicus) voles have much lower receptor den-
sity in this area (Insel and Shapiro, 1992; Ross et al.,
2009). Individual differences in OXTR density in the
nucleus accumbens within the prairie vole species are
associated with the probability of partner preference
behavior (a laboratory measure of monogamous mat-
ing systems) (King et al., in press) as well as monog-
amous behavior in prairie voles in the wild (Ophir
et al., 2012). Directly manipulating the numbers of
OXTR in the nucleus accumbens by viral vector gene
transfer enhances partner preference behavior in prai-
rie voles (Ross et al., 2009), although increasing the
levels of OXTR in this area is not sufficient to confer
partner preference behavior in the non-monogamous
meadow vole (Ross et al., 2009). These differences in
the neuroanatomical distribution of OXTR across spe-
cies, and even within related species, indicate a
strong association with social organization. Are these
differences present during development? Are the
adult differences a result of different developmental
trajectories? We can answer “yes” and “it depends”
to both of these questions. Species differences are
present during development, some differences are
transient, and species and experience-dependent dif-
ferences depend on the brain area in question. Identi-
fying the species-typical roles of OXT and OXTR in
development will be particularly valuable to further
our understanding of the neural basis of social
behavior.
Importantly, it is well appreciated that brains pre-
pare for adulthood through experience-dependent
developmental processes. Not only do species vary in
their developmental social experiences which then
shape adult species-typical behaviors and preferences
(Denenberg et al., 1964) but they also vary in devel-
opmental patterns of OXTR, to be reviewed below. If
we are to convince ourselves that developmental pat-
terns of OXTR are important for the emergence of
social behavior, then there must be a role for OXT-
OXTR signaling during development. Indeed, OXT is
a developmentally significant neuropeptide as several
studies have revealed the developmental effects of
OXT exposure on subsequent social behavior. The
developing brain is sensitive to early life experiences,
and the OXT system may be both a target and a medi-
ator of developmental experience (Bales and Perkey-
bile, 2012; Veenema, 2012; Hammock, 2015). In
other words, developmental experience shapes the
adult OXT system, and the infant OXT system medi-
ates some of the effects of the social environment.
There is ample evidence that the early environment
programs adult levels of OXT and OXTR contributing
to diversity in OXT function in adulthood. Addition-
ally, there is emerging evidence of sex, individual,
and species differences in the OXT system during
development, which likely contributes to altered
developmental trajectories, suggesting that OXT is a
mediator of the early environment. The early post-
natal environment exposes the infant to intense social
interactions, such as parent-infant and sibling interac-
tions. In humans as well as other mammalian species,
these early interactions expand to include a broad
range of extra-familial, cross-species, and other ani-
mate interactions. Species-typical developmentally
transient OXTR patterns might define social sensitive
periods for particular brain areas or stimuli.
ADULT OXTR ARE A TARGET OF
DEVELOPMENTAL EXPERIENCE
In the following paragraphs, we will discuss develop-
mental factors, including gonadal hormones and
maternal care, known to alter OXTR expression and
distribution into adulthood, supporting the hypothesis
that the OXT system is a target of developmental
2Vaidyanathan and Hammock
Developmental Neurobiology
experience [reviewed in (Bales and Perkeybile, 2012;
Veenema, 2012; Alves et al., 2015)].
Adult males and females show different levels of
OXTR in some brain regions. These differences could
be due to differences in exposure to gonadal hormones
during development (organizational effects) or changes
in hormone levels during adulthood (activational
effects). Estrogens regulate gene transcription of the
Oxtr gene in some brain areas. In adult rats, the transi-
tion to motherhood via pregnancy and parturition
enhances the expression of OXTR in several brain areas
post-partum which could be mimicked with estradiol
benzoate treatment after ovariectomy (Insel, 1986,
1990; Young et al., 1997; Young et al., 1998). Further-
more, there are sex differences in the expression of
OXTR that depend on gonadal hormones, in a brain-
region specific manner (de Kloet et al., 1986; Tribollet
et al., 1990; Bale and Dorsa, 1995a, 1995b; Bale et al.,
1995a, 1995b). These sex differences are maintained in
adulthood by gonadal steroids, but are also influenced
by neonatal exposure to gonadal steroids in a brain
region specific manner (Uhl-Bronner et al., 2005). As
an added layer of complexity, the direction of the effect
of sex steroid dependence of OXTR levels depends on
the species studied. For example, adult male rats have
higher levels of OXTR in the ventromedial hypothala-
mus than females, while in male mice, gonadal hor-
mones seem to suppress OXTR expression in this same
brain region (Insel et al., 1993). In meadow voles,
gonadal hormones and day length predict OXTR bind-
ing density, which is associated with seasonal social
affiliative behaviors (Beery and Zucker, 2010).
Maternal behavior and early-life experiences are
species-typical and are highly variable even within
individuals of the same species. Natural variation in
maternal care can confer enduring changes in OXTR
levels which are sex-specific (Francis et al., 2000;
Champagne et al., 2001; Francis et al., 2002). For
example, virgin female offspring born to high licking
and grooming mothers are quicker to exhibit maternal
behavior and also show significant differences in
OXTR distribution compared to the virgin female off-
spring of low licking and grooming mothers, and
these effects appear to depend on estrogens (Cham-
pagne et al., 2001). The offspring of high licking and
grooming and low licking and grooming dams also
show differences in Oxtr mRNA as well as estrogen
receptor alpha (ERa) during development, with a sig-
nificant effect of region and developmental age (Pena
et al., 2013). Manipulating the OXT system and
maternal care from infancy confers enduring changes
in the adult OXTR expression profile (Lukas et al.,
2010), but the mechanisms are unknown. Similarly,
in the biparental monogamous mandarin vole, pater-
nal deprivation during post-natal development
reduces adult OXTR expression in the nucleus accum-
bens and medial amygdala and disrupts social behav-
ior in a social recognition task in adulthood (Cao
et al., 2014).
Epigenetic mechanisms like methylation can
potentially contribute to the species and tissue diver-
sity of OXTR (Kusui et al., 2001; Kumsta et al.,
2013). In a recent study that aimed to test this
hypothesis in mice, methylation status of Oxtr was
associated with tissue specificity of expression in the
mammary glands and uterus (Mamrut et al., 2013) as
well as the brain (Harony-Nicolas et al., 2014) of
adult mice. While these studies were performed in
adult mice, it is possible that developmental experi-
ence leads to changes in methylation status of human
OXTR, to set up lifetime function of OXTR expres-
sion. This has been suggested in two recent studies in
humans which try to make a preliminary link
between child abuse (Smearman et al., 2016) or rat-
ings of parental care (Unternaehrer et al., 2015) and
OXTR methylation.
OXT/OXTR MAY BE A MEDIATOR
OF THE EARLY SOCIAL
AND SENSORY ENVIRONMENT
In the following paragraphs, we will provide evi-
dence that the OXT system is a potential mediator of
the early environment, as OXT activity during devel-
opment affects both short term and long term out-
comes. We describe some potent acute and long term
effects of OXT/OXTR manipulation in newborns.
Miller and Caldwell provide a very thorough tabu-
lated summary of research to date on neonatal manip-
ulations and the consequences of these manipulations
(Miller and Caldwell, 2015). Significantly, the pre-
cise neural circuit-based mechanisms of most of the
acute and long-term effects are not yet known.
Developmental OXT Has Acute Effects
There is growing evidence that OXT-OXTR has acute
effects on physiology or behavior during develop-
ment (Tyzio et al., 2006; Alberts, 2007; Pournajafi-
Nazarloo et al., 2007; Schaller et al., 2010; Mazzuca
et al., 2011; Meziane et al., 2015; Zheng et al., 2014)
[reviewed in (Kenkel et al., 2014; Hammock, 2015)].
During early postnatal development, rodents exhibit
huddling behavior, which primarily derives cues
from tactile and thermal peripheral sensations. At
postnatal day (P), 15 rat pups develop a filial hud-
dling behavior, guided by olfactory cues (Alberts,
OXT Receptor Dynamics in the Brain 3
Developmental Neurobiology
2007). Prior to this, rat pups are unable to differenti-
ate between a familiar conspecific and members of
other species (Alberts, 2007), which suggests that the
developmental learning of what it means to be a con-
specific occurs in the developmental period leading
up to P15. Maternal skin-to-skin contact is important
for acquiring these filial preferences and central
application of OXT increases contact between mother
and pups (Kojima and Alberts, 2011). In rats, daily
20-min maternal (and littermate) separation has been
shown to transiently reduce the amount of OXTR
ligand binding in dissected hippocampal membrane
homogenates during development on postnatal day 8,
yet transiently decrease at P4 and increase at P8 the
amount of ligand binding in the cingulate cortex
(Noonan et al., 1994). These same brain areas did not
show long term changes in OXTR ligand binding after
daily separation when measured at 1 year of age,
although it is unclear if the transient changes due to
altered maternal care lead to consequences for other
markers of developmental plasticity. Rat neonates
have peak expression in some brain areas in the first
week of life. The developmental curve was shifted in
a brain region dependent manner by maternal separa-
tion, suggesting that environmental inputs may shift
brain area sensitivity to OXT.
In the two populations of hypothalamic neurons
where OXT is produced, the supraoptic nucleus
(SON) and paraventricular nucleus (PVN), there is a
steady increase in OXT immunoreactive cell number
from postnatal day 1 to 21 (measured at P1, P8 and
P21), in male and female prairie voles (Yamamoto
et al., 2004). OXT immunoreactivity measured at
postnatal day 8, after a single neonatal OXT injec-
tion, shows no difference in males or females
between treatment group and control animals. How-
ever, at postnatal day 21, females that were given
neonatal OXT injections showed significantly
increased immunoreactivity in the PVN and not in
the SON compared to untreated females. Surpris-
ingly, females given an OXTR antagonist also showed
an increase in OXT immunoreactive cell number
(Yamamoto et al., 2004). Neonatal treatment with
OXT increases c-Fos immunoreactivity in the SON
in male pups, and treatment with OXT antagonist
increases c-Fos immunoreactivity in the SON of
female pups (Cushing et al., 2003). The important
distinction between OXT injections and OXT antago-
nist injections, is that the former is supplementary to
the endogenous OXT system (i.e., exogenous), and
the latter blocks the endogenous OXT system during
the neonatal period. Keeping in mind this distinction,
it can be hypothesized that male prairie vole pups are
more responsive to exogenous OXT, while female
prairie vole pups may have a more robust endogenous
OXT system, which makes them resilient to OXT
manipulations.
During development, there are several temporal
windows when different sensory systems and their
neural circuits become receptive to environmental
stimuli. Due to this, the acute effects of OXT may
depend on the specific OXTR-containing neural cir-
cuits that are undergoing maturation and develop-
ment at a given time. In immature neurons, GABA is
an excitatory neurotransmitter, and shortly before
parturition (E21) in rat neonates, there is a transient
switch from being excitatory to inhibitory (Tyzio
et al., 2006). This transient switch from depolarizing
to hyperpolarizing GABA
A
receptor signaling is trig-
gered by an OXT-mediated decrease in intracellular
[Cl
2
] (Tyzio et al., 2006). This transient switch has
been shown to exert neuroprotective and analgesic
effects on newborns (Tyzio et al., 2006; Khazipov
et al., 2008; Mazzuca et al., 2011; Tyzio et al., 2014).
Blocking OXT signaling in na
ıve mothers produces
offspring that display behavioral and electrophysio-
logical autism-like traits (Tyzio et al., 2014).
