Вы находитесь на странице: 1из 22

Frontiers in Neuroendocrinology 35 (2014) 89110

Contents lists available at ScienceDirect

Frontiers in Neuroendocrinology
journal homepage: www.elsevier.com/locate/yfrne

Review

Puberty and adolescence as a time of vulnerability to stressors that alter


neurobehavioral processes
Mary K. Holder 1, Jeffrey D. Blaustein ,1
Neuroscience and Behavior Program, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA
Center for Neuroendocrine Studies, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA

a r t i c l e i n f o a b s t r a c t

Article history: Puberty and adolescence are major life transitions during which an individuals physiology and behavior
Available online 1 November 2013 changes from that of a juvenile to that of an adult. Here we review studies documenting the effects of
stressors during pubertal and adolescent development on the adult brain and behavior. The experience
Keywords: of complex or compound stressors during puberty/adolescence generally increases stress reactivity,
Stress increases anxiety and depression, and decreases cognitive performance in adulthood. These behavioral
Immune challenge changes correlate with decreased hippocampal volumes and alterations in neural plasticity. Moreover,
Depression
stressful experiences during puberty disrupt behavioral responses to gonadal hormones both in sexual
Anxiety
Cognitive function
performance and on cognition and emotionality. These behavioral changes correlate with altered estro-
Sexual behavior gen receptor densities in some estrogen-concentrating brain areas, suggesting a remodeling of the brains
Estradiol response to hormones. A hypothesis is presented that activation of the immune system results in chronic
Progesterone neuroinammation that may mediate the alterations of hormone-modulated behaviors in adulthood.
Stress reactivity 2013 Elsevier Inc. All rights reserved.

1. Introduction (Tanner Stage III) predicts this sex difference better than age alone
(Hayward and Sanborn, 2002; Patton et al., 1996), suggesting that
Puberty is a major life transition from that of a non-reproduc- ovarian hormones, particularly in puberty/adolescence, play a role
tive juvenile into a reproductively competent adult. Puberty and in the etiology of affective disorders.
adolescence, the developmental period associated with puberty, Recently, studies have demonstrated that peri-pubertal or ado-
are times of great physiological, psychosocial and cultural changes. lescent exposure to stressful life experiences contributes greatly to
As such, puberty and adolescence are also times of great develop- an individuals vulnerability to mental disease (Ge et al., 2001;
mental plasticity (Dahl and Gunnar, 2009; Patton and Viner, 2007). Grant et al., 2003, 2004; Silverman et al., 1996; Turner and Lloyd,
The neuroplasticity of puberty may also contribute to vulnerability 2004). Yet little is known about how stress could disrupt normal
for the development of mental diseases (Andersen, 2003). The Na- neural development during the pubertal or adolescent period to
tional Comorbidity Survey Replication study, which assessed over produce a brain that is particularly vulnerable to mental diseases.
9000 people representative of the US population, indicate that Therefore, it is important to understand the neural mechanisms by
affective disorders such as anxiety, bipolar disorder, and major which a pubertal stressor may produce a brain that is susceptible
depression emerge during adolescence, with the peak age of onset to specic mental diseases. Using animal models, we are beginning
for these diseases of 14 years (Kessler et al., 2005; Paus et al., to learn the unique ways in which a pubertal organism responds to
2008). A major predictor of an individuals susceptibility to mental a stressor, and how the experience of stressors during pubertal
disease is his or her sex: women show higher rates of anxiety and development alters adult physiology and behavior.
depression, and men have higher rates of autism spectrum disor- The focus of this review is to present results of recent studies
ders, schizophrenia, and attention decit hyperactivity disorder that demonstrate (i) that stressors experienced in puberty alter
(Astbury, 2001; Eaton et al., 2012). However, the 2:1 femalemale steroid hormone-inuenced behaviors in adulthood and (ii) that
prevalence in depression and anxiety disorders emerges only after these behavioral changes may be mediated through alterations in
puberty (Angold and Costello, 2006). Moreover, the pubertal status the ongoing processes of brain development. First, we provide a
short background on some of the major developmental processes
Corresponding author. that occur during puberty and adolescence. Next, we discuss the
E-mail addresses: mkholder@cns.umass.edu (M.K. Holder), blaustein@cns. ability of stressors during pubertal development to alter the dis-
umass.edu (J.D. Blaustein). play of sexual behaviors, anxiety- and depression-like behaviors,
1
Address: 135 Hicks Way, Tobin Hall, Amherst, MA 01003-9271, USA. Fax: +1 413 and learning, memory and cognitive behaviors, giving special
545 0996.

0091-3022/$ - see front matter 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.yfrne.2013.10.004
90 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

attention to the possible neural correlates and mechanisms of hormonal changes of puberty (Sisk and Foster, 2004). While the
these behavioral responses. Finally, we postulate a novel mecha- biological hallmarks of pubertal development have been exten-
nism by which stressors may act to achieve enduring effects in sively categorized as the Tanner Stages, the biological hallmarks
the adult brain and behavior. of adolescence are more nebulous and include the neuroplasticity
and development of the cortical and limbic areas of the brain.
Pubertal development in humans comprises several physiolog-
2. Puberty and adolescence ical and physical changes (Fig. 1), including the development of
secondary sexual characteristics (e.g., breast budding in girls and
2.1. Denitions of puberty and adolescence testicular enlargement in boys and the growth of pubic hair) and
culminating in the onset of reproductive competence (e.g., menar-
Puberty and adolescence are commonly conated; however, in che and ovulatory menstrual cycles in girls and semenarche and
order to understand the ways that puberty and adolescence can spermarche in boys) (Marshall and Tanner, 1969, 1970; Tanner,
inuence the brains response to hormones, it is critical to disam- 1962). The pubertal processes are dependent on the presence of
biguate and use precise denitions of both puberty and adoles- estrogens and progestins in girls and androgens in boys (Tanner,
cence, as they are distinct processes. Puberty refers to the 1962). Typically girls begin pubertal development around ages
developmental transition from a non-reproductive state into a 1011 and have completed the process by ages 1516 (Marshall
reproductive state, culminating in reproductive competence (Sisk and Tanner, 1969; Tanner, 1962), whereas boys begin puberty
and Foster, 2004; Waylen and Wolke, 2004). Even though it is slightly later at ages 1112 and have completed it by 1617 (Mar-
sometimes referred to as a singular event, puberty is a prolonged, shall and Tanner, 1969; Tanner, 1962).
developmental process. The term is not only used to indicate the In female rodents, the rst stage of pubertal development is
attainment of reproductive competence, but also it is used to de- typically the rst external sign of ovarian activity (i.e., vaginal
scribe the reproductive changes culminating in reproductive com- opening), and puberty is considered complete with the onset of
petence. Throughout this review, we will refer to the changes that the rst reproductive (i.e., estrous) cycle. Although variable and
occur in reproductive status as pubertal developments and the dependent on housing conditions (e.g., Vandenbergh, 1967,
time during which these pubertal developments occur as the 1969), the day of vaginal opening is approximately postnatal day
pubertal period. In contrast, adolescence refers to the social and 35 (P35) in rats (Ojeda and Urbanski, 1994; Ojeda et al., 1976)
cognitive maturation associated with and resulting from the and P25 in C57Bl/6 mice and P30 in CD1 mice housed in single

