You are on page 1of 10

Journal of Comparative Physiology A

https://doi.org/10.1007/s00359-019-01376-8

REVIEW

A new view of sexual differentiation of mammalian brain


Margaret M. McCarthy1 

Received: 29 July 2019 / Revised: 16 October 2019 / Accepted: 23 October 2019


© Springer-Verlag GmbH Germany, part of Springer Nature 2019

Abstract
Establishment of enduring sex differences in brain and behavior occurs during pre- or perinatal development, depending on
species. For over 50 years the focus has been on gonadal steroid production by male fetuses and the impact on developing
brain. An increasing awareness of the importance of sex chromosome complement has broadened the focus but identifying
specific roles in development has yet to be achieved. Recent emphasis on transcriptomics has revealed myriad and unexpected
differences in gene expression in specific regions of male and female brains which may produce sex differences, serve a
compensatory role or provide latent sex differences revealed only in response to challenge. More surprising, however, has
been the consistent observation of a central role for inflammatory signaling molecules and immune cells in masculinization
of brain and behavior. The signal transduction pathways and specific immune cells vary by brain region, as does the neu-
roanatomical substrate subject to differentiation, reflecting substantial complexity emerging from what may be a common
origin, the maternal immune system. A working hypothesis integrating these various ideas is proposed.

Keywords  Androgens · Estrogens · Amygdala · Preoptic area · Prostaglandins

Introduction Sex determination begins with chromosome


complement
Sexual differentiation of the brain is a developmental pro-
cess occurring during a restricted critical window which is The biological construct of maleness and femaleness begins
operationally defined by the onset of androgen production by with the establishment of sex chromosome complement in
the fetal testis of males. The closing of the critical window the fertilized zygote and ends with reproductive competence
is when the blockade or removal of androgens in males is of the mature adult (Fig. 1). In mammals, a single gene on
without effect or similarly, treating females with testoster- the Y chromosome, Sry (Sex-determining region of the Y),
one is ineffective at inducing masculinization. In the first codes for the testis-determining factor protein (Tdf), which
scenario the process of masculinization has proceeded to initiates a cascade of gene expression within days of fer-
the point of no return and the same is true for feminization tilization that will lead to the creation of a testis from the
in the second scenario. Defining the critical period as begin- bipotential gonadal anlage (Goodfellow and Lovell-Badge
ning with the onset of hormone secretion and ending with 1993). If there is no Y chromosome, i.e. XX, or if the Sry
loss of sensitivity to hormones frames our view of the entire gene is missing or mutated, the gonadal anlage will proceed
process as being driven by hormones. This myopic view down a developmental pathway that culminates in the forma-
prevents consideration of other plausible biological variables tion of an ovary. As the embryo develops the formation of
that may be equally, if not more important, for establishing the reproductive tract and external genitalia are sculpted to
and maintaining sex differences in physiology and behavior. match the demands created by gonadal phenotype. One of
This review will highlight the importance of two such vari- the most profound consequences of the formation of a testis
ables, sex chromosome complement and the immune system. versus an ovary is not the production of many gametes ver-
sus only a few or the cyclical versus static production of ster-
oid hormones and creation of a sex specific hormonal milieu.
* Margaret M. McCarthy The most important difference is that one gonad, the testis, is
mmccarth@umaryland.edu
engineered to release gametes outside the body and therefore
1
Department of Pharmacology, University of Maryland, must be transferred to a viable mate, while the other gonad,
School of Medicine, MD, Baltimore, USA

13
Vol.:(0123456789)
Journal of Comparative Physiology A

Fig. 1  Classic view of sexual differentiation of the brain. Sexual dif- The female ovary will develop in the absence of signals from the
ferentiation begins with chromosome complement and is driven by Sry-induced gene expression cascade and will remain quiescent until
the Sry gene of the Y chromosome directing the formation of a testis puberty when cyclical production of estrogens and progestins begins.
from the bipotential gonad. Testosterone production by the resultant There is evidence for a sensitive period for feminization that occurs
testis will commence during late gestation and into the early postnatal slightly later in development. The masculinizing actions of testoster-
period of the rodent and masculinize select regions of the male brain. one and its aromatized by product, estradiol, are considered organi-
A separate process of defeminizaton specific to sex behavior removes zational and are essential for the activational effects of hormones at
the capacity for female-like receptive behavior (i.e. lordosis) in males. puberty on various reproductive parameters