Prader–Willi syndrome (PWS) is a neurodevelop-
mental disorder with a complex phenotype that
includes feeding and suckling problems from birth
(Holm et al., 1993). OXT injections 3–5 h after birth
rescues the feeding and suckling deficits in mice
lacking the Magel-2 gene, which is altered in PWS
(Schaller et al., 2010). In particular, the onset of
suckling behavior, which involves coordination of
sensory and motor processes to orient to olfactory
and tactile cues, is disturbed in these mice (Schaller
et al., 2010) and OXT may likely be important in
modulating these multisensory processes [reviewed
in (Hammock, 2015)], with long term consequences
for social behavior (Meziane et al., 2015). OXT mod-
ulation of oromotor behavior may be present in rats
as well. A very early study demonstrated that acute
administration of OXT into rat pup cisterna increases
autogrooming and oromotor behaviors (Pedersen
et al., 1988), including paw-sucking in rats (Nelson
and Alberts, 1997). These oromotor behaviors may
be functionally related to the OXT-facilitated suck-
ling processes as in the mouse.
Kramer et al. (2003) tested the effects of giving a
single dose of OXT or OXT-antagonist (OXTA) ver-
sus daily doses, on stress-related behavior in socially
isolated infant prairie voles. A single dose of OXTA
within the first 24 h after birth, decreased separation-
induced vocalizations in females but not males a
week later, suggesting a sex-specific effect of OXT.
In contrast, repeated daily treatment with the OXTA
had the opposite effect on separation-induced
4Vaidyanathan and Hammock
Developmental Neurobiology
vocalizations in females (Kramer et al., 2003). The
mechanisms of the sex-dependent effects are
unknown.
Developmental OXT Has Long
Term Effects
There is a significant amount of evidence that OXT-
OXTR during development has long-term effects on
adult physiology and/or behavior (Boer and Kruis-
brink, 1984; Boer, 1993; Boer et al., 1994; Bales and
Carter, 2003a, 2003b; Kramer et al., 2003; Bales
et al., 2004a, 2004b, 2007a, 2007b, 2013; Yamamoto
et al., 2004, 2006; Kramer et al., 2006; Jia et al.,
2008a, 2008b; Perry et al., 2009; Bales and Perkey-
bile, 2012; Eaton et al., 2012; Hashemi et al., 2013;
Meziane et al., 2015; Mogi et al., 2014; Tyzio et al.,
2014) reviewed in (Carter, 2003, 2007; Carter et al.,
2009; Young, 2013; Grinevich et al., 2014). Many of
these studies explore single doses of OXT given to
vole neonates and later determining the effects on
social behavior outcomes.
Neonatal OXT can have organizational effects on
the neural substrates underlying these changes in
adult physiology and behavior. Early postnatal OXT
injections have differential and sexually dimorphic
effects on adult animals (Kramer et al., 2003; Yama-
moto et al., 2004, 2006; Kramer et al., 2006; Mogi
et al., 2014). For example, a single neonatal dose of
OXT results in an increased partner preference in
male prairie voles in adulthood and decreased
anxiety-like behaviors (Carter et al., 2009). Female
prarie voles require a higher dose of postnatal OXT
injections, for an increased partner preference in
adulthood (Carter et al., 2009). A single dose of OXT
in newborn rats decreases the levels of dopamine and
serotonin metabolites in regions such as the striatum
and hypothalamus, in adult rats (Hashemi et al.,
2013), suggesting the potential indirect effect that
OXT can have on other neurotransmitter systems.
Several behaviors exhibited by the model systems
used in research are sexually dimorphic. Reproduc-
tive behavior and aggressive behavior are two exam-
ples that have been tested in the laboratory setting.
What is the neurological basis for this difference
and when do these divergent trajectories emerge?
Gonadal hormone exposure during early life has an
organizational effect on the brain and can induce per-
manent changes in the central nervous system. OXT
injections in male prairie voles during the neonatal
period increases pair-bonding in adulthood, and
males respond to a lower dose of OXT compared to
females, suggesting that male voles are particularly
sensitive to neonatal manipulations of OXT system.
OXT antagonists produce the opposite effect of
reduced pair-bonding in males (Bales and Carter,
2003a, 2003b). Intra-sexual aggression in adult prai-
rie voles is displayed by a significantly greater num-
ber of males than females. Adult female prairie voles
exhibit increased aggressive behavior toward strang-
ers after prolonged exposure to a male partner. OXT
injections during development increase adult intra-
sexual aggressive behavior in female prairie voles
following a few hours of exposure to a male partner
(Bales and Carter, 2003a, 2003b). The mechanisms
underlying the role of OXT in intra-sexual aggressive
behavior in females is unknown.
In adult females, many of the reproductive effects
of OXT are estrogen dependent, where estradiol ben-
zoate injections increase the female responses to
OXT (Caldwell et al., 1986). Neonatal manipulations
of OXT have been shown to alter ERaimmunoreac-
tivity, ERamRNA expression, as well as estrogen-
dependent responses during adulthood in female
prairie voles (Yamamoto et al., 2006; Pournajafi-
Nazarloo et al., 2007). In male prairie voles, develop-
mental manipulations of OXT seem to influence the
expression of sexual behavior and sperm transport in
adulthood (Bales et al., 2004a,b). There are sex-
differences in the brain after neonatal manipulations;
endogenous OXT during development has a sexually
dimorphic neural activation in males and females,
and exogenous OXT has been shown to affect neural
activity (measured by c-Fos immunoreactivity), spe-
cifically in males (Cushing et al., 2003; Yamamoto
et al., 2004).
Adult male mice deficient in Magel-2 gene, a can-
didate gene for PWS, show deficits in learning and
social behavior. These deficits are rescued by a single
dose of OXT in the adults. The deficits were also
shown to be rescued in adulthood by multiple doses
of OXT injections on six consecutive days from the
day of birth (Meziane et al., 2015). To better under-
stand the acute and long-term effects of OXT/OXTR
in development on social behavior outcomes, it is
important to know where the receptors are located
during development.
WHERE ARE THE EFFECTOR SITES
OF OXT DURING DEVELOPMENT?
The examples above provide strong evidence for an
organizational role for OXT/OXTR signaling during
development. Understanding the developmental, spe-
cies, and sex-specific circuit expression of OXTR at
the cellular level will be crucial to identifying the
mechanistic role of the OXT system during
OXT Receptor Dynamics in the Brain 5
Developmental Neurobiology
experience-dependent development. Where are these
effector sites? Anatomically, in both the adult and in
development, there is not substantial overlap between
all of the regions that express OXTR and the cells that
express OXT. The working model based on the avail-
able evidence indicate that OXT can act as a local
signal (i.e., synaptic transmission) and as a neurohor-
mone to modulate circuits at a distance from the
source of OXT (i.e., volume transmission) (Landgraf
and Neumann, 2004). This latter neuroendocrine
action of OXT can negate the need for OXT innerva-
tion into OXTR-expressing circuits, and thus
increases the dynamic developmental flexibility of
the system. The ontogeny of OXTR in the brain has
been explored prenatally and postnatally in rats,
mice, voles, and humans. Studies have approached
this question by predominantly two methods; expres-
sion profile of the mRNA transcript for OXTR, and
ligand binding studies to determine receptor distribu-
tion in the brain. Reporter mice have also been used,
especially to identify adult expression patterns. Until
recently, a truly convincing antibody (Mitre et al.,
2016) has not been available for use in mouse tissue.
The available data across developmental (prenatal
and postnatal) time points are presented in Figure 1.
At both the mRNA and protein level, the expres-
sion and distribution of OXTRs are significantly dif-
ferent during development compared to adulthood.
Using ligand binding receptor autoradiography, three
types of OXTR binding profiles have been identified:
(1) continuous binding from the time of first detection
through adulthood, (2) binding that has a pubertal
onset, and (3) transient binding in some brain areas
that only occurs during certain periods of
Figure 1 Coded summary of developmental maps of OXTR in various rodent species. Only studies
that include preweaning developmental time points are included here. M 5male; F 5female;
E5embryonic; P 5postnatal; ND 5no data; Signal intensity estimates5111 (peak) >
11 >1>1/2>2(none). Schematized patterns represent three types of brain-region specific
developmental profiles evident from developmental mapping studies across species. Some brain
regions follow the infant pattern, with a peak expression during pre-weaning development. Other
brain regions follow the pubertal pattern, evident at puberty. Finally, some brain regions show the
adult pattern, with a gradual onset throughout development which persists in adulthood. The spe-
cific brain regions which follow these patterns vary across species.
6Vaidyanathan and Hammock
Developmental Neurobiology
development. Transient OXTR expression in different
brain regions during development suggests that dif-
ferent regions are made sensitive to OXT during dif-
ferent stages of development. The functional role of
this transient expression is still largely unknown.
Hormonal imprinting (Csaba, 1986; Hashemi et al.,
2013) and experience-dependent sensitive period
plasticity (Hammock and Levitt, 2013; Hammock,
2015) are two non-mutually exclusive hypotheses
about the role of these transient patterns. Here, we
highlight developmental differences in OXTR, and
the similarities and differences in rodent species dur-
ing the early postnatal period will help to dissect the
roles of OXT in the development of species-typical
behaviors.
In rats (Chen et al., 2000) and mice (Tamborski
et al., 2016), detection of Oxtr mRNA expression is
possible as early as E12 by qPCR, and at E13 by in
situ hybridization in rats (Yoshimura et al., 1996). In
the developmental in situ hybridization study con-
ducted by Yoshimura et al. (1996) in rats, Oxtr
mRNA was detected by E13 in the dorsal motor
nucleus of the vagus nerve, with a progressive
increase in expression in other regions over time. The
authors of this study noted two distinct expression
patterns: transient and constant type (Yoshimura
et al., 1996). Both kinds of patterns were evident in
the brainstem, mesencephalon, diencephalon, and tel-
encephalon. Transient Oxtr mRNA during the early
postnatal period was detected in the caudate, puta-
men, cingulate cortex, anterior thalamic nuclei, ven-
tral tegmental area, and the hypoglossal nucleus,
while constant expression, once detected, persisted
into adulthood in the anterior olfactory nucleus, tenia
tecta, some amygdaloid nuclei, piriform cortex, the
ventromedial hypothalamic nucleus, subiculum, and
the dorsal motor nucleus of the vagus (Yoshimura
et al., 1996).
As alluded to above, OXTR detection by receptor
autoradiography has been used with great success
because it is easy to use and quantitative. Two types
of ligands have been used to map OXTR: tritiated-
OXT (
3
H-OXT) and iodinated- ornithine vasotocin
(
125
I-OVTA), a specific OXTR ligand. Because of the
sequence similarity between OXT and its paralog,
arginine-vasopressin (AVP), their cognate receptors
must be discriminated using labeled ligands that are
very specific or by competing off ligand binding with
highly selective agonists/antagonists. One of the ear-
liest developmental mapping studies to detect OXTR
by receptor autoradiography used
3
H-OXT in rats
(Snijdewint et al., 1989). In that report, numerous
binding sites for OXT were identified, including
developmentally transient expression in the dorsal
hippocampus and parietal cortex measured at post-
natal day 5, with adult levels of expression observed
on post-natal day 15. Because
3
H-OXT can bind to
other members of the OXT/AVP family of GPCRs,
some of the early results were not actually OXTR, but
included other receptor types. This is important to
keep in mind, because OXT and OXTR KO mice do
not have identical phenotypes, which indicates that
the OXT peptide can act at other receptors and the
OXTR may also signal effectively after AVP binding.