Fig. 1. A generalization of the approximate timing and events of pubertal development and adolescence in (A) humans and (B) rats. (A) The average ages of the pubertal
developments for girls (top) and boys (bottom) (Marshall and Tanner, 1969, 1970; Sizonenko, 1989; Tanner, 1962) and for (B) females (top) and males (bottom) (Korenbrot
et al., 1977; Lohmiller and Sonya, 2006; Ojeda and Urbanski, 1994; Ojeda et al., 1976; Parker and Mahesh, 1976; Vandenbergh, 1967, 1969). Puberty onset is dened by the
appearance of the secondary sex characteristics, and the offset is dened by the emergence of reproductive competence as indicated by ovulatory menstrual cycles and
mature, motile sperm. In humans, the ages ranging from 12 to 18 are commonly used as the age range of adolescence (Spear, 2000); whereas, in rats adolescence is
conceptualized by early, mid, and late adolescent periods (Tirelli et al., 2003). It should be noted that the times indicated for particular stages are quite variable, and can be
inuenced by nutrition and environmental conditions.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 91

sex rooms (Ismail & Blaustein, unpublished observations). Female parietal lobes rst increases in early adolescence and then de-
rats can have quite a short latency between vaginal opening and creases in later adolescence (Shaw et al., 2008). The age-related de-
estrous cycles. This latency can be as prolonged as a week crease in grey matter volume has been proposed to be due to the
(Lohmiller and Sonya, 2006), but can be as brief as the same day increase in myelination and axonal growth, resulting in a net in-
(Ojeda et al., 1976; Parker and Mahesh, 1976). In female mice, crease in white matter and subsequent decrease in grey matter
however, the pubertal period may last several weeks when females (Paus et al., 2008). Alternatively, this decrease in grey matter
are housed in the absence of male mice. While the presence of a may reect the synaptic reorganization in the form of proliferation
male or male odor can both advance vaginal opening and shorten of neurons and/or synapses, followed by subsequent cell death and
the interval between vaginal opening and rst estrus (i.e., synapse elimination (Giedd et al., 1999; Huttenlocher, 1979; Peta-
710 days instead of 20 days), this pubertal period is still consid- njek et al., 2011; Sowell et al., 2001). Interestingly, sex differences
ered extended, compared to rats (Vandenbergh, 1967, 1969). It is in region volume or neural density emerge as a consequence of the
important to note that girls also have an extended pubertal period brain remodeling. The hippocampal volume increases in girls,
of 23 years, if one considers the timing of analogous events whereas the volume of the amygdala (Giedd et al., 1997) increases
beginning with the development of pubic hair and breast budding in boys. Additionally, the bed nucleus of the stria terminalis is lar-
(Tanner Stage II) and ending with menarche and regular ovulatory ger and with a greater number of neurons in boys (Chung et al.,
menstrual cycles (Sizonenko, 1989). 2002).
The external markers of pubertal development in male rodents Rodents also experience neural development and brain remod-
are the separation of the prepuce from the glans penis, or preputial eling during the pubertal period. These changes take the form of
separation, and the presence of mature motile sperm in the caput gross morphological alterations, such as volumetric changes in
epididymis (Korenbrot et al., 1977). In rats, these events can be grey matter and neuronal cell numbers. The remodeling of the
separated by a few days to a week (Korenbrot et al., 1977), but pubertal brain in rodents also includes reorganization at the
the average day of preputial separation is P42. Although preputial pre- and post-synaptic levels (reviewed in Sisk and Zehr, 2005).
separation is also used as a marker of pubertal development in Important for this review is the fact that many, but not all, of the
male mice, the timing of separation and the presence of mature processes that result in the remodeling of the pubertal and adoles-
sperm remain to be determined. cent brain are driven by the pubertal hormones (Andersen et al.,
While animal work may inform the mechanism by which the 2002; Davis et al., 1996; Goldstein et al., 1990; Guillamon et al.,
pubertal brain is particularly vulnerable to insult, it can be difcult 1988; Nunez et al., 2000). Therefore, pubertal status, rather than
to reconcile animal work and the human condition. In particular, chronological age, is likely to be the critical factor. Unfortunately,
work in humans often does not precisely dene pubertal stage few studies focus on the precise, clearly dened, stage of reproduc-
but rather focuses on age or events that occur during adolescence. tive development, so it can be difcult to determine the effects of
Additionally, studies using rodents typically use chronological age, the stressors during this time frame on subsequent behavioral
rather than pubertal status, to categorize the animals using a clas- and neurobiological processes in adulthood.
sication system of early adolescence (P21P34), mid-adolescence
(P3546), and late adolescence (P4759) (Tirelli et al., 2003). An- 2.3. Puberty as a second period of organizational actions of steroid
other widely used convention for classications within the rat lit- hormones
erature denes the adolescent period as P2842 (Spear, 2000).
Therefore, the adolescent and pubertal periods of males are not Multiple sensitive periods exist for the organization of the brain
orthogonal; rather they are overlapping, resulting in ambiguity in and nervous system, primarily occurring during periods of rapid
the results of many experiments (Fig. 1). That is, it can be difcult developmental changes (Scott et al., 1974). One of the most well-
to determine whether the particular effects are due to manipula- studied and best-characterized periods is that of the perinatal per-
tion at a particular chronological age or to a particular reproductive iod in rodents. During the perinatal period, the steroid hormones,
stage. Therefore, in this review, we will focus on the pubertal stage testosterone and estradiol, have the ability to organize the neural
of development when possible. circuits and permanently alter the brain and behavior. That is, even
a brief exposure to testosterone or estradiol can sexually differen-
2.2. Puberty as time of neuroplasticity and neural development tiate the brain (Lenz et al., 2012; McCarthy, 2010; Phoenix et al.,
1959; Wallen, 2005). Organizational effects are contrasted with
The pubertal and adolescent periods are times of great neuro- the activational effects of hormones, which are transient modica-
plasticity and neural development in both humans and in rodent tions or modulation of activity that can alter physiology and
animal model species. The pubertal period shares many develop- behavior (Phoenix et al., 1959). Additionally, the organizational
mental processes with the prenatal and early postnatal periods: actions of hormones are also limited to critical or vulnerable win-
(I) neuronal growth (Phoenix et al., 1959; Pinos et al., 2001) or dows. Prior to and after these windows of plasticity, hormones may
death (Nunez et al., 2001); (II) growth of axonal projections or no longer affect the brains development (Phoenix et al., 1959). The
axonal sprouting (Benes et al., 2000; Dahl, 2004; Lewis et al., pubertal period is now understood to be one such period of orga-
2004); (III) reshaping of the dendritic tree by dendritic elaboration nization by steroid hormones (Sisk and Zehr, 2005).
or retraction (Goldstein et al., 1990; Meyer et al., 1978); (IV) syn- Implicit in the discussion of the remodeling of the pubertal and
apse formation (Bourgeois et al., 1994; Bourgeois and Rakic, adolescent brain is the conceptualization of the pubertal period as
1993) or elimination (Andersen et al., 2002; Gerocs et al., 1986; a second phase of steroid-dependent organization of behaviors and
Huttenlocher and Dabholkar, 1997; Meyer et al., 1978); and (V) their underlying neural circuitry. Therefore, an individuals life
myelination (Benes et al., 1994; Nunez et al., 2000; Woo et al., experience during pubertal development and adolescence may
1997). have enduring consequences, which profoundly affect adult life.
Some brain regions, such as the prefrontal cortex, continue to Furthermore, as many of the neurodevelopmental processes occur-
develop into adulthood in humans. In particular, gross morpholog- ring during the rather nebulous period of adolescence are hormon-
ical changes result from both increases in white matter by contin- ally-regulated or modulated, it may be that pubertal status and the
ued axonal myelination and changes in grey matter (Barnea-Goraly underlying hormonal milieu are the critical factors in an individ-
et al., 2005; Giedd et al., 1999; Paus et al., 1999; Reiss et al., 1996; uals susceptibility to the enduring consequences of experience. In-
Sowell et al., 2001). The volume of grey matter in the frontal and deed, stress during pubertal development and/or adolescence
92 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Fig. 2. Representation of the hypothalamuspituitarygonadal axis. Neurons in the preoptic area and mediobasal hypothalamus secrete gonadotropin-releasing hormone
(GnRH), a neurohormone, into the hypophyseal portal system of the median eminence. The portal blood carries GnRH into the pituitary gland and activates its receptor,
gonadotropin-releasing hormone receptor (GnRHR) located on gonadotrope cells. Once activated, the GnRHR triggers synthesis and secretion of pituitary gonadotropins:
follicle stimulating hormone (FSH) and luteinizing hormone (LH). LH and FSH then act in concert to stimulate the production of gonadal steroid hormones (e.g., androgens
from the testes and estrogens and progestins from the ovary) and maturation of gametes (e.g., the spermatozoa in the testes and an ovum in the ovary). The gonadal
hormones act in the hypothalamus and pituitary to regulate gonadotropin secretion via feedback loops. Solid lines, positive feedback; dashed lines, negative feedback.