the ovary, is restricted to releasing gametes inside the body were considered independent of any organizational effect
cavity which then with some assistance hopefully find their during development. This notion was empirically tested by
way to the inside of the uterus. In order for those gametes Phoenix and his trainees and essentially rejected in what is
to merge and become zygotes, in other words be fertilized, now considered a land mark paper published in Hormones
there must be a mechanism by which external gametes also and Behavior in 1959 (Phoenix et al. 1959). The experi-
gain access to the interior of the uterus. Both scenarios ment was simple, they treated pregnant Guinea pigs with
require a series of coordinated behaviors by both sexes that testosterone and then assessed the copulatory behavior of
begin with mate seeking, mate recognition and selection fol- the offspring as adults. The critical component was the use
lowed by copulation. of different doses of testosterone, a high dose that fully mas-
culinized the genitalia of the females in utero so that they
were externally indistinguishable from male littermates,
Sexual differentiation of behavior begins and a lower dose that did not noticeably impact the geni-
with the brain talia. Offspring of both treatment groups were compared
to those from untreated dams. The animals were tested in
A central question in the early days of behavioral neuroen- two important ways. First was to determine the impact of
docrinology (not called that at the time), was whether the prenatal androgen exposure on the display of female sexual
behaviors associated with male versus female copulation receptivity (lordosis). They found that females exposed to
were a function of the peripheral apparatus, i.e. a penis ver- either high or low dose testosterone did not exhibit lordosis
sus a vagina, or controlled internally by the brain. Hard as it in response to mounting by a male, thus the behavior change
is to believe now, the prevailing and favored theory was that was independent of the peripheral genitalia. Secondly, they
the body was the determinant of behavior. Thus, if an indi- assessed the propensity of the neonatally exposed animals
vidual found themselves with a penis, regardless of any other to engage in male copulatory behavior, finding that females
variables, they would attempt to mount and copulate with exposed to high dose testosterone developmentally where
other individuals possessing a vagina, and vice versa. More- highly responsive to adult testosterone and vigorously
over, while hormones were clearly required for adult copula- mounted other females. Interestingly they did not test the
tory behavior to be displayed, the actions of the hormones low dose exposed females, presumably because they were

13
Journal of Comparative Physiology A

so inculcated with the prevailing view that without a penis hormone milieu is markedly different in reproductively
no animal would attempt to mate like a male. Nonetheless, mature animals, (2) developmental programming by steroids
they concluded that the actions of the prenatal hormones dictates adult responses or (3) every cell in the body, includ-
were organizational and enduring. They went on to assert ing the brain, is either XX or XY within a given individual
the notion that the tissues which had been organized devel- and genes on sex chromosomes could have direct effects. All
opmentally to modify behavior was probably neural, but they three or any combination of two of these is also possible.
were “not prepared to suggest whether the site of action is
generalized or localized”. They further speculated that any
effects of androgens on the neural substrate would likely Sex differences in the brain involve more
reflect subtle changes in function as opposed to structure. than just hormones: Sex chromosome
The intervening 60 years have proved that wrong and filled complement
in the gaps by convincingly demonstrating that hormones
have highly localized and varied impact on the developing Because of the hegemony of hormones, the notion that
brain, playing major roles in determining cell birth, death, genes on the sex chromosomes might contribute to male
differentiation and connectivity. The impact of steroids and female differences in brain and behavior was largely
expands beyond neurons, also profoundly influencing astro- ignored until the early 2000’s when Art Arnold made use
cytes, oligodendrocytes and cells of non-neuronal origin of a novel mouse strain in which gonadal sex and genetic
such as the immune microglia and mast cells. The impact of sex could be dissociated (De Vries et al. 2002). Known
steroids is so powerful, complex and widespread one could as the 4-core-genotype, this mouse strain is engineered
argue the field has been held prisoner by the all encompass- so that the Sry gene is removed from the Y chromosome
ing effort to understand this class of signaling molecules. and translocated to an autosome. This allows for animals
One could also say the field has been myopic in its view, that are XX-Sry + and XY-Sry-. In other words, it creates
attributing every and all sex difference in brain or behav- XX animals with testis and XY animals with ovaries. By
ior to the action of steroid hormones. Indeed, the power of comparing endpoints in these mice to those that are XX
hormones was revered as so great that the circulating levels with ovaries and XY with testis, the relative contribution of
observed in adult females across the estrus cycle was consid- chromosomal sex versus gonadal sex can be assessed. Many
ered so remarkable in its power to control brain and behav- behaviors have been examined and generally those directly
ior that females could not be used as reliable subjects in associated with reproduction, i.e. mating and parenting, are
experimental neuroscience. We will never know for certain, entirely controlled by the gonads (Arnold et al. 2004). But
but this may have contributed to the fact that within a few many other behaviors and some physiological endpoints are
short years the majority of neuroscience research narrowed at least “influenced” by chromosomal sex. One of the most
its focus to only male subjects (Beery and Zucker 2010). notable is body weight, which is markedly impacted by chro-
The study of females was only for purposes of understand- mosomal sex but only if the gonads are removed (Chen et al.
ing reproduction. 2013). In another particularly remarkable study involving an
Now we are in a new era. In response to a growing aware- animal model of multiple sclerosis (MS), the chromosomal
ness of the negative consequences of not considering sex sex of the immune system and the nervous system were
as a biological variable during preclinical research, the separated so that individuals were XX in the periphery and
NIH created a new mandate that requires all funded studies XY in the brain. This revealed that the higher vulnerability
include subjects of both sexes and that data be disaggregated of females to MS was a function of the peripheral immune
and analyzed for an impact of sex. If an NIH funded investi- system, whereas the more severe disease pathology observed
gator insists on using subjects of only one sex in preclinical in males is the result of XY in the nervous system (Du et al.
research it requires extraordinary justification, i.e. studies 2014). This reinforces the view that the relationship between
of ovarian cancer can only be done in females for instance. gonadal sex and chromosomal sex can be both contextual
Attention to the importance of sex as a biological variable and tissue dependent.
has extended to Canadian and European funding agencies What is missing in our understanding of potential impact
as well. Some consider this to be over reach and possibly of chromosome complement on sex differences is whether
damaging, particularly in the neurosciences, but others have there is any effect during development. An in vitro exami-
embraced the new mandate and are finding unexpected and nation of dopamine neuron survival in cells derived from
surprising differences in the characteristics of various bio- embryos of the 4-core-genotypes model demonstrates a
logical processes. Observations of this sort often lead natu- clear effect of sex chromosome complement (Carruth et al.
rally to questions of “how” does said biological sex differ- 2002), and is consistent with inferential findings in vivo
ence come about? There are three generic answers to this (Dewing et al. 2003), so there is potential for direct effects
question. Sex differences can occur because; (1) the steroid but whether they constitute sexual differentiation. per se,