To get around the problem of specificity, several
groups have developed selective ligands for the
OXT/AVP family of GPCRs. The highly selective
ornithine vasotocin ligand (Elands et al., 1988) is the
most commonly used agent, which is commercially
available from NEN/Perkin Elmer (ornithine vasoto-
cin analog (vasotocin, d(CH2)5[Tyr(Me)
2
,Thr
4
,Or-
n
8
,[
125
I]Tyr
9
NH2]; ([
125
I]-OVTA, NEX254, Perkin-
Elmer, Boston, MA), and used in numerous studies
(Shapiro and Insel, 1989; Snijdewint et al., 1989; Tri-
bollet et al., 1989; Wang and Young, 1997; Chen
et al., 2000; Hammock and Levitt, 2013). The first
report of a developmental map of OXTR using this
ligand was in 1989. Tribollet et al. (Tribollet et al.,
1989) mapped OXTR ligand binding in rats and
clearly demonstrated that there were two phases of
expression: an “infant” pattern and an “adult” pattern.
Whereas Snijdewint et al. (1989) measured
3
H-OXT
ligand binding at E20, P1, P5, and P15, Tribollet
et al. sampled tissue at E12, E14, E16, E18, E20, and
P1, P3, P5, P10, P13, P16, P19, P22, P25, P30, P35,
P40, P45, P60, and P90. While the Snijdewint study
concluded that P5 expression was higher than P15,
with higher temporal resolution sampling, Tribollet
found a peak of expression in the cingulate cortex
closer to P10 in the rat. The onset of enhanced OXTR
in some brain areas in puberty is likely also due to
gonadal hormones at puberty (Tribollet et al., 1992).
The brain region specificity of OXTR expression
not only varies across development but also between
species. The earliest detectable OXTR ligand binding
signal is at E14 in rats (Tribollet et al., 1989) and
E16.5 in mice (Tamborski et al., 2016). While the
gestational periods of rats and mice are similar, there
is approximately a two-day difference in neural
development between rats and mice (Clancy et al.,
2001). Even closely related species vary in their
OXTR distribution during infant development. For
example, Wang and Young (1997) showed that dur-
ing postnatal development there is an accelerated
increase in OXTR ligand binding in the lateral septum
of the polygamous montane vole compared to
monogamous prairie vole, similar to the adults of
these species that showed a differential binding in the
OXT Receptor Dynamics in the Brain 7
Developmental Neurobiology
lateral septum (Insel and Shapiro, 1992). These find-
ings contribute to the hypothesis that species differen-
ces in adult social behavior have early developmental
origins.
The brain regions that show a transient develop-
mental OXTR expression are variable between spe-
cies. In rats (Sprague-Dawley), a transient binding
peak has been described at postnatal day 10 [with
adjacent sampling at P5 or P13 (Tribollet et al.,
1989) or P5 and P14 (Shapiro and Insel, 1989)], in
several regions such as the cingulate cortex, dorsal
subiculum, anterior and paraventricular thalamic
nuclei, nucleus accumbens, caudate putamen, mam-
millary complex, substantia gelatinosa of the spinal
trigeminal nucleus, and the reticular nucleus, lateral
septum, fornix, globus pallidus, and anterior CA1
region of the hippocampus (Shapiro and Insel, 1989;
Tribollet et al., 1989). Transient OXTR immunoreac-
tivity was observed in several sensory nuclei of the
brainstem of the rat neonate (Murata et al., 2011),
which at the electron microscope level, appeared to
be enriched in the Golgi apparatus. These regions in
rats can be contrasted with the transient expression in
C57BL/J6 mice when sampled at P0, P7, P14, P21,
P35, and P60: a transient developmental peak has
been identified throughout the neocortex at P14,
quantified in S1 neocortex (Hammock and Levitt,
2013). This transient post-natal expression by recep-
tor autoradiography in mice is also evident at the
level of mRNA (Fertuzinhos et al., 2014; Mitre et al.,
2016), as well as by immunohistochemistry with a
specific antibody to OXTR (Mitre et al., 2016).
Although the receptor autoradiography technique
is rapid and quantifiable, it offers less cell-specific
resolution. Reporter mice can also be used to deter-
mine the developmental origins of OXTR and these
mice have been made to assist in mapping the cell-
type specific expression of the mouse OXTR,asan
additional approach to complement in situ hybridiza-
tion studies, and out of necessity due to the historic
lack of suitable antibodies for OXTR in mouse. As an
example, the Oxtr-yfp mouse (Yoshida et al., 2009),
has a Venus yellow fluorescent protein coding region
in place of the OXTR coding region. Mice that have
two copies of the Venus-YFP are also knock-outs for
OXTR. These mice have not yet been used for a
developmental expression study.
The Oxtr-lacZ mouse contains the Oxtr promoter
driving the expression of beta galactosidase from the
lacZ gene (Gould and Zingg, 2003). While the coding
region for OXTR was still present in these mice, they
did show a knock-down of Oxtr expression in the
uterus (75% of expression in hemizygous mice and
50% expression in homozygous lacZ mice). These
mice have not been used in developmental studies,
but seem like a valuable tool to explore the effects of
partial loss of OXTR levels in OXTR circuitry.
The GENSAT consortium has made an Oxtr-EGFP
mouse (Gong et al., 2003). Similar to the two lines of
mice just described, this mouse has an Oxtr promoter
driving EGFP expression. An important difference,
however, is that this mouse is a BAC transgenic, and
thus the construct containing the Oxtr-EGFP ran-
domly inserted into the founder mouse genome. The
native OXTR locus in the mouse is presumably unaf-
fected. These mice have been used in developmental
characterization of expression of the transgene,
including embryonic and early post-natal ages. As
with ligand binding, mRNA, and protein studies, the
EGFP expression is striking in earlier ages, especially
for the neocortex.
A new tool has recently been made to be able to
selectively manipulate detectable OXTR circuits
(Hidema et al., 2016). This transgene has an HA
epitope-tagged OXTR that also includes an IRES Cre
construct. With these mice, OXTR cells can be identi-
fied easily with immunohistochemistry for the HA
tag and those same cells also express Cre recombi-
nase. These mice could be bred with a ROSA reporter
strain to detect expression, or combined with viral
vector technologies to perform tract tracing or opto-
genetics experiments, or in combination with other
mice with floxed transgenes to achieve selective dele-
tion of genes in OXTR circuits. This should be a valu-
able tool in the developmental dissection of the OXT-
dependent developmental emergence of social behav-
ior in mice.
We have tightly focused this review on the develop-
mental dynamics of brain OXTR, but it is important to
point out that the OXT/OXTR system has functions
beyond the brain and social behavior. For example,
emerging data indicate a role for OXT/OXTR in the
development of the gastrointestinal system and its reg-
ulation in adulthood (Welch et al., 2009; Welch et al.,
2014), metabolism (Chaves et al., 2013), osteogenesis
(Colaianni et al., 2015), and thermoregulation (Kasa-
hara et al., 2015). These pleiotropic OXT activities
may have downstream consequences that interact with
the brain and, as in the brain, may vary across devel-
opment, species, sex, and individual.
WHAT IS THE DEVELOPMENTAL OXTR
DISTRIBUTION PROFILE IN HUMANS?
OXT affects human social behavior [reviewed in
(Meyer-Lindenberg, 2008; Heinrichs et al., 2009;
Guastella and MacLeod, 2012; Zink and Meyer-
8Vaidyanathan and Hammock
Developmental Neurobiology
Lindenberg, 2012; Feldman, 2015)], although our
toolset to look at human-specific OXTR expression
and in vivo function is understandably limited. The
historic lack of a comprehensive set of tools to iden-
tify OXTRs makes it a challenge to map these recep-
tors in the human brain. Loup et al. (1989, 1991)
used
3
H-OXT and
125
I-OVTA to map OXT binding
sites in adult brain tissue. While,
125
I-OTA has high
specificity to OXTR in rats (Shapiro and Insel, 1989),
and subsequently in other rodents (Wang and Young,
1997; Takayanagi et al., 2005; Ross et al., 2009;
Hammock and Levitt, 2013), it appears not to be
selective for OXTR in the primate brain (Toloczko
et al., 1997; Freeman and Young, 2016). A com-
monly held interpretation of these results is that the
ligand specificity is lost between rodents and prima-
tes and thus the absence of specific binding in pri-
mate tissue with this ligand does not demonstrate that
there are not OXTR in the primate brain. More recent
studies demonstrate both OXTR mRNA and protein
in the human brain (Kang et al., 2011; Boccia et al.,
2013; Freeman et al., 2016).
The available information about OXTR expression
or binding in the developing human brain is sparse.
While ligand binding and immunohistochemical
studies have yet to be performed in human develop-
ment, data from the Human Brain Atlas indicate a
heightened transient profile of OXTR mRNA in the
human neocortex during the first postnatal years
(Kang et al., 2011). New ligands with better specific-
ity for the human OXTR are being developed (Man-
ning et al., 2008; Smith et al., 2012, 2013a, 2013b;
Freeman et al., 2016), and an antibody to primate
OXTR looks promising (Boccia et al., 2013), so it
may be possible to observe the developmental loca-
tions of OXTR in the human brain in the near future.
Based on recent studies in OXTR in mouse neocortex
(Marlin et al., 2015; Mitre et al., 2016), special atten-
tion to lateralization of OXTR expression is war-
ranted. A clear picture of the developmental
distribution in humans is important to evaluate the
clinical application of OXT in human disorders like
autism and schizophrenia [argued in (Insel, 2016)].
IMPLICATIONS OF DEVELOPMENTALLY
TRANSIENT OXTR
What is the potential significance of a neocortical
OXTR signal (e.g., in humans in the early postnatal
years, in the cingulate cortex in rats, and widespread
throughout upper layers II/III in the neocortex of
mice) in the context of development? During the first
two postnatal weeks in altricial rodents, early experi-
ences are mediated by chemosensory and thermotac-
tile processes (Brunjes and Alberts, 1979; Kojima
and Alberts, 2011), and unless a pup is separated
from the litter, the entire neonatal period is contin-
gent with social chemosensory and thermotactile
inputs. By the end of the second postnatal week, as
the eyes and ears open, input-dependent signals for
visual and auditory information emerge in the neo-
cortex, joining the activity-dependent input in soma-
tosensory cortex (Colonnese et al., 2010), and this
period coincides with peak levels of white matter
stimulation-induced LTP in layers II/III (Rozas et al.,
2001). We have hypothesized that OXT, via develop-
mentally transient OXTR, may be important in shap-
ing the experience-dependent plasticity of the
neocortex during the developmental onset of multi-
sensory processes, because of their location in layers
II/III (Hammock and Levitt, 2013; Hammock, 2015),
known for its heavy cortico-cortical projections.
Direct experimental evidence supports this conjecture
(Zheng et al., 2014). In an effort to understand mech-
anisms of developmental multisensory plasticity in
layers II/III of the neocortex, at postnatal day 14,
Zheng et al. compared typically developing mice to
mice that had one of two modes of sensory depriva-
tion since birth—either whisker trimming or constant
dark rearing. In the neocortex, animals that had been
whisker trimmed not only showed reduced frequency
of miniature excitatory post-synaptic currents
(mEPSCs) in layer II/III pyramidal neurons of S1
neocortex in the area representing the whiskers but
also in primary visual cortex and primary auditory
cortex. This suggests that the reduction of input from
a single modality (the whiskers) affected the develop-
ment of the neocortical processing of the other
modalities. The same phenomenon was observed
after constant dark-rearing: in addition to reduced
mEPSC frequency in the primary visual cortex, there
was also a reduction evident in primary somatosen-
sory cortex and primary auditory cortex. To identify
potential mechanisms that might mediate the effects
of sensory deprivation on this cross-modal neocorti-
cal plasticity, Zheng et al. performed separate tran-
scriptomic analyses of both the neocortex and the
thalamus/hypothalamus. The most robust transcript
to emerge from this analysis after either form of
deprivation was Oxt mRNA in the hypothalamus.