results in profound alterations in physiology and behavior as indi- the brain: estrogen receptor (ER) a and b, also referred to as
cated by (I) increases in anxiety and depression (Ge et al., 2001; ESR1 and ESR2, respectively. High levels of ERa are present in
Patton and Viner, 2007); (II) risk-taking and novelty-seeking hypothalamic and limbic nuclei in the rat brain, including but
behaviors (Arnett, 1996; Roberti, 2004; Wagner, 2001); (III) altera- not limited to the anteroventral periventricular nucleus (AVPV),
tions in learning and cognition (Im-Bolter et al., 2013); and (IV) medial preoptic area (mPOA), bed nucleus of the stria terminalis
drug and alcohol use and abuse (Bekman et al., 2010; Crawford (BNST), arcuate nucleus, paraventricular nucleus of the hypothala-
et al., 2003; MacPherson et al., 2010). In this review, we will con- mus (PVN), ventromedial nucleus of the hypothalamus (VMN), and
ne ourselves to the ability of stressors in the pubertal and adoles- medial amygdala (MeA) (Shughrue et al., 1996, 1997). The distribu-
cent periods to alter the brains response to gonadal (Section 3) and tion of ERb is similar to ERa, but it is more highly expressed in the
adrenal (Section 4) hormones. mPOA, hippocampus, and cerebellum (Shughrue et al., 1996, 1997).
While no qualitative sex differences in the distribution of ERs in
3. Gonadal steroid hormones particular brain regions have been reported, there are sex differ-
ences in the density of ERs (Rainbow et al., 1982). Compared to
3.1. Hypothalamicpituitarygonadal (HPG) axis male rats, female rats have higher concentrations of ERs in the
BNST peri-pubertally (Kuhnemann et al., 1994) and in the mPOA,
The hypothalamus and preoptic areas of the brain control much AVPV, and VMN pubertally and into adulthood (Brown et al.,
of the neuroendocrine activity of vertebrates via regulation of hor- 1988; Kuhnemann et al., 1994). Specically, female rats have a
mone secretion. The secretion of the gonadal hormones, primarily higher density of ERa in the VMN during pubertal development
estrogens and progestins in females and androgens in males, is (Yokosuka et al., 1997) and in adulthood (Yamada et al., 2009)
controlled by the HPG axis (Fig. 2). The primary sources of gonadal and more ERb in the lateral amygdaloid nucleus and the dentate
hormones are the ovaries in females and the testes in males. The gyrus and CA3 region of the hippocampus in adulthood (Zhang
ovary primarily produces two classes of steroid hormones from et al., 2002).
cholesterol: estrogens and progestins. Of the estrogens, the most Progesterone acts on two main receptors in the brain: PRA and
behaviorly potent is estradiol (Feder and Silver, 1974) and will be PRB. The PRs are expressed throughout the female and male rat
discussed in this review. Progestins, or progestogens, are the sec- brains, especially in the BNST, mPOA, AVPV, PVN, VMN and arcuate
ond class of steroid hormones synthesized in the ovary. Progester- nucleus (Parsons et al., 1982; Rainbow et al., 1982). Treatment
one is the most biologically potent of the progestins. The testes with estradiol also induces the transcription of the genes for PRs
produce androgens, of which testosterone is the primary androgen within these brain regions (Intlekofer and Petersen, 2010; Parsons
discussed in this review. et al., 1982). Adult female rats have a higher density of PRs within
the VMN and AVPV, compared to adult male rats (Rainbow et al.,
3.2. Receptors of the HPG axis 1982). Prepubertal male rats have lower levels of PRs in the PVN
compared to adult males (Romeo et al., 2005). It is unknown
In addition to acting other mechanisms of action to be discussed whether PR levels differ in pre-, peri-, and post-pubertal the brains
later, estradiol acts on two main so-called nuclear receptors in of female rodents.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 93