13
Journal of Comparative Physiology A

is unclear. The sexual differentiation of the brain is clas- et al. 2011). MicroRNAs provide regulatory control of tran-
sically viewed as occurring during a critical period when scription and a single mRNA might impact multiple genes,
testicularly produced androgens in the male fetus gain access thereby modulating coordinated gene networks. Third, there
to the CNS and initiate a variety of molecular and cellu- are some genes that escape X-inactivation, resulting in a
lar processes that are the manifestation of masculinization. double dose in females and thereby creating the potential
The critical period begins with the production of steroids for a sex difference in gene expression (reviewed in (Arnold
late in gestation in rodents and about mid-gestation in pri- et al. 2016). In addition to being rich in microRNAs, the
mates, including humans, and ends within the first few days X-chromosome is also notably rich in genes relevant to brain
of postnatal life or prior to birth, respectively (reviewed in development and neural functioning, as well as immune sys-
(McCarthy et al. 2017a). In females there is a parallel sensi- tem genes (Bianchi et al. 2012). Lastly, while the Y chro-
tive period, which is distinct from a critical period in that it mosome is small, it is not completely devoid of genes and
involves sensitivity to an exogenous substance. In our rodent these too could have direct effects on the developing brain.
models, a newborn female can be injected with testosterone All of these are in the realm of possibility and none of them
or its aromatized byproduct estradiol, for up to 6–10 days exclude another, but to-date there has been no empirical evi-
postnatally and still be masculinized for some endpoints, dence that sex chromosome complement directly impacts
i.e. remain “sensitive”. This has provided an excellent tool sexual differentiation of the brain (Fig. 2).
for revealing the mechanisms of brain masculinization as
it is relatively easy to manipulate the hormonal milieu and
other physiological parameters in a newborn pup compared
to a developing fetus, which is further confounded by the Sex differences in the brain involve more
hormonal environment of pregnancy. However, in newborn than just hormones: transcription regulatory
males masculinization cannot be blocked or reversed easily networks (TRNs)
because the process was initiated in utero and like a train
that has left the station, it is difficult to impede its forward Steroid receptors are nuclear transcription factors and this
progression. In both cases, natural masculinization in males logically led to the assumption that androgen receptors
or induced masculinization in females, the brain is remark- (AR), estrogen receptors (ER) or both would induce a set
ably immature at the time that it is being “programmed” of genes that drive the brain masculinization process. This
or “organized”, especially in light of the fact that most of reasonable and plausible scenario leads to a series of test-
the physiology and behavior that will be impacted does not able predictions; (1) there will be sex differences in gene
appear until the animals are reproductively mature. The pul- expression, some genes more highly expressed in males and
satile versus cyclic release of luteinizing hormone from the some higher in females, (2) treatment of females with tes-
pituitary is a hallmark of puberty, and sexual behavior is not tosterone will recapitulate the pattern seen in males, (3) the
expressed until well after that, nor is territorial aggression, androgen-induced genes will be directly relevant to brain
courtship, nest building and maternal aggression. And so development, and (4) identifying those genes will solve the
there is tremendous dynamism occurring from the time of mystery of brain sexual differentiation. An analysis of the
sexual differentiation and sex differences in brain and behav- transcriptome of the neonatal preoptic area, arguably the
ior, but what is constant across this period is sex chromo- most sexually dimorphic region of the brain, found equal
some complement. This makes it difficult to identify effects numbers of genes expressed at higher levels in males versus
that might be restricted to a critical or sensitive period. females (Nugent et al. 2015), in direct contradiction to pre-
Further complicating our ability to attribute developmen- diction #1. Treating females with testosterone (or estradiol)
tal effects to sex chromosome complement is the increas- does increase the level of some mRNA and protein from
ing awareness that autosomes are impacted by whether some genes normally higher in males (Quadros et al. 2002),
they share the nucleus with an XX or XY set. There are but just as frequently does not. More importantly, the genes
several ways this can occur. First is that when a cell has that are regulated do not seem to have direct relationship
an XX compliment, one of the X chromosomes is subject to neural development, thus neither prediction #2 or #3 are
to inactivation. This inactivation is achieved by epigenetic strongly supported. One of the more surprising findings to
repression via DNA methylation and histone modification, emerge in recent years are the profound and pervasive sex
an energetically expensive process that could deplete the differences in the transcriptome, both developmentally and
cell of precious enzymes and substrates that may be needed in adulthood, in animals and humans (Nugent et al. 2015;
for other epigenetic regulatory processes, thereby creating Werling et al. 2016; Labonte et al. 2017). The differences
a heterochromatic sink. A second route for sex chromo- involve large number of genes, belying the notion that a few
some regulation of autosomes is via microRNAs, which select genes are responsible for the masculinization of the
are particularly abundant on the X chromosome (Pinheiro brain.