Either form of sensory deprivation led to reduced Oxt
mRNA and protein at P14. Zheng et al. went on to
characterize the effects of direct manipulation of
OXT and observed that increasing OXT levels
increased mEPSC frequency in all three primary sen-
sory areas and that decreasing OXT decreased
mEPSC frequency in all three primary sensory areas.
OXT Receptor Dynamics in the Brain 9
Developmental Neurobiology
Thus, manipulating OXT levels was sufficient to
recapitulate the effects of 2 weeks of sensory depriva-
tion on multi-modal neocortical activity. Transient
OXTR binding patterns may reflect a developmental
sensitive period for functional activation by OXT in
those regions through volume transmission. Addi-
tionally, transient binding profiles may also reflect
developmental changes such as neurite growth, den-
dritic branching, synaptic pruning, and synaptogene-
sis. OXT, perhaps increased by social contexts, may
make contingent multisensory inputs more likely to
induce neural activity during activity-dependent mul-
tisensory neocortical development, right at a time
when these inputs are first available to the neocortex.
This might permit socially contingent sensory inputs
to increase and/or refine the neocortical real estate
devoted to detecting and processing social inputs
(Hammock, 2015).
CONCLUSIONS AND FUTURE
DIRECTIONS
In this review, we highlighted data on diverse pat-
terns of brain expression of OXTR due to experience,
gonadal sex, and species. These differences are brain
region specific. For each of these dimensions, there is
evidence that receptor quantity influences some
aspect of social behavior. We also highlighted emerg-
ing research on species differences in developmental
patterns of expression. This leads to the hypothesis
that developmental differences between (and individ-
ual differences within) species can contribute to
species-typical developmental trajectories (or indi-
vidual and sex differences in trajectories). Transient
peaks of OXTR expression may define sensitive peri-
ods for OXT to shape the brain development of those
areas, and perhaps in a social-environment dependent
manner. In other words, OXTR may be a developmen-
tal plasticity gene that serves as a transducer of the
social environment to fine-tune the experience-
dependent plasticity of the social brain (Hammock
and Levitt, 2006; Hammock and Levitt, 2013;
Hammock, 2015).
This is a fairly broad hypothesis which needs to be
broken down into smaller testable questions.
a. How does the social or sensory environment
begin to elicit OXT activity in socially na
ıve
neonates?
b. How does OXT modulate sensory activity in
neonates?
a. Which sensory modalities? Is it species
dependent?
b. At what level(s)? Sensation? Perception?
Motivation? Cognition?
c. What cells and circuits are especially affected
by OXT during development? How are circuit
dynamics affected?
d. Is there a true sensitive period for OXT effects?
Does this have implications for human clinical
efforts attempting to use OXT to improve social
behavior?
e. How does region specific OXT/OXTR activity
in development affect emergence of species
typical behavior?
f. How do species and individual level differences
in OXT system genetics lead to differences in
function- both during development and in
adulthood?
g. How do sex differences in the OXT system lead
to differences in function during development
and in adulthood?
h. What is the experience-dependent developmen-
tal trajectory of this system in humans?
A focus on OXT-dependent neural circuit activity
during development and the mechanisms of sensitive
period plasticity will greatly enhance our understanding
of the role of OXT in social behavior development.
The authors declare no conflict of interest in preparing this
work.
REFERENCES
Alberts JR. 2007. Huddling by rat pups: Ontogeny of indi-
vidual and group behavior. Dev Psychobiol 49:22–32.
Alves E, Fielder A, Ghabriel N, Sawyer M, Buisman-
Pijlman FT. 2015. Early social environment affects the
endogenous oxytocin system: A review and future direc-
tions. Front Endocrinol (Lausanne) 6:32.
Bale TL, Davis AM, Auger AP, Dorsa DM, McCarthy
MM. 2001. CNS region-specific oxytocin receptor
expression: Importance in regulation of anxiety and sex
behavior. J Neurosci 21:2546–2552.
Bale TL, Dorsa DM. 1995a. Regulation of oxytocin receptor
messenger ribonucleic acid in the ventromedial hypothala-
mus by testosterone and its metabolites. Endocrinology
136:5135–5138.
Bale TL, Dorsa DM. 1995b. Sex differences in and effects
of estrogen on oxytocin receptor messenger ribonucleic
acid expression in the ventromedial hypothalamus. Endo-
crinology 136:27–32.
Bale TL, Dorsa DM, Johnston CA. 1995a. Oxytocin recep-
tor mRNA expression in the ventromedial hypothalamus
during the estrous cycle. J Neurosci 15:5058–5064.
10 Vaidyanathan and Hammock
Developmental Neurobiology
Bale TL, Pedersen CA, Dorsa DM. 1995b. CNS oxytocin
receptor mRNA expression and regulation by gonadal
steroids. Adv Exp Med Biol 395:269–280.
Bales KL, Abdelnabi M, Cushing BS, Ottinger MA, Carter
CS. 2004a. Effects of neonatal oxytocin manipulations on
male reproductive potential in prairie voles. Physiol
Behav 81:519–526.
Bales KL, Carter CS. 2003a. Developmental exposure to
oxytocin facilitates partner preferences in male prairie
voles (Microtus ochrogaster). Behav Neurosci 117:854–
859.
Bales KL, Carter CS. 2003b. Sex differences and develop-
mental effects of oxytocin on aggression and social
behavior in prairie voles (Microtus ochrogaster). Horm
Behav 44:178–184.
Bales KL, Perkeybile AM. 2012. Developmental experien-
ces and the oxytocin receptor system. Horm Behav 61:
313–319.
Bales KL, Perkeybile AM, Conley OG, Lee MH, Guoynes
CD, Downing GM, Yun CR, et al. 2013. Chronic intra-
nasal oxytocin causes long-term impairments in partner
preference formation in male prairie voles. Biol Psychia-
try 74:180–188.
Bales KL, Pfeifer LA, Carter CS. 2004b. Sex differences
and developmental effects of manipulations of oxytocin
on alloparenting and anxiety in prairie voles. Dev Psy-
chobiol 44:123–131.
Bales KL, Plotsky PM, Young LJ, Lim MM, Grotte N,
Ferrer E, Carter CS. 2007a. Neonatal oxytocin manipula-
tions have long-lasting, sexually dimorphic effects on
vasopressin receptors. Neuroscience 144:38–45.
Bales KL, van Westerhuyzen JA, Lewis-Reese AD, Grotte
ND, Lanter JA, Carter CS. 2007b. Oxytocin has dose-
dependent developmental effects on pair-bonding and
alloparental care in female prairie voles. Horm Behav 52:
274–279.
Beery AK, Lacey EA, Francis DD. 2008. Oxytocin and
vasopressin receptor distributions in a solitary and a
social species of tuco-tuco (Ctenomys haigi and Cten-
omys sociabilis). J Comp Neurol 507:1847–1859.
Beery AK, Zucker I. 2010. Oxytocin and same-sex social
behavior in female meadow voles. Neuroscience 169:
665–673.
Boccia ML, Petrusz P, Suzuki K, Marson L, Pedersen CA.
2013. Immunohistochemical localization of oxytocin
receptors in human brain. Neuroscience 253C:155–164.
Boer GJ. 1993. Chronic oxytocin treatment during late ges-
tation and lactation impairs development of rat offspring.
Neurotoxicol Teratol 15:383–389.
Boer GJ, Kruisbrink J. 1984. Effects of continuous adminis-
tration of oxytocin by an accurel device on parturition in
the rat and on development and diuresis in the offspring.
J Endocrinol 101:121–129.
Boer GJ, Quak J, de Vries MC, Heinsbroek RP. 1994. Mild
sustained effects of neonatal vasopressin and oxytocin
treatment on brain growth and behavior of the rat. Pep-
tides 15:229–236.
Brunjes PC, Alberts JR. 1979. Olfactory stimulation indu-
ces filial preferences for huddling in rat pups. J Comp
Physiol Psychol 93:548–555.
Caldwell JD, Prange AJ Jr, Pedersen CA. 1986. Oxytocin
facilitates the sexual receptivity of estrogen-treated
female rats. Neuropeptides 7:175–189.
Campbell P, Ophir AG, Phelps SM. 2009. Central vaso-
pressin and oxytocin receptor distributions in two species
of singing mice. J Comp Neurol 516:321–333.
Cao Y, Wu R, Tai F, Zhang X, Yu P, An X, Qiao X, et al.
2014. Neonatal paternal deprivation impairs social recog-
nition and alters levels of oxytocin and estrogen receptor
alpha mRNA expression in the MeA and NAcc, and
serum oxytocin in mandarin voles. Horm Behav 65:57–
65.
Carter CS. 2003. Developmental consequences of oxytocin.
Physiol Behav 79:383–397.
Carter CS. 2007. Sex differences in oxytocin and vasopres-
sin: Implications for autism spectrum disorders? Behav
Brain Res 176:170–186.
Carter CS, Boone EM, Pournajafi-Nazarloo H, Bales KL.
2009. Consequences of early experiences and exposure to
oxytocin and vasopressin are sexually dimorphic. Dev
Neurosci 31:332–341.
Champagne F, Diorio J, Sharma S, Meaney MJ. 2001. Nat-
urally occurring variations in maternal behavior in the rat
are associated with differences in estrogen-inducible cen-
tral oxytocin receptors. Proc Natl Acad Sci USA 98:
12736–12741.
Chaves VE, Tilelli CQ, Brito NA, Brito MN. 2013. Role of
oxytocin in energy metabolism. Peptides 45:9–14.
Chen Q, Schreiber SS, Brinton RD. 2000. Vasopressin and
oxytocin receptor mRNA expression during rat telen-
cephalon development. Neuropeptides 34:173–180.
Clancy B, Darlington RB, Finlay BL. 2001. Translating
developmental time across mammalian species. Neuro-
science 105:7–17.
Colaianni G, Sun L, Zaidi M, Zallone A. 2015. The “love
hormone” oxytocin regulates the loss and gain of the fat-
bone relationship. Front Endocrinol (Lausanne) 6:79.
Colonnese MT, Kaminska A, Minlebaev M, Milh M,
Bloem B, Lescure S, Moriette G, et al. 2010. A conserved
switch in sensory processing prepares developing neocor-
tex for vision. Neuron 67:480–498.
Csaba G. 1986. Receptor ontogeny and hormonal imprint-
ing. Experientia 42:750–759.
Cushing BS, Yamamoto Y, Hoffman GE, Carter CS. 2003.
Central expression of c-Fos in neonatal male and female
prairie voles in response to treatment with oxytocin.
Brain Res Dev Brain Res 143:129–136.
de Kloet ER, Voorhuis DA, Boschma Y, Elands J. 1986.
Estradiol modulates density of putative ’oxytocin recep-
tors’ in discrete rat brain regions. Neuroendocrinology
44:415–421.
Denenberg VH, Hudgens GA, Zarrow MX. 1964. Mice
reared with rats: Modification of behavior by early expe-
rience with another species. Science 143:380–381.
OXT Receptor Dynamics in the Brain 11
Developmental Neurobiology
Dolen G, Darvishzadeh A, Huang KW, Malenka RC. 2013.