Androgen receptors (ARs) have a high degree of overlap with gonadal, primarily ovarian, steroid hormones in regulation of sex-
the distribution of ERs, particularly in the MeA, BNST, mPOA, ual behavior (Section 3.4.1), depression- and anxiety-like behav-
PVN, VMN and arcuate nucleus (Commins and Yahr, 1985; iors (Section 3.4.2), and cognitive behavior (Section 3.4.3).
Sheridan, 1978; Simerly et al., 1990). Adult male rats have a greater Throughout this review, we discuss alterations in the brain that
number and intensity of AR-staining in the hippocampus (Xiao and may mediate the enduring effects of stressors on the response to
Jordan, 2002), BNST and PVN (Herbison, 1995) compared to ovarian hormones.
females. Adult male mice also express increased levels of AR in
the mPOA and BNST (Lu et al., 1998). It is unknown whether AR 3.4.1. Sexual behavior in adulthood
levels change following pubertal development. Ovariectomized (OVX) mice, unlike rats, guinea pigs, and ham-
Steroid hormone receptors can act via two mechanisms. The sters, typically require several weekly injections of estradiol and
classic, or genomic, pathway involves the ligand-dependent dimer- progesterone followed by mating tests with sexually active males
ization and binding of the receptors to specic hormone response before they begin to express high levels of sexual receptivity
elements on target genes, leading to changes in gene transcription (Blaustein, 2008). However, in the course of an extensive experi-
(Mangelsdorf et al., 1995; Novac and Heinzel, 2004). This classic ment to determine the most appropriate doses of estradiol and
activity of steroid receptors results in long lasting changes in cellu- progesterone to induce female sexual behavior in C57Bl/6 mice,
lar function. The genomic actions of the receptors also involve the we discovered that mice shipped at six weeks old failed to respond
recruitment of co-activators and co-repressors, but discussion of to estradiol and progesterone injections in adulthood with typical
this is beyond the scope of this review (Tetel et al., 2009). More re- increases in the level of sexual receptivity in response to mount
cent evidence suggests that steroid hormone receptors can also attempts by a male mouse (Laroche et al., 2009b). However, mice
activate cytoplasmic signal transduction pathways (Filardo and shipped in adulthood (12 weeks old) expressed the expected
Thomas, 2012; McEwen et al., 2012; Nilsson et al., 2001; behavioral response, indicating that the stress of shipping during
Vasudevan and Pfaff, 2008). pubertal development perturbs the behavioral response to ovarian
hormones later in life (Fig. 3). This vulnerability is limited to the
3.3. Pubertal maturation of the HPG axis pubertal period: C57Bl/6 mice shipped at four, ve or six weeks
old do not respond to estradiol and progesterone in adulthood with
The pubertal maturation of the HPG axis begins with the reac- the usual high levels of receptivity. In fact, mice shipped at six
tivation of GnRH neurons. While GnRH neurons maintain or in- weeks display the most profound perturbations in behavioral re-
crease expression of GnRH mRNA and proteins (Araki et al., sponses (Laroche et al., 2009b). Mice shipped at three weeks old
1975; Jakubowski et al., 1991; Wiemann et al., 1989), their secre- or those shipped at seven weeks old or later responded to estradiol
tory activity is quiescent during the prepubertal period (Sisk and and progesterone with typical levels of sexual receptivity in adult-
Foster, 2004). At the onset of the pubertal period, the frequency hood. Therefore, in C57Bl/6 mice, this period between four to six
and amplitude of GnRH secretion increases gradually until the weeks of age represents a sensitive or vulnerable period in which
GnRH neurons re in synchrony to produce intermittent pulses stressors have an enduring effect on adult behavioral response to
of GnRH release (Plant and Barker-Gibb, 2004). Much work in re- ovarian hormones.
cent years has helped to elucidate the triggers for the reinstate- This sensitivity is not limited to female mice. Male mice shipped
ment of the GnRH pulsatility. A variety of permissive signals may at six weeks old also have a slightly decreased response to testos-
converge on GnRH neurons, including photoperiod (i.e. melatonin), terone in adulthood compared to mice shipped in adulthood. That
energy balance or metabolic (i.e., body fat, leptin), and psychoso- is, peripubertally-shipped male mice display fewer mounts and a
cial signals (Ellison et al., 2012; Sisk and Foster, 2004). These sig- trend towards longer intromission latencies (Laroche et al.,
nals are mediated by the general excitatory and inhibitory 2009b). Recently, McCormick and colleagues (2013) demonstrated
neurotransmitters, glutamate and GABA, respectively (Parent that social instability experienced during pubertal development
et al., 2005). (P3045) leads to reduced sexual performance in male rats in
Most of the work examining the re-emergence of GnRH pulsa- adulthood. Specically, pubertally-stressed males have fewer ejac-
tility has centered on kisspeptin and its receptor, GPR54 (for fur- ulations, a longer latency to ejaculate, and a reduced copulatory
ther review see Ojeda et al., 2006; Sisk and Foster, 2004). efciency, as indicated by more mounts and intromissions prior
Mutations in the genes for either kisspeptin or GPR54 are associ- to ejaculation. This effect is conned to sexual performance, as
ated with low levels of gonadotropin and gonadal steroid levels
and a failure to undergo spontaneous puberty in both human
and rodent models (Oakley et al., 2009). Kisspeptin transcription
increases dramatically prior to puberty, and GPR54 expression be-
comes localized to the GnRH secretory neurons (Gottsch et al.,
2004; Ramaswamy et al., 2008). Additionally, kisspeptin signaling
is both necessary and sufcient for pulsatile GnRH secretion
(Kauffman et al., 2007). Finally, kisspeptin neurons in the arcuate
nucleus express ERs, which may in part modulate steroid feedback
of the HPG axis (Navarro et al., 2011).

3.4. Enduring effects of stressors on responses to gonadal steroids

The pubertal period is important for the maturation of the


stress axis (see Section 4). Further, stressors experienced during
puberty/adolescence alter stress reactivity. However, prior to a Fig. 3. The effect of pubertal shipping on sexual receptivity of C57Bl/6 mice. LQs of
rather serendipitous discovery (reviewed in detail elsewhere: female mice shipped during the peripubertal period. Mice shipped at six weeks and
tested at fourteen weeks display signicantly lower LQs as compared to mice
Blaustein and Ismail, 2013), it was unknown that pubertal stressors shipped at twelve weeks and tested at fourteen weeks during weekly test session 3,
could alter the brains response to gonadal hormones. Here, we dis- 4, and 5. Data represents mean SEM; *p < 0.05. Reprinted with permission from
cuss the ability of a pubertal stressor to alter the response to The Endocrine Society.
94 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