13
Journal of Comparative Physiology A

Fig. 2  Incorporation of sex chromosomes and transcriptomics in sex- aspects of sex chromosome complement to the process of sexual dif-
ual differentiation of the brain. In recent years it has become increas- ferentiation. Future studies addressing convergence of sex chromo-
ingly clear that sex chromosome complement in mammals (XX some effects and steroid hormones will be valuable in this regard. A
vs XY) is an important contributor to sex differences in brain and second emerging concept is sex differences in the transcriptome, and
behavior. However because of the continuous presence of this modi- in particular the potential for differences in networks as opposed to
fier since the time of conception, it is difficult to attribute particular isolated gene expression

So where do sex differences in the transcriptome come as a function of sex is found in differential gene expression,
from and what does it mean? Independent of, or, in tandem alternative splice variants and promoter usage, genome wide
with sex chromosome complement, there is a strong poten- methylation and histone modifications associated with pro-
tial for sex differences in transcription regulatory networks moter activation in rats, mice and humans (Trabzuni et al.
(TRNs). Transcription factors (TF) interact directly with 2013; Ghahramani et al. 2014; Nugent et al. 2015; Shen et al.
DNA or via co-activators and co-repressors in a multi-com- 2015; Werling et al. 2016; Labonte et al. 2017). Of particular
ponent transcriptional complex. They modulate the activity interest would be to know if TRNs are established devel-
at gene promoters and interact with distal regulatory ele- opmentally during the critical period and then maintained
ments known as enhancers. The binding of individual TF’s across the life span (Fig. 2).
can be weak and imprecise and so cooperativity between
them and associated proteins provides the robust regula-
tion needed to precisely regulate the roughly 20,000 genes Sex differences in the brain involve
in the mammalian genome. TRNs determine cell fate dur- more than just hormones: Immune
ing development and adult maintenance of homeostasis. and inflammatory mediators
How networks are established and maintained is still being
explored experimentally. Network stability is influenced by The cellular and molecular mechanisms mediating the mas-
both intrinsic (i.e. cell cycle progression) and extrinsic (i.e. culinization of specific brain regions has been elucidated in
metabolic state) factors (reviewed in (Wilkinson et al. 2017). considerable detail for some regions and a surprising pat-
Steroid receptors are classical TFs that recognize and inter- tern has emerged. It was predicted that factors impacting
act with palindromic sequences embedded in promoters but cell number, dendritic growth and synapse density would
also associate with a variety of co-factors, some specific most likely be growth factors, neurotransmitters and other
to steroid receptors and others more general, such as c-fos modulators of neuronal activity such as ion changes. Instead,
(Barrett 1998; Beato and Klug 2000). Steroids can also influ- an over preponderance of signaling pathways and cell types
ence TRNs by modulating the activity of DNA methylat- involved in immune and inflammatory responses was discov-
ing enzymes (Nugent et al. 2015), and histone modifying ered to be central to masculinization of multiple endpoints
enzymes (Matsuda et al. 2011). Evidence that TRNs differ in multiple brain regions (Fig. 3).

13
Journal of Comparative Physiology A

Fig. 3  New view of sexual differentiation of the brain involving in high levels of phagocytosis during the critical period for sexual
immune modulators. Converging evidence from studies exploring the differentiation, thereby modulating divers endpoints such as synap-
cellular and molecular mechanisms establishing sex differences in togenesis and cell survival. Cytokines and associated proteins are also
brain and behavior implicates the innate immune cells of the brain, expressed at higher levels in males and in one brain region result in
microglia, as well as mast cells which are found both in the periphery the death of neurons that would otherwise survive. Thus there both a
and centrally. Males consistently have either more of these immune complexity and a consistency in the involvement of the immune sys-
cells in specific brain regions or the cells are in a more activated tem in masculinization of brain and behavior in rodents
state and producing inflammatory signaling molecules are engaging

Sex differences in synaptic patterning are (Theoharides et al. 2012). In the brain mast cells are usu-
directed by prostaglandins and histamine ally found in the meninges but during the critical period for
sexual differentiation there is a population deep in the neu-
The preoptic area was noted above as touting some of the ropil of the preoptic area, and there are significantly more
most robust sex differences in the brain which come in the in males than females. At the close of the sensitive period
form of synaptic patterning and differential cell death. This the numbers drop precipitously in both sexes (Lenz et al.
brain region is also of interest due to its centrality to the 2018). During the time that mast cells are residing within
control of both mating and parenting behavior (Numan 1994; the neuropil, they are also constitutively degranulating in
Hull and Dominguez 2007). The neurons in this region are males and releasing histamine. Nearby microglia respond to
replete with estrogen and androgen receptors but the key the released histamine with increased prostaglandin produc-
players initiating the differentiation of the synaptic pattern tion, in particular PGE2, which is usually associated with
(i.e. more dense dendritic spine synapses in males), are two fever (Lazarus et al. 2007), but also has signaling capac-
types of immune cells, microglia and mast cells (Lenz et al. ity through G-protein coupled receptors linked to adenyl
2013; Lenz et al. 2018). Microglia are immune cells unique cyclase, cAMP production and activation of protein kinase A
to the brain and are modified macrophages that take up per- (PKA) (Sugimoto and Narumiya 2007). In the preoptic area
manent residence there early in embryonic development this activated PKA phosphorylates a subunit of the AMPA
(Ginhoux et al. 2013). Mast cells are also immune cells but glutamate receptor and this in turn promotes the traffick-
originate in the bone marrow and are distributed throughout ing of the receptor to the membrane and, via mechanisms
the body, particularly at surfaces such as skin, nasal epithe- not entirely understood, promotes the formation and stabi-
lium, mouth eyes etc. They are considered first respond- lization of excitatory dendritic spine synapses (Lenz et al.
ers to any tissue disturbances and are packed with large 2011). The increase in dendritic spines endures into adult-
secretory granules ready to dump histamine, serotonin and hood and positively correlates with the expression of male
other signaling molecules (Silver and Curley 2013). They sexual behavior (Wright et al. 2008). The preoptic area is
are also capable of making prostaglandins and cytokines essential for male sexual behavior and if it is damaged or