Social reward requires coordinated activity of nucleus
accumbens oxytocin and serotonin. Nature 501:179–184.
Dubois-Dauphin M, Pevet P, Barberis C, Tribollet E,
Dreifuss JJ. 1992. Localization of binding sites for oxyto-
cin in the brain of the golden hamster. Neuroreport 3:
797–800.
Eaton JL, Roache L, Nguyen KN, Cushing BS, Troyer E,
Papademetriou E, Raghanti MA. 2012. Organizational
effects of oxytocin on serotonin innervation. Dev Psycho-
biol 54:92–97.
Elands J, Barberis C, Jard S, Tribollet E, Dreifuss JJ,
Bankowski K, Manning M, et al. 1988. 125I-labelled
d(CH2)5[Tyr(Me)2,Thr4,Tyr-NH2(9)]OVT: A selective
oxytocin receptor ligand. Eur J Pharmacol 147:197–207.
Feldman R. 2015. Sensitive periods in human social devel-
opment: New insights from research on oxytocin, syn-
chrony, and high-risk parenting. Dev Psychopathol 27:
369–395.
Ferguson JN, Aldag JM, Insel TR, Young LJ. 2001. Oxyto-
cin in the medial amygdala is essential for social recogni-
tion in the mouse. J Neurosci 21:8278–8285.
Fertuzinhos S, Li M, Kawasawa YI, Ivic V, Franjic D,
Singh D, Crair M, et al. 2014. Laminar and temporal
expression dynamics of coding and noncoding RNAs in
the mouse neocortex. Cell Rep 6:938–950.
Francis DD, Champagne FC, Meaney MJ. 2000. Variations
in maternal behaviour are associated with differences in
oxytocin receptor levels in the rat. J Neuroendocrinol 12:
1145–1148.
Francis DD, Young LJ, Meaney MJ, Insel TR. 2002. Natu-
rally occurring differences in maternal care are associated
with the expression of oxytocin and vasopressin (V1a)
receptors: Gender differences. J Neuroendocrinol 14:
349–353.
Freeman SM, Inoue K, Smith AL, Goodman MM, Young LJ.
2014a. The neuroanatomical distribution of oxytocin recep-
tor binding and mRNA in the male rhesus macaque
(Macaca mulatta). Psychoneuroendocrinology 45:128–141.
Freeman SM, Smith AL, Goodman MM, Bales KL. 2016.
Selective localization of oxytocin receptors and vasopres-
sin 1a receptors in the human brainstem. Soc Neurosci
10:1–11.
Freeman SM, Walum H, Inoue K, Smith AL, Goodman
MM, Bales KL, Young LJ. 2014b. Neuroanatomical dis-
tribution of oxytocin and vasopressin 1a receptors in the
socially monogamous coppery titi monkey (Callicebus
cupreus). Neuroscience 273:12–23.
Freeman SM, Young LJ, 2016. Comparative perspectives on
oxytocin and vasopressin receptor research in rodents and
primates: Translational implications. J Neuroendocrinol
28, doi:10.1111/jne.12382.
Gimpl G, Fahrenholz F. 2001. The oxytocin receptor sys-
tem: Structure, function, and regulation. Physiol Rev 81:
629–683.
Gong S, Zheng C, Doughty ML, Losos K, Didkovsky N,
Schambra UB, Nowak NJ, et al. 2003. A gene expression
atlas of the central nervous system based on bacterial arti-
ficial chromosomes. Nature 425: 917–925.
Gould BR, Zingg HH. 2003. Mapping oxytocin receptor
gene expression in the mouse brain and mammary gland
using an oxytocin receptor-LacZ reporter mouse. Neuro-
science 122: 155–167.
Grinevich V, Desarmenien MG, Chini B, Tauber M,
Muscatelli F. 2014. Ontogenesis of oxytocin pathways in
the mammalian brain: Late maturation and psychosocial
disorders. Front Neuroanat 8: 164.
Guastella AJ, MacLeod C. 2012. A critical review of the
influence of oxytocin nasal spray on social cognition in
humans: Evidence and future directions. Horm Behav 61:
410–418.
Hammock EA. 2015. Developmental perspectives on oxy-
tocin and vasopressin. Neuropsychopharmacology 40:
24–42.
Hammock E, Levitt P. 2013. Oxytocin receptor ligand
binding in embryonic tissue and postnatal brain develop-
ment of the C57BL/6J mouse. Front Behav Neurosci 7:
195.
Hammock EAD, Levitt P. 2006. The discipline of neurobe-
havioral development: The emerging interface of processes
that build circuits and skills. Hum Dev 49: 294–309.
Harony-Nicolas H, Mamrut S, Brodsky L, Shahar-Gold H,
Barki-Harrington L, Wagner S. 2014. Brain region-
specific methylation in the promoter of the murine oxyto-
cin receptor gene is involved in its expression regulation.
Psychoneuroendocrinology 39: 121–131.
Hashemi F, Tekes K, Laufer R, Szegi P, Tothfalusi L,
Csaba G. 2013. Effect of a single neonatal oxytocin treat-
ment (hormonal imprinting) on the biogenic amine level
of the adult rat brain: Could oxytocin-induced labor cause
pervasive developmental diseases? Reprod Sci 20: 1255–
1263.
Heinrichs M, von Dawans B, Domes G. 2009. Oxytocin,
vasopressin, and human social behavior. Front Neuroen-
docrinol 30: 548–557.
Hidema S, Fukuda T, Hiraoka Y, Mizukami H, Hayashi R,
Otsuka A, Suzuki S, et al. 2016. Generation of Oxtr
cDNA(HA) -Ires-Cre mice for gene expression in an oxy-
tocin receptor specific manner. J Cell Biochem 117:
1099–1111.
Holm VA, Cassidy SB, Butler MG, Hanchett JM,
Greenswag LR, Whitman BY, Greenberg F. 1993. Prader-
Willi syndrome: Consensus diagnostic criteria. Pediatrics
91: 398–402.
Insel TR. 1986. Postpartum increases in brain oxytocin
binding. Neuroendocrinology 44: 515–518.
Insel TR. 1990. Regional changes in brain oxytocin recep-
tors post-partum: Time-course and relationship to mater-
nal behaviour. J Neuroendocrinol 2: 539–545.
Insel TR. 2016. Translating oxytocin neuroscience to the
clinic: A national institute of mental health perspective.
Biol Psychiatry 79: 153–154.
Insel TR, Gelhard R, Shapiro LE. 1991. The comparative
distribution of forebrain receptors for neurohypophyseal
12 Vaidyanathan and Hammock
Developmental Neurobiology
peptides in monogamous and polygamous mice. Neuro-
science 43: 623–630.
Insel TR, Shapiro LE. 1992. Oxytocin receptor distribution
reflects social organization in monogamous and polyga-
mous voles. Proc Natl Acad Sci USA 89: 5981–5985.
Insel TR, Winslow JT. 1991. Central administration of oxy-
tocin modulates the infant rat’s response to social isola-
tion. Eur J Pharmacol 203: 149–152.
Insel TR, Young L, Witt DM, Crews D. 1993. Gonadal ste-
roids have paradoxical effects on brain oxytocin recep-
tors. J Neuroendocrinol 5: 619–628.
Jia R, Tai FD, An SC, Broders H, Ding XL, Kong Q, Zhao
L, et al. 2008b. Effects of neonatal oxytocin treatment on
aggression and neural activities in mandarin voles. Phys-
iol Behav 95: 56–62.
Jia R, Tai F, An S, Broders H, Sun R. 2008a. Neonatal
manipulation of oxytocin influences the partner prefer-
ence in mandarin voles (Microtus mandarinus). Neuro-
peptides 42: 525–533.
Kang HJ, Kawasawa YI, Cheng F, Zhu Y, Xu X, Li M,
Sousa AM, et al. 2011. Spatio-temporal transcriptome of
the human brain. Nature 478: 483–489.
Kasahara Y, Tateishi Y, Hiraoka Y, Otsuka A, Mizukami
H, Ozawa K, Sato K, et al. 2015. Role of the oxytocin
receptor expressed in the rostral medullary raphe in ther-
moregulation during cold conditions. Front Endocrinol
(Lausanne) 6: 180.
Kenkel WM, Yee JR, Carter CS. 2014. Is oxytocin a
maternal-foetal signalling molecule at birth? Implications
for development. J Neuroendocrinol 26:739–749.
Khazipov R, Tyzio R, Ben-Ari Y. 2008. Effects of oxytocin
on GABA signalling in the foetal brain during delivery.
Prog Brain Res 170:243–257.
King LB, Walum H, Inoue K, Eyrich NW, Young LJ. Vari-
ation in the oxytocin receptor gene predicts brain region-
specific expression and social attachment. Biol Psychiatry
DOI: http://dx.doi.org/10.1016/j.biopsych.2015.12.008
Klopfer PH. 1971. Mother love: What turns it on? Am Sci
59:404–407.
Kojima S, Alberts JR. 2011. Warmth from skin-to-skin
contact with mother is essential for the acquisition of fil-
ial huddling preference in preweanling rats. Dev Psycho-
biol 53:813–827.
Kramer KM, Choe C, Carter CS, Cushing BS. 2006. Devel-
opmental effects of oxytocin on neural activation and
neuropeptide release in response to social stimuli. Horm
Behav 49:206–214.
Kramer KM, Cushing BS, Carter CS. 2003. Developmental
effects of oxytocin on stress response: Single versus
repeated exposure. Physiol Behav 79:775–782.
Kumsta R, Hummel E, Chen FS, Heinrichs M. 2013. Epige-
netic regulation of the oxytocin receptor gene: Implica-
tions for behavioral neuroscience. Front Neurosci 7:83.
Kusui C, Kimura T, Ogita K, Nakamura H, Matsumura Y,
Koyama M, Azuma C, et al. 2001. DNA methylation of
the human oxytocin receptor gene promoter regulates
tissue-specific gene suppression. Biochem Biophys Res
Commun 289:681–686.
Landgraf R, Neumann ID. 2004. Vasopressin and oxytocin
release within the brain: A dynamic concept of multiple
and variable modes of neuropeptide communication.
Front Neuroendocrinol 25:150–176.
Lee HJ, Caldwell HK, Macbeth AH, Tolu SG, Young WS
III. 2008a. A conditional knockout mouse line of the oxy-
tocin receptor. Endocrinology 149:3256–3263.
Lee HJ, Caldwell HK, Macbeth AH, Young WS III. 2008b.
Behavioural studies using temporal and spatial inactiva-
tion of the oxytocin receptor. Prog Brain Res 170:73–77.
Loup F, Tribollet E, Dubois-Dauphin M, Dreifuss JJ. 1991.
Localization of high-affinity binding sites for oxytocin
and vasopressin in the human brain. An autoradiographic
study. Brain Res 555:220–232.
Loup F, Tribollet E, Dubois-Dauphin M, Pizzolato G,
Dreifuss JJ. 1989. Localization of oxytocin binding sites
in the human brainstem and upper spinal cord: An autora-
diographic study. Brain Res 500:223–230.
Lukas M, Bredewold R, Neumann ID, Veenema AH. 2010.
Maternal separation interferes with developmental
changes in brain vasopressin and oxytocin receptor bind-
ing in male rats. Neuropharmacology 58:78–87.
Mamrut S, Harony H, Sood R, Shahar-Gold H, Gainer H,
Shi YJ, Barki-Harrington L, et al. 2013. DNA methyla-
tion of specific CpG sites in the promoter region regulates
the transcription of the mouse oxytocin receptor. PLoS
One 8:e56869.