sexual motivation in the pubertally-stressed male rats is unaf-


fected. The stressed males also have lower plasma testosterone
levels, but there is no difference in the levels of androgen or estro-
gen receptors in the mPOA of the stressed and control male rats
(McCormick et al., 2013).
Several controlled stressors were used to duplicate the effect of
shipping in the laboratory. However, standard restraint stress, food
restriction for 36 h, or a multiple stressor regimen (combination of
heat, light and restraint) did not inuence the behavioral response
to estradiol and progesterone in adulthood (Laroche et al., 2009a).
Shipping is a complex, multifactor stressor: animals experience
noise and vibration, temperature uctuations, possible predator
odors, disruption in social hierarchy, etc. In situations such as
our laboratory, they also transition from a light:dark cycle with
lights on early in the morning at the breeders to a shifted and re- Fig. 4. The effect of pubertal immune challenge on sexual receptivity in CD1 mice.
versed light:dark cycle typical for behavioral testing (light off in Lordosis quotient on test weeks 4 and 5 of CD1 mice treated with either saline or
the late morning; lights on in the early evening). It is possible that LPS at 6 or 8 weeks old. Mice treated with LPS at six weeks display signicantly
the shipping stressor may also have elements of circadian disrup- lower LQs as compared to mice treated with saline at six weeks during the weekly
test session 4 and 5. Mice treated with LPS at eight weeks display signicantly
tion beyond the random and intermittent exposure to light and
lower LQs as compared to mice treated with saline at eight weeks only during
dark during the process of shipping itself. It is therefore under- weekly test session 5. Data represents mean SEM; *p < 0.05. Reprinted with
standable that relatively simple stressors are unable to elicit simi- permission from Elsevier, Inc.
lar perturbations in response to gonadal hormones.
Due to the similarities in duration of effect and in behavior of
recently shipped animals and sick animals, injection of the bacte- 3.4.2. Ovarian hormone-modulated depression- and anxiety-like
rial endotoxin lipopolysaccharide (LPS) (Laugero and Moberg, behavior in adulthood
2000) was used as a stressor. LPS causes a robust inammatory While more commonly studied in premenstrual dysphoric dis-
and immune response and induces the display of sickness behavior order (Schmidt et al., 1998) or post-partum depression (Bloch
that lasts for less than forty-eight hours (Anisman, 2009; Dantzer, et al., 2003), changes in hormone levels may contribute to the
2001). Female C57Bl/6 mice injected with LPS at four, ve or six development of mental disease, such as depression and/or anxiety
weeks, but not three, seven, eight, or ten weeks old expressed a de- disorders in women. Studies of women with premenstrual dys-
crease in sexual receptivity in adulthood, compared to saline-in- phoric disorder or post-partum depression demonstrate that ovar-
jected controls (Laroche et al., 2009a), duplicating the same ian hormones trigger anxiety and/or depression during the luteal
developmental sensitive period demonstrated by mice shipped at phase, or at times when ovarian hormone concentrations are rela-
these same ages (Laroche et al., 2009b). Subsequent work has dem- tively low (Bloch et al., 2003; Schmidt et al., 1998).
onstrated that pubertal and adult animals display similar increases Animal models of depression-like behavior include the Porsolt
sickness behavior (e.g., lethargy, huddling, piloerection, and ptosis) forced swim test and the tail suspension test. In these tests, depres-
and decreases in body weight following LPS administration (Ismail sive symptoms of behavioral despair are inferred from decreases in
and Blaustein, 2013; Ismail et al., 2011, 2012; Olesen et al., 2011). times spent swimming and struggling to escape and an increase in
The effects of shipping and LPS exposure are not idiosyncratic to time spent immobile (ONeil and Moore, 2003; Palanza et al.,
the inbred C57Bl/6 mice strain, as similar results have been ob- 2001). Another widely used test measures anhedonia, dened by
tained using the outbred CD1 mice strain (Fig. 4) (Ismail et al., a decrease in sucrose preference (Grippo et al., 2003; Konkle
2011). However, the sensitive period is shifted; in female C57Bl/6 et al., 2003). This test typically involves the presentation of two
mice, the onset of the sensitive period is four weeks old, but in bottles, one with water and another with a solution of 110% su-
CD1 mice, this vulnerability does not occur until after four weeks. crose. The consumption of the sucrose relative to the water is mea-
The sensitive period may last longer in CD1 mice as indicated by sured. Mice and rats typically prefer the sucrose solution, and
the effects of injection of LPS and shipping at eight weeks, but failure to show this preference is taken as a sign of anhedonia.
not ten weeks old (Ismail et al., 2011). Estradiol also modulates depression-like behaviors in rodents
It should be noted that the effects of the controlled stressors, (Bekku and Yoshimura, 2005; Dalla et al., 2004; Okada et al.,
such as restraint or food deprivation, and LPS in pubertal male 1997; Rachman et al., 1998; Suda et al., 2008). OVX increases,
mice have not been examined. Although shipping is effective in and estradiol-replacement decreases, duration of immobility in
reducing behavioral response to testosterone, the efcacy of these the tail suspension (Bernardi et al., 1989) and forced-swim test
other stressors is unknown. Additionally, the effects of shipping or (Koss et al., 2012; Okada et al., 1997; Rocha et al., 2005). Strain dif-
LPS have not been extended to male CD1 mice. ferences in the anti-depressive properties of estradiol have been
The decreased response to estradiol and progesterone in adult- reported. For example, Long-Evans rats have an anti-depressive re-
hood following the pubertal stressors may result from a decrease sponse to estradiol. In contrast, Wistar rats show an increase in
in ERa in some brain areas. That is, CD1 mice shipped at six weeks, depression-like behavior following the withdrawal of estradiol
but not at four weeks, express a decrease in ERa-immunopositive (Koss et al., 2012). Additionally, female mice with a knockout of
cells in the mPOA, VMN and arcuate nucleus, but not in the AVPV the aromatase gene, which are decient in estradiol (as aromatase
(Ismail et al., 2011). It is unknown whether this arises from poten- converts testosterone to estradiol), display greater depression-like
tial volumetric changes in the brain areas, through cell death, or behaviors compared to wild-type mice (Dalla et al., 2004). Overall,
phenotypic changes in which cells express lower levels of ERa. the evidence supports the hypothesis that estradiol is anti-
Additionally, the neuronal (e.g., glutamatergic or GABAergic) or depressive.
even the neural cell type (e.g., neuron or glia) in which this de- LPS exposure during the sensitive period perturbs the anti-
crease in ERa occurs is unknown. Nonetheless, these data suggest depressive properties of estradiol in both C57Bl/6 and CD1 mice
a cellular basis for the decreased behavioral response after the (Fig. 5). Instead of decreasing the behavioral responses to estradiol,
pubertal stressor. as previously demonstrated in sexual behavior, estradiol increased
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 95