13
Journal of Comparative Physiology A

lesioned males lose interest in copulating. The increase in of GnRH neuronal activity to achieve these diverse ends
excitatory input created by generating a higher density of and this control is established during the critical period
glutamatergic synapses promotes mating by enhancing the for sexual differentiation. If a neonatal female is treated
neural responsiveness to salient cues from receptive females. with a large dose of testosterone or its aromatized end
The critical point is that two immune cells, microglia and product, estradiol, she will be sterile as an adult (Gorski
mast cells, are essential to the establishment of this node in and Barraclough 1963). The steroid-induced sterility is
the neural circuit controlling male copulation. In support of not because of any fault with the ovary or the pituitary
this conclusion, depleting microglia selectively during the but rather the loss of the synchronized firing of the GnRH
critical period completely abolishes the capacity for mating neurons required for a surge of LH from the pituitary to
in males when they mature, but has no impact on females induce ovulation. The follicules of the ovary develop per-
(VanRyzin et al. 2016). fectly well, they simply cannot release the ova, nor do they
form the corpus luteum required to maintain pregnancy.
The firing rate of GnRH neurons is not controlled by direct
Sex differences in cell death are directed action of estrogens because they do not express estrogen
by immune signaling receptors (Shivers et al. 1983). Instead it is the afferent
input to the GnRH neurons which is critical, and an essen-
In addition to a sex difference in synaptic patterning in the tial node for the synchronization required for the LH surge
preoptic area are volumetric differences in which a subnu- is the AVPV. The AVPV is a region of the preoptic area
cleus is larger in one sex. The most famous such nucleus is and contains a variety of different cell types. Dopamine
actually named for that fact, the sexually dimorphic nucleus neurons residing here are more numerous in females than
of the preoptic area, or more simply the SDN (Gorski et al. males (Simerly et al. 1997), but they are only a small per-
1980). The topic of intense study, the SDN is arguably the centage of the population (which is not to suggest they
most robust volumetric sex difference in the brain and its dis- are unimportant). Inhibitory GABAergic neurons in this
covery in the 1970’s heralded a new era of connecting brain region all express estrogen receptors, and some have a dual
morphology to physiology and behavior. It also provided the identity as glutamatergic as well, but regardless there are
first opportunity to identify the specific cellular mechanisms more of them in females and they project monosynapti-
by which steroids mediate sexual differentiation. Ironically, cally onto GnRH neurons (Ottem et al. 2004). The AVPV
attempts at both have largely failed. The precise function of is just one part of a complex intercalating network of col-
the SDN remains unclear although it is strongly implicated lections of cells that control GnRH activity but in a very
in partner preference (Roselli and Balthazart 2011). As to simplistic sense the more neurons in the AVPV the greater
mechanism, we know that the SDN starts with essentially the potential to induce the surge of LH necessary for ovu-
the same number of neurons in males and females but they lation. The number of neurons in the AVPV is established
die in females due to lack local of estradiol, aromatized from during the critical period for sexual differentiation, just as
peripheral androgens in males (Jacobson and Gorski 1981; with the SDN, but in this case estradiol appears to induce
Davis et al. 1996a). But HOW estradiol is providing survival cell death, thereby reducing the number of surviving cells
support to the SDN neurons in males has been a mystery, in males (Forger 2006).
and many obvious answers such as regulation of cell death That estradiol is acting as a cell killer instead of a
and survival pathways have been largely negative (Forger cell survival factor is surprising, but what is even more
2009). The possibility that immune cells such as microglia intriguing is that the process involves signaling pathways
and mast cells may be key mediators of SDN volume is an normally associated with inflammation (Krishnan et al.
active area of investigation. 2009). Tumor necrosis factor-alpha, or Tnfα, is a cytokine
A second nucleus within the preoptic area also differs involved in the acute phase of an inflammatory response,
in volume but in the opposite direction, being larger in and it acts through two related receptors, Tnfr1 and Tnfr2.
females than males (Davis et al. 1996b). The anteroventral Tnfr1 is directly associated with cell death, but it is Tnfr2
periventricular nucleus, or more simply the AVPV, is a key that is critical to cell survival in the AVPV of females.
node for the control of LH release from the pituitary by The impact of Tnfα is indirect via phosphorylation of
controlling the activity of GnRH neurons (Gu and Simerly NFκB, which increases the cell survival factor Bcl2. In
1997; Clarkson and Herbison 2006; Polston and Simerly males, estrogens induce yet another protein, Traip, which
2006). The pattern of LH release is sexually dimorphic, inhibits NFκB and allows for increased expression of the
being continuously pulsatile in males but cyclical in Bax mediated cell death pathway and thereby reducing the
females and characterized by a surge that is essential for overall cell population of the AVPV selectively in males
inducing ovulation. It requires markedly distinct control (reviewed in (Petersen et al. 2012).