Manning M, Stoev S, Chini B, Durroux T, Mouillac B,
Guillon G. 2008. Peptide and non-peptide agonists and
antagonists for the vasopressin and oxytocin V1a, V1b,
V2 and OT receptors: Research tools and potential thera-
peutic agents. Prog Brain Res 170:473–512.
Marlin BJ, Mitre M, D’Amour JA, Chao MV, Froemke RC.
2015. Oxytocin enables maternal behaviour by balancing
cortical inhibition. Nature 520:499–504.
Mazzuca M, Minlebaev M, Shakirzyanova A, Tyzio R,
Taccola G, Janackova S, Gataullina S, et al. 2011. New-
born analgesia mediated by oxytocin during delivery.
Front Cell Neurosci 5:3.
Meyer-Lindenberg A. 2008. Impact of prosocial neuropepti-
des on human brain function. Prog Brain Res 170:463–470.
Meziane H, Schaller F, Bauer S, Villard C, Matarazzo V,
Riet F, Guillon G, et al. 2015. An early postnatal oxytocin
treatment prevents social and learning deficits in adult
mice deficient for magel2, a gene involved in Prader-
Willi syndrome and autism. Biol Psychiatry 78: 85–94.
Miller TV, Caldwell HK. 2015. Oxytocin during develop-
ment: Possible organizational effects on behavior. Front
Endocrinol (Lausanne) 6:76.
Mitre M, Marlin BJ, Schiavo JK, Morina E, Norden SE,
Hackett TA, Aoki CJ, et al. 2016. A distributed network
for social cognition enriched for oxytocin receptors.
J Neurosci 36:2517–2535.
Mogi K, Ooyama R, Nagasawa M, Kikusui T. 2014. Effects
of neonatal oxytocin manipulation on development of
social behaviors in mice. Physiol Behav 133:68–75.
OXT Receptor Dynamics in the Brain 13
Developmental Neurobiology
Murata Y, Li MZ, Masuko S. 2011. Developmental expres-
sion of oxytocin receptors in the neonatal medulla oblon-
gata and pons. Neurosci Lett 502:157–161.
Nelson E, Alberts JR. 1997. Oxytocin-induced paw sucking
in infant rats. Ann N Y Acad Sci 807:543–545.
Neumann ID, Landgraf R. 2012. Balance of brain oxytocin
and vasopressin: Implications for anxiety, depression,
and social behaviors. Trends Neurosci 35:649–659.
Noonan LR, Caldwell JD, Li L, Walker CH, Pedersen CA,
Mason GA. 1994. Neonatal stress transiently alters the
development of hippocampal oxytocin receptors. Brain
Res Dev Brain Res 80:115–120.
Olazabal DE, Young LJ. 2006. Species and individual dif-
ferences in juvenile female alloparental care are associ-
ated with oxytocin receptor density in the striatum and
the lateral septum. Horm Behav 49:681–687.
Ophir AG, Gessel A, Zheng DJ, Phelps SM. 2012. Oxyto-
cin receptor density is associated with male mating tactics
and social monogamy. Horm Behav 61:445–453.
Pedersen CA, Ascher JA, Monroe YL, Prange AJ Jr. 1982.
Oxytocin induces maternal behavior in virgin female rats.
Science 216:648–650.
Pedersen CA, Caldwell JD, Drago F, Noonan LR, Peterson
G, Hood LE, Prange AJ Jr. 1988. Grooming behavioral
effects of oxytocin. Pharmacology, ontogeny, and com-
parisons with other nonapeptides. Ann N Y Acad Sci
525:245–256.
Pedersen CA, Prange AJ Jr. 1979. Induction of maternal
behavior in virgin rats after intracerebroventricular
administration of oxytocin. Proc Natl Acad Sci USA 76:
6661–6665.
Pena CJ, Neugut YD, Champagne FA. 2013. Developmen-
tal timing of the effects of maternal care on gene expres-
sion and epigenetic regulation of hormone receptor levels
in female rats. Endocrinology 154:4340–4351.
Perry AN, Paramadilok A, Cushing BS. 2009. Neonatal
oxytocin alters subsequent estrogen receptor alpha pro-
tein expression and estrogen sensitivity in the female rat.
Behav Brain Res 205:154–161.
Pournajafi-Nazarloo H, Carr MS, Papademeteriou E,
Schmidt JV, Cushing BS. 2007. Oxytocin selectively
increases ERalpha mRNA in the neonatal hypothalamus
and hippocampus of female prairie voles. Neuropeptides
41:39–44.
Rabhi M, Stoeckel ME, Calas A, Freund-Mercier MJ.
1999. Historadioautographic localisation of oxytocin and
vasopressin binding sites in the central nervous system of
the merione (Meriones shawi). Brain Res Bull 48:147–
163.
Ross HE, Freeman SM, Spiegel LL, Ren X, Terwilliger EF,
Young LJ. 2009. Variation in oxytocin receptor density
in the nucleus accumbens has differential effects on affili-
ative behaviors in monogamous and polygamous voles.
J Neurosci 29:1312–1318.
Rozas C, Frank H, Heynen AJ, Morales B, Bear MF,
Kirkwood A. 2001. Developmental inhibitory gate con-
trols the relay of activity to the superficial layers of the
visual cortex. J Neurosci 21:6791–6801.
Schaller F, Watrin F, Sturny R, Massacrier A, Szepetowski
P, Muscatelli F. 2010. A single postnatal injection of oxy-
tocin rescues the lethal feeding behaviour in mouse new-
borns deficient for the imprinted Magel2 gene. Hum Mol
Genet 19:4895–4905.
Shapiro LE, Insel TR. 1989. Ontogeny of oxytocin recep-
tors in rat forebrain: A quantitative study. Synapse 4:
259–266.
Smearman EL, Almli LM, Conneely KN, Brody GH, Sales
JM, Bradley B, Ressler KJ, et al. 2016. Oxytocin receptor
genetic and epigenetic variations: Association with child
abuse and adult psychiatric symptoms. Child Dev 87:
122–134.
Smith AL, Freeman SM, Stehouwer JS, Inoue K, Voll RJ,
Young LJ, Goodman MM. 2012. Synthesis and evalua-
tion of C-11, F-18 and I-125 small molecule radioligands
for detecting oxytocin receptors. Bioorg Med Chem 20:
2721–2738.
Smith AL, Freeman SM, Voll RJ, Young LJ, Goodman
MM. 2013a. Carbon-11 N-methyl alkylation of L-
368,899 and in vivo PET imaging investigations for neu-
ral oxytocin receptors. Bioorg Med Chem Lett 23:902–
906.
Smith AL, Freeman SM, Voll RJ, Young LJ, Goodman
MM. 2013b. Investigation of an F-18 oxytocin receptor
selective ligand via PET imaging. Bioorg Med Chem Lett
23:5415–5420.
Snijdewint FG, Van Leeuwen FW, Boer GJ. 1989. Ontog-
eny of vasopressin and oxytocin binding sites in the brain
of Wistar and Brattleboro rats as demonstrated by light-
microscopical autoradiography. J Chem Neuroanat 2:3–
17.
Soloff MS, Alexandrova M, Fernstrom MJ. 1979. Oxytocin
receptors: Triggers for parturition and lactation? Science
204:1313–1315.
Takayanagi Y, Yoshida M, Bielsky IF, Ross HE,
Kawamata M, Onaka T, Yanagisawa T, et al. 2005. Per-
vasive social deficits, but normal parturition, in oxytocin
receptor-deficient mice. Proc Natl Acad Sci USA 102:
16096–16101.
Tamborski S, Mintz EM, Caldwell HK. 2016. Sex differen-
ces in the embryonic development of the central oxytocin
system in mice. J Neuroendocrinol 28, doi:10.1111/
jne.12364.
Toloczko DM, Young L, Insel TR. 1997. Are there oxyto-
cin receptors in the primate brain? Ann N Y Acad Sci
807:506–509.
Tribollet E, Audigier S, Dubois-Dauphin M, Dreifuss JJ.
1990. Gonadal steroids regulate oxytocin receptors but
not vasopressin receptors in the brain of male and female
rats. An autoradiographical study. Brain Res 511:129–
140.
Tribollet E, Dubois-Dauphin M, Dreifuss JJ, Barberis C,
Jard S. 1992. Oxytocin receptors in the central nervous
system. Distribution, development, and species differen-
ces. Ann N Y Acad Sci 652: 29–38.
Tribollet E, Charpak S, Schmidt A, Dubois-Dauphin M,
Dreifuss JJ. 1989. Appearance and transient expression of
14 Vaidyanathan and Hammock
Developmental Neurobiology
oxytocin receptors in fetal, infant, and peripubertal rat
brain studied by autoradiography and electrophysiology.
J Neurosci 9: 1764–1773.
Tyzio R, Cossart R, Khalilov I, Minlebaev M, Hubner CA,
Represa A, Ben-Ari Y, et al. 2006. Maternal oxytocin trig-
gers a transient inhibitory switch in GABA signaling in the
fetal brain during delivery. Science 314: 1788–1792.
Tyzio R, Nardou R, Ferrari DC, Tsintsadze T, Shahrokhi
A, Eftekhari S, Khalilov I, et al. 2014. Oxytocin-mediated
GABA inhibition during delivery attenuates autism
pathogenesis in rodent offspring. Science 343: 675–679.
Uhl-Bronner S, Waltisperger E, Martinez-Lorenzana G,
Condes Lara M, Freund-Mercier MJ. 2005. Sexually
dimorphic expression of oxytocin binding sites in fore-
brain and spinal cord of the rat. Neuroscience 135: 147–
154.
Unternaehrer E, Meyer AH, Burkhardt SC, Dempster E,
Staehli S, Theill N, Lieb R, et al. 2015. Childhood mater-
nal care is associated with DNA methylation of the genes
for brain-derived neurotrophic factor (BDNF) and oxyto-
cin receptor (OXTR) in peripheral blood cells in adult
men and women. Stress 18: 451–461.
Veenema AH. 2012. Toward understanding how early-life
social experiences alter oxytocin- and vasopressin-
regulated social behaviors. Horm Behav 61: 304–312.
Wang Z, Young LJ. 1997. Ontogeny of oxytocin and vaso-
pressin receptor binding in the lateral septum in prairie
and montane voles. Brain Res Dev Brain Res 104: 191–
195.
Welch MG, Margolis KG, Li Z, Gershon MD. 2014. Oxyto-
cin regulates gastrointestinal motility, inflammation, mac-
romolecular permeability, and mucosal maintenance in
mice. Am J Physiol Gastrointest Liver Physiol 307:
G848–G862.
Welch MG, Tamir H, Gross KJ, Chen J, Anwar M,
Gershon MD. 2009. Expression and developmental regu-
lation of oxytocin (OT) and oxytocin receptors (OTR) in
the enteric nervous system (ENS) and intestinal epithe-
lium. J Comp Neurol 512: 256–270.
Williams JR, Carter CS, Insel T. 1992. Partner preference
development in female prairie voles is facilitated by mat-
ing or the central infusion of oxytocin. Ann N Y Acad
Sci 652: 487–489.
Yamamoto Y, Carter CS, Cushing BS. 2006. Neonatal
manipulation of oxytocin affects expression of estrogen
receptor alpha. Neuroscience 137: 157–164.
Yamamoto Y, Cushing BS, Kramer KM, Epperson PD,
Hoffman GE, Carter CS. 2004. Neonatal manipulations
of oxytocin alter expression of oxytocin and vasopressin
immunoreactive cells in the paraventricular nucleus of
the hypothalamus in a gender-specific manner. Neuro-
science 125: 947–955.