depression-like behavior in the tail suspension and forced swim Ovarian hormones also inuence anxiety-like behaviors in fe-
tests (Ismail et al., 2012). Specically, in mice treated with saline male rodents. Both rats (Diaz-Veliz et al., 1989, 1991, 1997; Mora
during pubertal development or adulthood or LPS in adulthood, et al., 1996; Olivera-Lopez et al., 2008; Pandaranandaka et al.,
estradiol induced the expected decrease in depression-like behav- 2006, 2009) and mice (Galeeva and Tuohimaa, 2001) display cycles
iors, as measured by a decrease in immobility. However, in mice in anxiety-like behaviors that correlate with the estrous cycle. Spe-
treated with LPS during pubertal development, estradiol treatment cically, anxiety-like behaviors are increased during diestrus,
increases the duration of immobility (Ismail et al., 2012). That is, when estradiol levels are low, and females display a reduction in
the pubertal immune challenge reversed the effects of estradiol. anxiety during proestrus and estrus (when estradiol levels are ele-
Interestingly, neither estradiol treatment, nor pubertal LPS expo- vated). Moreover, exogenous estradiol treatment during diestrus
sure, affected the sucrose preference test, which measures anhedo- reduces anxiety-like behaviors to proestrus-like levels (Marcondes
nia. While it is possible that a higher concentration of sucrose et al., 2001). OVX also increases anxiety levels in rats (Diaz-Veliz
would have resulted in a decrease in sucrose preference, these data et al., 1997; Pandaranandaka et al., 2009; Picazo et al., 2006) and
may suggest that different mechanisms may mediate behavioral mice (Ogawa et al.). Replacement of estradiol and progesterone de-
despair and anhedonia aspects of depression-like behaviors. creases levels of anxiety-like behaviors in female rats (Diaz-Veliz
Typical animal models of anxiety-like behaviors are based upon et al., 1989, 1991, 1997; Mora et al., 1996; Olivera-Lopez et al.,
the innate aversion of rodents to open spaces. Two such tests are 2008; Pandaranandaka et al., 2006, 2009). OVX mice, however, dis-
the elevated plus maze and the open eld arena. The elevated plus play a biphasic response to estradiol; while chronic treatment with
maze is based upon the animals fear of open and unprotected low doses of estradiol reduces anxiety-like behaviors (Tomihara
spaces, with decreased time exploring the open arms interpreted et al., 2009), higher levels of estradiol may increase anxiety
as an increase in anxiety-like behavior (Roy and Chapillon, 2004; (Morgan and Pfaff, 2001, 2002; Tomihara et al., 2009).
Wall and Messier, 2001). The open eld test measures the explor- As with the anti-depressive properties of estradiol, LPS expo-
atory behavior of the rat or mouse, as well as its willingness to sure during the pubertal period perturbs the anxiolytic properties
move away from the arena wall and into the center. Reductions of estradiol and progesterone in both C57Bl/6 and CD1 mice. In
in both exploratory behavior and entries into the center are taken control OVX mice, an injection of estradiol benzoate followed
as evidence for an increase in anxiety (Roy and Chapillon, 2004). 44 h later by an injection of progesterone, similar to treatment
The lightdark box also takes advantage of the rodents natural used in tests of sexual receptivity, decreases anxiety-like behavior
preference for dark spaces, so increases in time spent in the light as assessed in the elevated plus maze, the lightdark box, and the
compartment are interpreted as decreases in anxiety-like behavior. marble burying task (Fig. 6) (Olesen et al., 2011). In OVX mice that
Other measures of anxiety include enhanced startle reexes, de- received the LPS during the pubertal period, hormone treatment
creases in the approach to novel food or social interactions (File either blocked the anxiolytic effect of estradiol and progesterone
and Seth, 2003; Johnston and File, 1991), and more rapid marble or had an anxiogenic effect (Olesen et al., 2011). That is, an im-
burying in the marble burying test (which also may model compul- mune challenge in puberty either eliminates or reverses the behav-
sive behavior (Li et al., 2006)). ioral responses to ovarian hormones in adulthood.
It should be noted that there was a tendency for LPS exposure
during pubertal development to affect the display of depression-
and anxiety-like behaviors even in the absence of hormones. OVX
mice exposed to LPS in the pubertal peroid and treated with oil in
adulthood showed a reduction in time spent immobile, indicating a
decrease in depression-like behavior (Ismail et al., 2012). There
was also a tendency for pubertal LPS injection to reduce anxiety-
like behaviors in adulthood in both the lightdark box and the ele-
vated plus maze. This is consistent with previous research demon-
strating that adolescent stressors (Conrad and Winder, 2011) or

Fig. 5. The effects of pubertal immune challenge on depression-like behavior in


adult female mice. (A) Duration of immobility during the forced swim test in C57Bl/
6 mice treated with either saline or LPS at six and eight weeks old and implanted Fig. 6. The effects of pubertal immune challenge on anxiety-like behavior in CD1
with estradiol or oil vehicle capsules in adulthood. (B) Duration of immobility mice. Estradiol followed by progesterone administration 48 h later increased the
during the tail suspension test in CD1 mice treated with saline or LPS at six and ten latency to bury marbles in the saline- but not the LPS-treated mice compared to
weeks old and implanted with estradiol or oil vehicle capsules in adulthood. Data respective oil controls. Data represents mean SEM; *p < 0.05. Reprinted with
represents mean SEM; *p < 0.05. Reprinted with permission from Elsevier, Inc. permission from Elsevier, Inc.
96 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

prenatal LPS treatment (Wang et al., 2010) reduce anxiety-like during the vulnerable period, (Ismail and Blaustein, 2013). As with
behaviors. Nevertheless, in all cases, hormone treatment was many other studies, the cognitive tasks used in this study depend
either without an effect or caused an increase in depression- and upon the innate exploratory behavior of the mice, decreases of
anxiety-like behaviors in mice that were injected with LPS during which are indicators of anxiety. Therefore, the possibility cannot
the pubertal period. be excluded that the apparent decreases in cognition are a result
The alteration of the behavioral properties of the ovarian hor- of the increased anxiety- and depression-like behaviors.
mones, particularly estradiol, may be mediated in part by alter- Estradiol affects cognition in both a long lasting (from hours to
ation of the ovarian hormone receptors in the brain. While the days) manner and a short-term or rapid manner (occurring within
antidepressant effects of estradiol are mediated predominantly minutes). The long-lasting effects are consistent with the classical,
by ERb (Osterlund, 2010; Rocha et al., 2005), both subtypes of ER gene-transcription actions of ERs, while membrane receptors act-
have been implicated in the mediation of the anxiolytic properties ing through signal transduction pathways mediate the short-term
of estradiol (Choleris et al., 2003, 2006; Imwalle et al., 2005; Krezel effects (McEwen et al., 2012). The preponderance of data demon-
et al., 2001; Ogawa et al., 1998). Therefore, the inability of estra- strate that the pro-cognitive effects of estradiol are mediated
diol, or estradiol and progesterone, to decrease depression- and/ primarily through ERb, rather than ERa (reviewed in Luine and
or anxiety-like behaviors in mice treated during pubertal develop- Frankfurt, 2012). The numbers and types of dendritic spines, which
ment with LPS suggests an alteration in ERa and/or ERb in relevant are associated with increases in innervation and serve as sites of
brain regions, such as the BNST, amygdala or hippocampus. synaptic input, are critical mediators of neural plasticity in the
Although the effects of shipping stress on ERa levels in some brain adult brain (reviewed in Urbanska et al., 2012). Moreover, the
areas described above are suggestive, this hypothesis remains to be mechanisms of hippocampal learning, memory, and cognition in-
directly tested in future experiments. volve dendritic remodeling. In the CA1 region of the hippocampus
of adult male (Jedlicka et al., 2008; Leuner et al., 2003) and female
3.4.3. Estradiol-modulated cognitive behaviors in adulthood rats (Beltran-Campos et al., 2011; Woolley, 2000), learning is cor-
Cognition, learning, and memory are complex processes that in- related with increases in dendritic spine density. Estradiol in-
volve the learning or acquisition of information, the consolidation creases synaptic activity in the CA1 region of the hippocampus
and storing of information, and then the retrieval and utilization of via alteration of both excitatory (Smejkalova and Woolley, 2010)
the information. Some of the most commonly used learning and and inhibitory transmission (Huang and Woolley, 2012), which
memory tasks involve spatial memory, or learning where an object can lead to new synaptic connections as indicated by dendritic
or place is in relation to the environment. Spatial memory is typi- spines (Yankova et al., 2001). Estradiol may enhance cognition
cally measured using the radial arm maze, in which animals must through the generation of new spines or an alteration of existing
learn the orientation of arms using spatial cues in order to retrieve spines (reviewed in Luine and Frankfurt, 2012). Therefore, it is pos-
food, and the Morris water maze, in which animals learn the loca- sible that the experience of a pubertal stressor may interfere with
tion of a submerged escape platform. Other memory tasks involve these aspects of synaptic plasticity and learning and memory. This
the ability to recognize a familiar object or conspecic animal or a is an attractive hypothesis to be tested in subsequent experiments.
familiar object in a new place. The recognition tasks take advan-
tage of the innate exploratory behavior of rodents. One major
assumption of the recognition tasks is that rodents more readily 4. Adrenal steroids
investigate a new or novel object or conspecic than one that is
familiar (Luine, 2008; Luine and Frankfurt, 2012). Both spatial 4.1. Hypothalamicpituitaryadrenal (HPA) axis
memory tasks and recognition tasks depend upon an intact hippo-
campus (Broadbent et al., 2004). The physiological response to a stressor occurs in two distinct
In adulthood, estradiol generally has a positive effect on a vari- components: activation of the sympathetic nervous system and
ety of learning tasks, including spatial and recognition memory activation of the hypothalamicpituitaryadrenal (HPA) axis. The
(reviewed in Luine, 2008), but the specic effects of estradiol de- sympathetic nerves and adrenal medulla immediately release cat-
pend upon the cognitive task and the brain regions(s) involved. echolamines following the experience of a stressor (Kvetnansky
Estradiol impairs performance in striatal or amygdalar tasks, in et al., 2009). This release of catecholamine in the periphery results
which some learned, associative response is required in female rats in increased glucose utilization, heart rate and pupil dilation, bron-
(Davis et al., 2005; Korol, 2004). In hippocampal, or spatial or place
tasks, estradiol enhances performance in female rats (Daniel et al.,
1997; Davis et al., 2005; Luine et al., 1998; Sandstrom and Wil-
liams, 2004) and mice (Li et al., 2004; Xu and Zhang, 2006).
Pubertal exposure to LPS disrupts the positive cognitive effects
of estradiol on social memory, as measured by social discrimina-
tion and recognition tasks, and object memory, as measured by no-
vel object recognition and novel object placement tests (Ismail and
Blaustein, 2013). More specically, in mice treated with saline dur-
ing pubertal development or with LPS or saline in adulthood, estra-
diol improves social memory, as measured by increases in the
duration of investigation of a novel stimulus mouse. In contrast,
in mice treated with LPS during pubertal development, estradiol
treatment failed to increase the time spent investigating the novel
mouse (Ismail and Blaustein, 2013). Estradiol also increased per-
formance in the object recognition and object placement recogni-
tion tasks in mice treated with saline during the pubertal period Fig. 7. The effect of pubertal immune challenge on cognitive behavior in CD1 mice.
Estradiol increase the percentage of time spent investigating the novel object
or treated in adulthood (Fig. 7). Additionally, estradiol treatment during the object recognition test in mice in treated pubertally with saline, but not
failed to increase the time spent investigating the novel object or LPS compared to respective oil controls. Data represents mean SEM; *p < 0.05.
the novel placement of a familiar object in mice treated with LPS Reprinted with permission from Elsevier, Inc.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 97