13
Journal of Comparative Physiology A

Sex differences in cell death are directed genes. This does seem to be the case for the genes cod-
by immune cells ing for the COX-1 and COX-2 enzymes (Amateau and
McCarthy 2004), but there are no other clear demonstra-
The amygdala is a complex collection of subnuclei tions of direct steroid-hormone induced immune related
involved in a variety of social and emotional behaviors gene expression. On a more global level immune-related
ranging from juvenile social play, to mating to fear mem- genes appear to be epigenetically repressed in females
ories. Play behavior in rats is expressed predominantly as opposed to males, suggesting higher overall immune
during the period between weaning and puberty, and is related transcription in regions of male brain (McCarthy
often seen more frequently in males but depends upon con- et al. 2017b). This is puzzling given that androgens are
text (Bredewold et al. 2014; Argue and McCarthy 2015). generally anti-inflammatory, not pro-inflammatory. A
The medial amygdala has long been identified as a brain second source is the sex chromosome complement, with
region critical to this sex difference (Meaney and McE- the X chromosome being enriched in immune associated
wen 1986), but how that is the case was unknown until genes (Bianchi et al. 2012). As discussed above, there are
microglia were considered as potential sculptures of the many genes that escape X-inactivation, resulting in a dou-
neural circuit of play. In developing males, the microglia ble dose in females, which would be counter to the notion
of the amygdala are highly phagocytic and they selectively of higher immune activation in males. Lastly there is the
target newly born astrocytes (VanRyzin et al. 2019). The potential that the source is external to the individual and
ability of microglia to engulf and kill new born cells in instead derives from the maternal immune system. During
a directed manner is a recently discovered phenomenon pregnancy the maternal immune system must be repressed
referred to as “phagoptosis”, and about which we still have in order for successful growth of the fetus. However, there
much to learn (Brown and Neher 2014). In the develop- is increasing evidence that repression against male fetuses
ing amygdala, newly born astrocytes express proteins on is less strong than that against female fetuses (reviewed
their surface that are interpreted by microglia as “eat me” in (McCarthy 2019). Flipping that around, the maternal
signals, as opposed to “don’t eat me” signals found on immune system is more apt to recognize a male fetus as
other cells. In males, the number of astrocytes express- foreign and mount a reaction against it. This could result in
ing eat me signals is higher, and they are therefore more increased levels of diffusible inflammatory signals, such as
frequently consumed. The end result is a reduced popula- prostaglandins or cytokines, in the male fetuses, or could
tion of astrocytes in the male medial amygdala compared evoke a reactive immune response from the male fetus
to females, and this then correlates with greater neuronal itself. Lastly, there is the potential for maternal immune
activation during play bouts, which occur some 3–4 weeks cells gaining access to the male fetus and even invading
later (VanRyzin et al. 2019). Thus, once again, the immune the brain. Demonstrating any or all of these will be tech-
system is the key determinant of neural circuit construc- nically challenging for many reasons. One of the biggest
tion that will impact sex differences in physiology and is separating what is maternal in origin from what is fetal
behavior as the animal matures. in origin. Equally daunting is the accurate identification
and quantification of immune cells in the nervous system
as these cells are both small and rare. Overcoming these
hurdles will provide essential information for determining
A new view of sexual differentiation the veracity of this new view of sexual differentiation of
of the brain the brain.

Based on the above observations, we propose the novel


working hypothesis that neuroinflammatory mediators and Funding This work was supported by RO1MH52716 and
R01DA039062 to MMM.
immune cells are primary drivers of the masculinization
of specific brain regions and behaviors, while steroids
are either enablers that act to augment and enhance the
inflammatory signaling, or perhaps are actually acting in
References
an anti-inflammatory capacity in order to prevent runa-
way inflammation. But this raises the essential question of Amateau SK, McCarthy MM (2004) Induction of PGE(2) by estradiol
why do males have more of these signaling molecules and mediates developmental masculinization of sex behavior. Nat
immune cells than females? There are multiple potential Neurosci 7:643–650
sources, beginning with a direct effect of testosterone or Argue KJ, McCarthy MM (2015) Characterization of juvenile play
in rats: importance of sex of self and sex of partner. Biol Sex
estradiol on the transcription of inflammation associated Differ 6:16