Yoshida M, Takayanagi Y, Inoue K, Kimura T, Young LJ,
Onaka T, Nishimori K. 2009. Evidence that oxytocin
exerts anxiolytic effects via oxytocin receptor expressed
in serotonergic neurons in mice. J Neurosci 29: 2259–
2271.
Yoshimura R, Kimura T, Watanabe D, Kiyama H. 1996.
Differential expression of oxytocin receptor mRNA in the
developing rat brain. Neurosci Res 24: 291–304.
Young LJ. 1999. Frank A. Beach Award. Oxytocin and
vasopressin receptors and species-typical social behav-
iors. Horm Behav 36: 212–221.
Young LJ. 2013. When too much of a good thing is bad:
Chronic oxytocin, development, and social impairments.
Biol Psychiatry 74: 160–161.
Young LJ, Huot B, Nilsen R, Wang Z, Insel TR. 1996. Spe-
cies differences in central oxytocin receptor gene expres-
sion: Comparative analysis of promoter sequences.
J Neuroendocrinol 8: 777–783.
Young LJ, Lim MM, Gingrich B, Insel TR. 2001. Cellular
mechanisms of social attachment. Horm Behav 40: 133–
138.
Young LJ, Muns S, Wang Z, Insel TR. 1997. Changes in
oxytocin receptor mRNA in rat brain during pregnancy
and the effects of estrogen and interleukin-6.
J Neuroendocrinol 9: 859–865.
Young LJ, Wang Z, Donaldson R, Rissman EF. 1998.
Estrogen receptor alpha is essential for induction of oxy-
tocin receptor by estrogen. Neuroreport 9: 933–936.
Zheng JJ, Li SJ, Zhang XD, Miao WY, Zhang D, Yao H,
Yu X. 2014. Oxytocin mediates early experience-
dependent cross-modal plasticity in the sensory cortices.
Nat Neurosci 17: 391–399.
Zingg HH, Laporte SA. 2003. The oxytocin receptor.
Trends Endocrinol Metab 14: 222–227.
Zink CF, Meyer-Lindenberg A. 2012. Human neuroimag-
ing of oxytocin and vasopressin in social cognition. Horm
Behav 61: 400–409.
OXT Receptor Dynamics in the Brain 15
Developmental Neurobiology
... First, we recognize that results reported here may not be translatable to humans because of significant species differences in the permeability of the placental and the fetal blood-brain barriers that may ultimately affect the extent of Oxt transfer, 50,72,73 and variations in the distribution of the Oxtr in the brain that may modify the potential consequences. 74,75 At best, our results provide evidential support for Oxtr downregulation in the fetal brain after labor induction with Oxt, which until now, has largely been a theoretical concern. 76,77 Second, considerable species differences in neurodevelopmental ontogeny could potentially influence the interpretation of our neurobehavioral outcomes. ...
... 76,77 Second, considerable species differences in neurodevelopmental ontogeny could potentially influence the interpretation of our neurobehavioral outcomes. 74,75,78,79 Though performing these studies in P10 pups, the equivalent of a term human neonate, 78 would seem ideal, it is confounded by the rich social-sensory experiences of the pups during maternal nurturing and littermate contact during the first 10 days of life, in contrast to the absence of these experiences with the in utero Oxt exposure paradigm relevant to obstetric practice. Third, because of constraints related to the size of the post-weaning rats, we performed functional connectivity experiments between P25-30. ...
Article
Full-text available
Despite six decades of the use of exogenous oxytocin for management of labor, little is known about its effects on the developing brain. Motivated by controversial reports suggesting a link between oxytocin use during labor and autism spectrum disorders (ASDs), we employed our recently validated rat model for labor induction with oxytocin to address this important concern. Using a combination of molecular biological, behavioral, and neuroimaging assays, we show that induced birth with oxytocin leads to sex-specific disruption of oxytocinergic signaling in the developing brain, decreased communicative ability of pups, reduced empathy-like behaviors especially in male offspring, and widespread sex-dependent changes in functional cortical connectivity. Contrary to our hypothesis, social behavior, typically impaired in ASDs, was largely preserved. Collectively, our foundational studies provide nuanced insights into the neurodevelopmental impact of birth induction with oxytocin and set the stage for mechanistic investigations in animal models and prospective longitudinal clinical studies.
... Oxytocin neurons in caudal regions of the PVH coordinate various aspects of ingestive behavior and autonomic regulation (15)(16)(17)(18)(19)(20)(21)(22)(23). Oxytocin neural circuits mature relatively late in development, as mature oxytocin peptide is not detected until after birth, and axon outgrowth progressively increases during the first few weeks of postnatal life (24)(25)(26). Descending oxytocin projections that arise from caudal PVH neurons and target the DVC are negligible at postnatal day 0 (P0), but markedly increase in density postnatally, reaching adult-like levels by weaning (27). ...
Preprint
During postnatal life, the adipocyte-derived hormone leptin is required for proper targeting of neural inputs to the paraventricular nucleus of the hypothalamus (PVH) and impacts the activity of neurons containing agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus. Activity-dependent developmental mechanisms are known to play a defining role during postnatal organization of neural circuits, but whether leptin-mediated postnatal neuronal activity specifies neural projections to the PVH or impacts downstream connectivity is largely unexplored. Here, we blocked neuronal activity of AgRP neurons during a discrete postnatal period and evaluated development of AgRP inputs to defined regions in the PVH, as well as descending projections from PVH oxytocin neurons to the dorsal vagal complex (DVC) and assessed their dependence on leptin or postnatal AgRP neuronal activity. In leptin-deficient mice, AgRP inputs to PVH neurons were significantly reduced, as well as oxytocin-specific neuronal targeting by AgRP. Moreover, downstream oxytocin projections from the PVH to the DVC were also impaired, despite the lack of leptin receptors found on PVH oxytocin neurons. Blocking AgRP neuron activity specifically during early postnatal life reduced the density of AgRP inputs to the PVH, as well as the density of projections from PVH oxytocin neurons to the DVC, and these innervation deficits were associated with dysregulated autonomic function. These findings suggest that postnatal targeting of descending PVH oxytocin projections to the DVC requires leptin-mediated AgRP neuronal activity, and represents a novel activity-dependent mechanism for hypothalamic specification of metabolic circuitry, with consequences for autonomic regulation. Significance statement Hypothalamic neural circuits maintain homeostasis by coordinating endocrine signals with autonomic responses and behavioral outputs to ensure that physiological responses remain in tune with environmental demands. The paraventricular nucleus of the hypothalamus (PVH) plays a central role in metabolic regulation, and the architecture of its neural inputs and axonal projections is a defining feature of how it receives and conveys neuroendocrine information. In adults, leptin regulates multiple aspects of metabolic physiology, but it also functions during development to direct formation of circuits controlling homeostatic functions. Here we demonstrate that leptin acts to specify the input-output architecture of PVH circuits through an activity-dependent, transsynaptic mechanism, which represents a novel means of sculpting neuroendocrine circuitry, with lasting effects on how the brain controls energy balance.
... Hence, hormones like oxytocin could play a central role in attachment development. Oxytocin is a neuropeptide that is produced in the hypothalamus and is released from the posterior pituitary gland (Vaidyanathan & Hammock, 2017). ...
Article
The current study explored longitudinally whether oxytocin receptor gene methyla-tion (OXTRm) changes moderated the association between parental sensitivity changes and children's attachment changes over three waves. Six hundred six Flemish children (10-12 years, 42.8%-44.8% boys) completed attachment measures and provided sali-vary OXTRm data on seven CpG sites. Their parents reported their sensitive parenting. Results suggest that OXTRm changes hardly link to attachment (in)security changes after the age of 10. Some support was found for interaction effects between parental sensitivity changes and OXTRm changes on attachment changes over time. Effects suggest that for children with increased OXTRm in the promotor region and decreased methylation in the inhibitor region over time, increased parental sensitivity was associated with increased secure attachment and decreased insecure attachment over time.
... A putative endocrinological correlate of attachment is oxytocin (Feldman & Bakermans-Kranenburg, 2017). Oxytocin is a neurohormone produced in the hypothalamus and released from the posterior pituitary gland (Vaidyanathan & Hammock, 2017). Its most commonly known function is to induce labor and breastfeeding, but studies have also found oxytocin to be related to social affiliation (Winslow & Insel, 2002), and the establishment of social and attachment bonding (Swain et al., 2014). ...
... Reduced protein expression may be a biological mediator of some aspects of the psychogenesis and psychopathology of BPD. Notably, oxytocin's effects at the central level largely depend on the dynamics and distribution of its receptors in different brain areas (Vaidyanathan and Hammock, 2017). According to some authors (Herpertz and Bertsch, 2015), a deficit in the oxytocin system could be related to the abnormal stress response and emotional instability characteristic of BPD. ...
... Oxytocin exerts its effects via the oxytocin receptor (OXTR) [21]. OXTR is a G-protein-coupled receptor (GPCR), mostly expressed in the brain but also present in other body tissues, and has been proven to be involved in the development of social skills [38]. This prompted researchers to examine the OXTR gene as a prospective candidate for depression susceptibility. ...
Article
Full-text available
Background Depression is a common psychiatric disorder that negatively affects mood and thoughts. Association studies of OXTR polymorphisms with depression have been performed repeatedly. However, the results of these studies were inconsistent. The aim of the present study was to perform a meta-analysis of case–control studies that have investigated the relationship between the OXTR polymorphism (rs53576) and depression risk. Methods Four databases, PubMed, ScienceDirect, Springer Link, and Google Scholar, were searched, and a total of 10 studies were involved in the meta-analysis. ReviewManager (RevMan) 5.4 software was used to perform a meta-analysis of the eligible studies. Results A significant association between OXTR rs53576 and depression was found in the recessive model (Odds Ratio (OR) AA vs. AG + GG = 1.28, 95% Confidence Interval (CI) [1.02–1.59], P = 0.03), while there was no association with the other two genetic models (dominant model: OR AA + AG vs. GG = 1.01, 95% CI [0.87–1.18], P = 0.87; allelic model: OR A vs. G = 0.95, 95% CI [0.83–1.09], P = 0.46). A significant association was observed in the Caucasian populations (OR 1.29, 95% CI [1.01, 1.64], P = 0.04), while the Asian populations showed no significant association (OR 1.22, 95% CI [0.71, 2.09], P = 0.48). Conclusions This meta-analysis is to date the first to provide a comprehensive investigation of the association of the OXTR rs53576 polymorphism with depression, and its results reflect the data currently available from the literature and can serve as a guide for further research.
... There was a similar increase in female IT expression around sexual maturity. Across vertebrate species, it has been repeatedly demonstrated that oxytocin/vasopressin systems develop in a way that influences subsequent expression of reproductive and parenting behaviors (e.g., Hammock, 2015;Vaidyanathan & Hammock, 2017). Based on conserved functions observed across vertebrates, it is possible that IT is involved in priming beetles for reproduction, but further research is needed to determine how the development of inotocinergic system is related to reproduction. ...