chodilation, decreased gut motility, and vasoconstriction, all of MRs. The highest densities of GRs are in the hippocampus, medial
which would promote survival in the case of an immediate physi- prefrontal cortex, and the amygdala, which provides input, via the
cal stressor (Kvetnansky et al., 2009). The HPA axis, however, re- BNST, to the PVN (Ulrich-Lai and Herman, 2009). The hippocampus
sponds to a stressor by releasing glucocorticoids (primarily and infralimbic medial prefrontal cortex (mPFC) are particularly
cortisol in humans, primates, and some other species and primarily involved in the negative feedback loop, whereas the amygdala
corticosterone in rats and mice) in a slower, and potentially, a more and prelimbic mPFC activate the HPA axis (Ulrich-Lai and Herman,
long-lasting way (Fig. 8). Blood levels of glucocorticoids peak 2009).
around 1020 min following a stressor; the glucocorticoids then
initiate gene transcription, beginning approximately one hour 4.3. Pubertal maturation of the HPA axis and stress response
following the stressor (Sapolsky et al., 2000; Schommer et al.,
2003). The HPA axis serves to maintain allostasis, in which Maturation of the HPA axis precedes the maturation of the HPG
adaptive changes occur to both achieve physiological stability axis (Fig. 1). This maturation results in increased secretion of hor-
(McEwen, 2000) and inuence a host of other processes, such as mones by the adrenal glands, termed adrenarche, which in humans
energy metabolism (Brandi et al., 1993; Rizza et al., 1982; Vitaliano takes places around 68 years of age (Parker, 1991) and is the re-
et al., 2002), immune function (Straub et al., 2011), cardiovascular sult of increased 17-hydroxylase activity. At this stage of develop-
health (Sapolsky and Share, 1994; Whitworth et al., 1995), and as ment, cortisol increases in humans (Apter et al., 1979; Walker
recent evidence suggests, even mental health (Pariante and Miller, et al., 2001), and corticosterone increases in rats and mice (Pigna-
2001; Tafet and Bernardini, 2003). telli et al., 2006; Spinedi et al., 1997). Basal corticosterone concen-
trations increase in both male and female rats during the peri-
4.2. Receptors of the HPA axis pubertal periods and peak around P4560. Moreover, females have
a steeper increase than males, resulting in a higher concentration
Two types of receptors mediate the genomic actions of the glu- of plasma corticosterone levels in female rats even in adulthood
cocorticoids: the mineralocorticoid (MR) and the glucocorticoid (Pignatelli et al., 2006). Other steroids not typically associated with
(GR) receptor. While both MR and GR bind corticosterone, GRs the adult rat adrenal gland are present in the pre-pubertal period.
are more directly involved in mediating a negative feedback loop Specically, cortisol, androstenedione, and 17-hydroxyprogester-
for corticosterone secretion. MRs have a higher afnity for cortico- one are detected with peak concentrations at P20 in both female
sterone than GRs; therefore, the majority of MRs being occupied and male rats; these peaks correlate with an increase in 17-
even under low basal levels of glucocorticoids (reviewed in De hydroxylase expression in the adrenal glands (Pignatelli et al.,
Kloet et al., 2008). As only approximately 20% of GRs are occupied 2006).
in basal conditions, GRs are more sensitive to changes in the con- Unlike gonadal hormones, basal ACTH and corticosterone levels
centrations of glucocorticoids (reviewed in Datson et al., 2008). are relatively stable during pubertal development and into adult-
Moreover, GRs are more widely distributed in the brain than hood (Pignatelli et al., 2006; Romeo et al., 2004a,b). Additionally,