13
Journal of Comparative Physiology A

Arnold AP, Xu J, Grisham W, Chen X, Kim YH, Itoh Y (2004) Mini- Gorski RA, Harlan RE, Jacobson CD, Shryne JE, Southam AM (1980)
review: Sex chromosomes and brain sexual differentiation. Endo- Evidence for the existence of a sexually dimorphic nucleus in the
crinology 145:1057–1062 preoptic area of the rat. J Comp Neurol 193:529–539
Arnold AP, Reue K, Eghbali M, Vilain E, Chen X, Ghahramani N, Gu GB, Simerly RB (1997) Projections of the sexually dimorphic
Itoh Y, Li J, Link JC, Ngun T, Williams-Burris SM (2016) The anteroventral periventricular nucleus in the female rat. J Comp
importance of having two X chromosomes. Philos Trans R Soc Neurol 384:142–164
Lond Ser B Biol Sci 371:20150113 Hull EM, Dominguez JM (2007) Sexual behavior in male rodents.
Barrett TJaS, Thomas C (1998) Steroid receptors at the nexus of tran- Hormones Behav 52:45–55
scriptional regulation. J Cell Biochem Suppl 30:198–193 Jacobson CD, Gorski RA (1981) Neurogenesis of the sexually dimor-
Beato M, Klug J (2000) Steroid hormone receptors: an update. Hum phic nucleus of the preoptic area in the rat. J Comp Neurol
Reprod Update 6:225–236 196:519–529
Beery AK, Zucker I (2010) Sex bias in neuroscience and biomedical Krishnan S, Intlekofer KA, Aggison LK, Petersen SL (2009) Central
research. Neurosci Biobehav Rev 35:565–572 role of TRAF-interacting protein in a new model of brain sexual
Bianchi I, Lleo A, Gershwin ME, Invernizzi P (2012) The X chromo- differentiation. Proc Natl Acad Sci USA 106:16692–16697
some and immune associated genes. J Autoimmun 38:J187–J192 Labonte B et al (2017) Sex-specific transcriptional signatures in human
Bredewold R, Smith CJ, Dumais KM, Veenema AH (2014) Sex-spe- depression. Nat Med
cific modulation of juvenile social play behavior by vasopressin Lazarus M, Yoshida K, Coppari R, Bass CE, Mochizuki T, Lowell
and oxytocin depends on social context. Front Behav Neurosci BB, Saper CB (2007) EP3 prostaglandin receptors in the median
8:216 preoptic nucleus are critical for fever responses. Nat Neurosci
Brown GC, Neher JJ (2014) Microglial phagocytosis of live neurons. 10:1131–1133
Nat Rev Neurosci 15:209–216 Lenz KM, Wright CL, Martin RC, McCarthy MM (2011) Prostaglandin
Carruth LL, Reisert I, Arnold AP (2002) Sex chromosome genes E regulates AMPA receptor phosphorylation and promotes mem-
directly affect brain sexual differentiation. Nat Neurosci brane insertion in preoptic area neurons and glia during sexual
5:933–934 differentiation. PloS One 6:e18500
Chen X, McClusky R, Itoh Y, Reue K, Arnold AP (2013) X and Y Lenz KM, Nugent BM, Haliyur R, McCarthy MM (2013) Microglia
chromosome complement influence adiposity and metabolism in are essential to masculinization of brain and behavior. J Neurosci
mice. Endocrinology 154:1092–1104 33:2761–2772
Clarkson J, Herbison AE (2006) Postnatal development of kisspeptin Lenz KM, Pickett LA, Wright CL, Davis KT, Joshi A, McCarthy MM
neurons in mouse hypothalamus; sexual dimorphism and projec- (2018) Mast Cells in the Developing Brain Determine Adult Sex-
tions to gonadotropin-releasing hormone neurons. Endocrinology ual Behavior. J Neurosci 38:8044–8059
147:5817–5825 Matsuda KI, Mori H, Nugent BM, Pfaff DW, McCarthy MM, Kawata
Davis EC, Popper P, Gorski RA (1996a) The role of apoptosis in sexual M (2011) Histone deacetylation during brain development is
differentiation of the rat sexually dimorphic nucleus of the preop- essential for permanent masculinization of sexual behavior. Endo-
tic area. Brain Res 734:10–18 crinology 152:2760–2767
Davis EC, Shryne JE, Gorski RA (1996b) Structural sexual dimor- McCarthy MM (2019) Sex differences in neuroimmunity as an inherent
phisms in the anteroventral periventricular nucleus of the rat hypo- risk factor. Neuropsychopharmacology 44:38–44
thalamus are sensitive to gonadal steroids perinatally, but develop McCarthy MM, De Vries GJ, Forger NG (2017) Sexual differentiation
peripubertally. Neuroendocrinology 63:142–148 of the brain: a Fresh look at mode, mechanisms and meaning.
De Vries GJ, Rissman EF, Simerly RB, Yang LY, Scordalakes EM, Hormones, brain and behavior. Pfaff DaJ, San Diego, pp 3–32
Auger CJ, Swain A, Lovell-Badge R, Burgoyne PS, Arnold AP McCarthy MM, Nugent BM, Lenz KM (2017) Neuroimmunology and
(2002) A model system for study of sex chromosome effects neuroepigenetics in the establishment of sex differences in the
on sexually dimorphic neural and behavioral traits. J Neurosci brain. Nat Rev Neurosci 18:471–484
22:9005–9014 Meaney MJ, McEwen BS (1986) Testosterone implants into the amyg-
Dewing P, Shi T, Horvath S, Vilain E (2003) Sexually dimorphic gene dala during the neonatal period masculinize the social play of
expression in mouse brain precedes gonadal differentiation. Brain juvenile female rats. Brain Res 398:324–328
Res Mol Brain Res 118:82–90 Nugent BM, Wright CL, Shetty AC, Hodes GE, Lenz KM, Mahurkar
Du S, Itoh N, Askarinam S, Hill H, Arnold AP, Voskuhl RR (2014) A, Russo SJ, Devine SE, McCarthy MM (2015) Brain feminiza-
XY sex chromosome complement, compared with XX, in the CNS tion requires active repression of masculinization via DNA meth-
confers greater neurodegeneration during experimental autoim- ylation. Nat Neurosci 18:690–697
mune encephalomyelitis. Proc Natl Acad Sci USA 111:2806–2811 Numan M (1994) Maternal behavior. In: Knobil E, Neill JD (eds)
Forger NG (2006) Cell death and sexual differentiation of the nervous Physiology of reproduction. Raven Press, New York, pp 108–302
system. Neuroscience 138:929–938 Ottem EN, Godwon JG, Krishnan S, Petersen SL (2004) Dual-phe-
Forger NG (2009) Control of cell number in the sexually dimorphic notype GABA/glutamate neurons in adult preoptic area: Sexual
brain and spinal cord. J Neuroendocrinol 21:393–399 dimorphism and function. J Neurosci 24:8097–8105
Ghahramani NM, Ngun TC, Chen PY, Tian Y, Krishnan S, Muir S, Petersen SL, Krishnan S, Aggison LK, Intlekofer KA, Moura PJ (2012)
Rubbi L, Arnold AP, de Vries GJ, Forger NG, Pellegrini M, Vilain Sexual differentiation of the gonadotropin surge release mecha-
E (2014) The effects of perinatal testosterone exposure on the nism: a new role for the canonical NfkappaB signaling pathway.
DNA methylome of the mouse brain are late-emerging. Biol Sex Front Neuroendocr 33:36–44
Differ 5:8 Phoenix CH, Goy RW, Gerall AA, Young WC (1959) Organizing
Ginhoux F, Lim S, Hoeffel G, Low D, Huber T (2013) Origin and dif- action of prenatally administered testosterone proprionate on the
ferentiation of microglia. Front Cell Neurosci 7:45 tissues mediating mating behavior in the female guinea pig. Endo-
Goodfellow PN, Lovell-Badge R (1993) SRY and sex determination in crinology 65:369–382
mammals. Annu Rev Genet 27:71–92 Pinheiro I, Dejager L, Libert C (2011) X-chromosome-located micro-
Gorski RA, Barraclough CA (1963) Effects of low dosages of androgen RNAs in immunity: might they explain male/female differences?
on the differentiation of hypothalamic regulatory control of ovula- The X chromosome-genomic context may affect X-located
tion in the rat. Endorcrinology 73:210–216 miRNAs and downstream signaling, thereby contributing to the