Article
Full-text available
Parental care is thought to evolve through modification of behavioral precursors, which predicts that the mechanistic changes occur in the genes underlying those traits. The duplicated gene system of oxytocin/vasopressin has been broadly co-opted across vertebrates to influence parenting, from a pre-duplication ancestral role in water balance. It remains unclear whether co-option of these genes for parenting is limited to vertebrates. Here, we experimentally tested for associations between inotocin gene expression and water balance, parental acceptance of offspring, and active parenting in the subsocial beetle Nicrophorus orbicollis, to test whether this single copy homologue of the oxytocin/vasopressin system, has similarly been co-opted for parental care in a species with elaborate parenting. As expected, inotocin was associated with water balance in both sexes. Inotocin expression increased around sexual maturation in both males and females, although more clearly in males. Finally, we found that expression of inotocin was not associated with acceptance of larvae but was associated with a transition to male but not female parenting. Moreover, level of offspring provisioning behavior and gene expression were positively correlated in males but uncorrelated in females. Our results suggest a broad co-option of this system for parenting that may have existed prior to gene duplication, and that inotocin may be associated with flexibility in parenting behavior.
Article
Full-text available
Introduction: Dysfunctions in the oxytocin system have been reported in patients with borderline personality disorder (BPD). Deficits could be related to interpersonal hypersensitivity, which has been previously associated with failures in social cognition (SC) in this disorder, especially in Theory of Mind (ToM) skills. The aim of this work is to study the links between the oxytocin system and SC impairments in patients with BPD. Method: Plasma oxytocin levels (OXT) and protein expression of oxytocin receptors in blood mononuclear cells (OXTR) were examined in 33 patients with a diagnosis of BPD (age: M 28.85, DT = 8.83). Social cognition was assessed using the Movie for the Assessment of Social Cognition (MASC). Statistical associations between biochemical factors and different response errors in MASC were analyzed through generalized linear regression controlling for relevant clinical factors. Results: Generalized linear regression showed a significant relationship between lower OXTR and overmentalization in BPD patients (OR = 0.90). Conclusions: This work supports the relationship between alterations in the oxytocin system and ToM impairments observed in BPD patients, enhancing the search for endophenotypes related to the phenotypic features of the disorder to improve current clinical knowledge and address more specific therapeutic targets.
Article
Chronic exposure to trauma and violence can promote aggressive behavior. Oxytocin and variants in the oxytocin receptor (OXTR) gene may play a role in the etiology of proactive, that is, goal‐oriented instrumental aggression, or reactive aggression, which typically occurs in response to emotionally triggering situations. The current study builds on previous findings that experienced and witnessed trauma in childhood predicts higher levels of appetitive aggression, a form of proactive aggression characterized by the enjoyment of participating in violent behavior. The current study explores the role of OXTR rs2254298 and rs53576 variants in appetitive and reactive aggression. Adult males living in Cape Town, South Africa, and at risk for violent behavior completed the Appetitive Aggression Scale (AAS) and Buss–Perry Aggression Questionnaire (BPAQ). OXTR rs2254298 and rs53576 were successfully genotyped via restriction fragment length polymorphism (RFLP) analysis in 238 and 239 participants, respectively. Regression analysis showed that rs2254298 G/G and A/G genotypes and the rs53576 A/G genotype were significantly associated with lower AAS scores ( p < .001) compared to the A/A genotype. Additionally, genotype interaction analyses conducted in 232 participants, found that the combination of rs2254298 A/G and rs53576 G/G genotypes produced opposite effects on appetitive and reactive aggression. Specifically, this combination was associated with a 0.29‐point increase in AAS scores ( p = .032) and a 0.13‐point decrease in BPAQ scores ( p = .037) when compared to A‐allele homozygosity for both variants. These results suggest that genetic variation in a signaling system involved in influencing environmental and social salience may contribute to appetitive aggression.
Article
Full-text available
Oxytocin is a neuropeptide important for social behaviors such as maternal care and parent–infant bonding. It is believed that oxytocin receptor signaling in the brain is critical for these behaviors, but it is unknown precisely when and where oxytocin receptors are expressed or which neural circuits are directly sensitive to oxytocin. To overcome this challenge, we generated specific antibodies to the mouse oxytocin receptor and examined receptor expression throughout the brain. We identified a distributed network of female mouse brain regions for maternal behaviors that are especially enriched for oxytocin receptors, including the piriform cortex, the left auditory cortex, and CA2 of the hippocampus. Electron microscopic analysis of the cerebral cortex revealed that oxytocin receptors were mainly expressed at synapses, as well as on axons and glial processes. Functionally, oxytocin transiently reduced synaptic inhibition in multiple brain regions and enabled long-term synaptic plasticity in the auditory cortex. Thus modulation of inhibition may be a general mechanism by which oxytocin can act throughout the brain to regulate parental behaviors and social cognition. SIGNIFICANCE STATEMENT Oxytocin is an important peptide hormone involved in maternal behavior and social cognition, but it has been unclear what elements of neural circuits express oxytocin receptors due to the paucity of suitable antibodies. Here, we developed new antibodies to the mouse oxytocin receptor. Oxytocin receptors were found in discrete brain regions and at cortical synapses for modulating excitatory-inhibitory balance and plasticity. These antibodies should be useful for future studies of oxytocin and social behavior.
Article
Full-text available
Background: Oxytocin (OXT) modulates several aspects of social behavior. Intranasal OXT is a leading candidate for treating social deficits in patients with autism spectrum disorder, and common genetic variants in the human OXTR gene are associated with emotion recognition, relationship quality, and autism spectrum disorder. Animal models have revealed that individual differences in Oxtr expression in the brain drive social behavior variation. Our understanding of how genetic variation contributes to brain OXTR expression is very limited. Methods: We investigated Oxtr expression in monogamous prairie voles, which have a well-characterized OXT system. We quantified brain region-specific levels of Oxtr messenger RNA and oxytocin receptor protein with established neuroanatomic methods. We used pyrosequencing to investigate allelic imbalance of Oxtr mRNA, a molecular signature of polymorphic genetic regulatory elements. We performed next-generation sequencing to discover variants in and near the Oxtr gene. We investigated social attachment using the partner preference test. Results: Our allelic imbalance data demonstrate that genetic variants contribute to individual differences in Oxtr expression, but only in particular brain regions, including the nucleus accumbens, where oxytocin receptor signaling facilitates social attachment. Next-generation sequencing identified one polymorphism in the Oxtr intron, near a putative cis-regulatory element, explaining 74% of the variance in striatal Oxtr expression specifically. Males homozygous for the high expressing allele display enhanced social attachment. Conclusions: Taken together, these findings provide convincing evidence for robust genetic influence on Oxtr expression and provide novel insights into how noncoding polymorphisms in OXTR might influence individual differences in human social cognition and behavior.
Article
Oxytocin (OT) knock-out mice fail to recognize familiar conspecifics after repeated social exposures, despite normal olfactory and spatial learning abilities. OT treatment fully restores social recognition. Here we demonstrate that OT acts in the medial amygdala during the initial exposure to facilitate social recognition. OT given before, but not after, the initial encounter restores social recognition in OT knock-out mice. Using c-Fos immunoreactivity (Fos-IR) as a marker of neuronal activation in this initial encounter, we found similar neuronal activation in the wild-type (WT) and OT knock-out mouse in olfactory bulbs, piriform cortex, cortical amygdala, and the lateral septum. Wild-type, but not OT knock-out mice exhibited an induction of Fos-IR in the medial amygdala. Projections sites of the medial amygdala also failed to show a Fos-IR induction in the OT knock-out mice. OT knock-out, but not WT, mice showed dramatic increases in Fos-IR in the somatosensory cortex and the hippocampus, suggesting alternative processing of social cues in these animals. With site-specific injections of OT and an OT antagonist, we demonstrate that OT receptor activation in the medial amygdala is both necessary and sufficient for social recognition in the mouse.
Article
In the last several decades, sophisticated experimental techniques have been used to elucidate the neurobiology of the oxytocin and vasopressin systems in rodents. Using a suite of methodologies, including electrophysiology, site-specific selective pharmacology, receptor autoradiography, in vivo microdialysis, and genetic and optogenetic manipulations, we have gained unprecedented knowledge about how these neuropeptides engage neural circuits to regulate behavior, particularly social behavior. Based on this foundation of information from rodent studies, we have started generating new hypotheses and frameworks about how the oxytocin and vasopressin systems could be acting in humans to influence social cognition. However, despite the recent inundation of publications using intranasal oxytocin in humans, we still know very little about the neurophysiology of the oxytocin system in primates more broadly. Furthermore, the design and analysis of these human studies have remained largely uninformed of the potential neurobiological mechanisms underlying their findings. While the methods available to study the oxytocin and vasopressin systems in humans are incredibly limited due to practical and ethical considerations, there is great potential to fill the gaps in our knowledge by developing better nonhuman primate models of social functioning. Behavioral pharmacology and receptor autoradiography have been used to study the oxytocin and vasopressin systems in nonhuman primates, and there is now great potential to broaden our understanding of the neurobiology of these systems. In this review, we discuss comparative findings in receptor distributions in rodents and primates, with perspectives on the functionality of conserved regions of expression in these distinct mammalian clades. We also identify specific ways that established technologies can be used to answer basic research questions in primates. Finally, we highlight areas of future research in nonhuman primates that are experimentally poised to yield critical insights into the anatomy, physiology, and behavioral effects of the oxytocin system, given its remarkable translational potential. This article is protected by copyright. All rights reserved.
Article
Intranasal oxytocin affects a suite of human social behaviors, including trust, eye contact, and emotion recognition. However, it is unclear where oxytocin receptors (OXTR) and the structurally related vasopressin 1a receptors (AVPR1a) are expressed in the human brain. We have previously described a reliable, pharmacologically informed receptor autoradiography protocol for visualizing these receptors in postmortem primate brain tissue. We used this technique in human brainstem tissue to identify the neural targets of oxytocin and vasopressin. To determine binding selectivity of the OXTR radioligand and AVPR1a radioligand, sections were incubated in four conditions: radioligand alone, radioligand with the selective AVPR1a competitor SR49059, and radioligand with a low or high concentration of the selective OXTR competitor ALS-II-69. We found selective OXTR binding in the spinal trigeminal nucleus, a conserved region of OXTR expression in all primate species investigated to date. We found selective AVPR1a binding in the nucleus prepositus, an area implicated in eye gaze stabilization. The tissue's postmortem interval was not correlated with either the specific or nonspecific binding of either radioligand, indicating that it will not likely be a factor in similar postmortem studies. This study provides critical data for future studies of OXTR and AVPR1a in human brain tissue.
Article
Childhood abuse can alter biological systems and increase risk for adult psychopathology. Epigenetic mechanisms, alterations in DNA structure that regulate the gene expression, are a potential mechanism underlying this risk. While abuse associates with methylation of certain genes, particularly those in the stress response system, no study to date has evaluated abuse and methylation of the oxytocin receptor (OXTR). However, studies support a role for OXTR in the link between abuse and adverse adult outcomes, showing that abuse can confer greater risk for psychiatric symptoms in those with specific OXTR genotypes. This study therefore sought to (a) assess the role of epigenetics in the link between abuse and psychopathology and (b) begin to integrate the genetic and epigenetic literature by exploring associations between OXTR genotypes and DNA CpG methylation. Data on 18 OXTR CpG sites, 44 single nucleotide polymorphisms, childhood abuse, and adult depression and anxiety symptoms were assessed in 393 African American adults (age = 41 ± 12.8 years). Overall, 68% of genotypes were associated with methylation of nearby CpG sites, with a subset surviving multiple test correction. Child abuse associated with higher methylation of two CpG sites yet did not survive correction or serve as a mediator of psychopathology. However, abuse interacted with CpG methylation to predict psychopathology. These findings suggest a role for OXTR in understanding the influence of early environments on adult psychiatric symptoms.