13
Journal of Comparative Physiology A

enhanced immune response of females. BioEssays News Rev Mol Kalogeromitros D (2012) Mast cells and inflammation. Biochim
Cell Dev Biol 33:791–802 Biophys Acta 1822:21–33
Polston EK, Simerly RB (2006) Ontogeny of the projections from the Trabzuni D, Ramasamy A, Imran S, Walker R, Smith C, Weale ME,
anteroventral periventricular nucleus of the hypothalamus in the Hardy J, Ryten M (2013) Widespread sex differences in gene
female rat. J Comp Neurol 495:122–132 expression and splicing in the adult human brain. Nat Commun
Quadros PS, Goldstein AY, De Vries GJ, Wagner CK (2002) Regu- 4:2771North American Brain Expression C
lation of sex differences in progesterone receptor expression in VanRyzin JW, Yu SJ, Perez-Pouchoulen M, McCarthy MM (2016)
the medial preoptic nucleus of postnatal rats. J Neuroendocrinol Temporary depletion of microglia during the early postnatal
14:761–767 period induces lasting sex-dependent and sex-independent effects
Roselli C, Balthazart J (2011) Sexual differentiation of sexual behavior on behavior in rats. eNeuro 3
and its orientation. Front Neuroendocr 32:109 VanRyzin JW, Marquardt AE, Argue KJ, Vecchiarelli HA, Ashton
Shen EY, Ahern TH, Cheung I, Straubhaar J, Dincer A, Houston I, SE, Arambula SE, Hill MN, McCarthy MM (2019) Microglial
de Vries GJ, Akbarian S, Forger NG (2015) Epigenetics and sex phagocytosis of newborn cells is induced by endocannabinoids
differences in the brain: a genome-wide comparison of histone-3 and sculpts sex differences in Juvenile Rat Social Play. Neuron
lysine-4 trimethylation (H3K4me3) in male and female mice. Exp Werling DM, Parikshak NN, Geschwind DH (2016) Gene expression
Neurol 268:21–29 in human brain implicates sexually dimorphic pathways in autism
Shivers BD, Harlan RE, Morrell JI, Pfaff DW (1983) Absence of spectrum disorders. Nat Commun 7:10717
oestradiol concentration in cell nuclei of LHRH-immunoreactive Wilkinson AC, Nakauchi H, Gottgens B (2017) Mammalian transcrip-
neurones. Nature 304:345–347 tion factor networks: recent advances in interrogating biological
Silver R, Curley JP (2013) Mast cells on the mind: new insights and complexity. Cell Syst 5:319–331
opportunities. Trends Neurosci 36:513–521 Wright CL, Burks SR, McCarthy MM (2008) Identification of prosta-
Simerly RB, Zee MC, Pendleton JW, Lubahn DB, Korach KS (1997) glandin E2 receptors mediating perinatal masculinization of adult
Estrogen receptor-dependent sexual differentiation of dopaminer- sex behavior and neuroanatomical correlates. Dev Neurobiol 68
gic neurons in the preoptic region of the mouse. Proc Natl Acad
Sci USA 94:14077–14082 Publisher’s Note Springer Nature remains neutral with regard
Sugimoto Y, Narumiya S (2007) Prostaglandin E receptors. J Biol tojurisdictional claims in published maps and institutional affiliations.
Chem 282:11613–11617
Theoharides TC, Alysandratos KD, Angelidou A, Delivanis DA, Sis-
manopoulos N, Zhang B, Asadi S, Vasiadi M, Weng Z, Miniati A,

13

You might also like