Fig. 8. Representation of the hypothalamuspituitaryadrenal axis. The experience of a stressor cause sensory, hindbrain, limbic, and cortical regions to signal to neurons in
the paraventricular nucleus of the hypothalamus (PVN). The PVN then secretes corticotrophin releasing hormone (CRH) and arginine vasopressin (AVP) into the hypophyseal
portal system of the median eminence. The portal blood carries CRH and AVP into the pituitary gland and activates their respective receptors to triggers synthesis and
secretion of adrenocorticotropic hormone (ACTH) from the anterior pituitary. ACTH then stimulates the release of glucocorticoids (cortisol in humans and primates and
corticosterone in rats and mice) from the adrenal cortex. The elevated levels of glucocorticoids suppress further release of CRH, AVP and ACTH by binding to receptors in the
PVN, hippocampus, and prefrontal cortex and pituitary. (+), solid lines, positive feedback; (), dashed lines negative feedback.
98 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

pre-pubertal (P28) and adult male and female rats respond to an 4.4.1. Stress response in adulthood
acute stressor (e.g. restraint stress) with comparable increases in Examinations of the enduring effects of stressors in the pubertal
ACTH and corticosterone; however, pre-pubertal male and female period on adult HPA function and stress reactivity in adulthood
rats have a more prolonged, stress-induced corticosterone re- have yielded somewhat mixed results. It has generally been re-
sponse compared to adult rats (Romeo et al., 2004a,b). In adult rats, ported that stressors experienced in puberty and adolescence in-
repetitive exposure to the same stressor results in a habituation of crease or augment basal HPA function (Bazak et al., 2009;
the corticosterone response (Girotti et al., 2006; Harris et al., 2004; Jacobson-Pick and Richter-Levin, 2010; Pohl et al., 2007; Schmidt
Helmreich et al., 1997; Magarinos and McEwen, 1995; Marti and et al., 2007, 2010b; Sterlemann et al., 2008; Uys et al., 2006a,b)
Armario, 1997), but male rats that experience repeated stressors or responses to stressors (Isgor et al., 2004; Jacobson-Pick and
in the pre-pubertal period do not show this same blunting of cor- Richter-Levin, 2010; Mathews et al., 2008; Weathington et al.,
ticosterone response. In fact, they have higher peaks of ACTH, cor- 2012) as indicated by corticosterone levels in adulthood (Table 1).
ticosterone and adrenal progesterone, but a more rapid return to While the basal increases of corticosterone occur in both male and
baseline levels (Doremus-Fitzwater et al., 2009; Romeo et al., female rats and male CD1 mice, there also may be interactions with
2006). Pubertal female rats do not show this increase in corticoste- gonadal hormones, as Mathews et al. (2008) reported stressor-in-
rone response (Doremus-Fitzwater et al., 2009). duced increases in corticosterone only in female rats in the dies-
It is well-established that gonadal hormones inuence HPA trous stage of the estrous cycle. There may also be interactions
reactivity. In adult male rats, testosterone decreases the peak of with circadian rhythms, as some reports indicate that basal corti-
corticosterone levels and recovery time from a stressor (Handa costerone levels are increased in the morning (Schmidt et al.,
et al., 1994; McCormick et al., 1998, 2002; Viau, 2002; Viau and 2007, 2010b; Sterlemann et al., 2008; Uys et al., 2006b) and de-
Meaney, 1996), whereas the ovarian hormones increase these creased in the afternoon (Sterlemann et al., 2008; Toth et al.,
measures in females (Carey et al., 1995; McCormick et al., 2002; 2008a). Pubertal stressors lead to enhanced basal corticosterone
Redei et al., 1994; Viau and Meaney, 1991; Young et al., 2001). and decreases in the expression of GR and MR in the hippocampus
While it seems plausible that the differential levels of gonadal in adulthood (Isgor et al., 2004; Schmidt et al., 2007; Sterlemann
hormones would contribute to the altered stress reactivity in et al., 2008; Uys et al., 2006b), suggesting enduring alterations in
pre-pubertal and adult animals, the protracted stress response in the negative feedback of the HPA axis.
pre-pubertal male and female rats is not affected by the presence In contrast, several studies report that there are no enduring
or absence of gonadal hormones (Romeo et al., 2004a,b). In effects of a stressor during puberty in male or female rats on either
mid-pubertal (P40) male rats, androgens modulate HPA reactivity, basal (Watt et al., 2009; Weathington et al., 2012) or stress-
indicating that pubertal development need not be complete before induced plasma ACTH or corticosterone level following a second
adult-like patterns of stress reactivity emerge (Gomez et al., 2004). stressor in adulthood (Bourke and Neigh, 2011; Maslova et al.,
It is intriguing that this is also around the same time during which 2002; Mathews et al., 2008; McCormick et al., 2005, 2008;
the adult-like sex differences in corticosterone levels emerge Toledo-Rodriguez and Sandi, 2007; Wilkin et al., 2012; Wright
(Pignatelli et al., 2006). et al., 2008) (Table 1). The adolescent stressors used in these
One of the key brain regions of the HPA axis, the PVN, exhibits studies include physical (e.g. restraint, foot shock, sleep
maturation in functionality during the pubertal period. The PVN deprivation), social (e.g., social instability or isolation), or predator
volume, cellular size, and morphology are similar in pubertal and odor; in adulthood these animals experienced well-established and
adult rats (Romeo et al., 2007). Moreover, the numbers of CRH commonly-used stressor procedures (e.g., restraint, foot-shock,
and AVP cells are also similar (Romeo et al., 2006). There is, how- elevated platform, and the forced swim). However, differences in
ever, a greater activation (i.e., Fos expression) of cells within the procedures may explain the apparent discrepancies. In several of
PVN following a single stressor in pre-pubertal compared to adult the studies in which no effect of pubertal stressor on adult stress
male rats (Romeo et al., 2006). This greater activation is specic to reactivity was reported, the animals experienced the stressor
the CRH cells, suggesting a correlation between this increased beginning at 30 (McCormick et al., 2005, 2008) or 40 days old
activation of CRH cells and the protracted corticosterone response (Wright et al., 2008), whereas the animals demonstrating
following a stressor in pre-pubertal males. alterations in stress response in adulthood experienced the
stressors starting pre-pubertally (at or before P28) (Ilin and
4.4. Enduring effects of commonly-used stressors Richter-Levin, 2009; Isgor et al., 2004; Jacobson-Pick and
Richter-Levin, 2010; Pohl et al., 2007; Schmidt et al., 2007;
In humans, the experience of extreme stress and trauma during Sterlemann et al., 2008; Weathington et al., 2012). It will be
adolescence is associated with a greater likelihood of mood disor- important to determine if the difference in the ndings between
ders, such as anxiety, major depression, and post-traumatic stress the different studies is due to the presence of a vulnerable period
disorder (PTSD) (Ge et al., 2001; Grant et al., 2003, 2004; Silverman ending prior to P30. That is, perhaps pre-pubertal, not pubertal,
et al., 1996; Turner and Lloyd, 2004). In recent years, several stud- rats and mice are sensitive to this enduring alteration of HPA axis
ies with animal models have addressed the enduring effects of function.
stressors experienced during the juvenile and pubertal periods
on stress reactivity in adulthood (Section 4.4.1). These studies sup- 4.4.2. Anxiety- and depression-like behavior in adulthood
port the notion that stress experienced in pubertal development In general, the experience of repeated stressors such as chronic
leads to an increase in anxiety- and depression-like behaviors in variable stress (Ilin and Richter-Levin, 2009; Jacobson-Pick and
both male and female rats (Section 4.4.2). There is also the sugges- Richter-Levin, 2010, 2012; Wilkin et al., 2012), elevated platform
tion that alterations in cognition occur in adulthood following stress (Avital et al., 2006; Avital and Richter-Levin, 2005), social
pubertal stress (Section 4.4.3). It is important to note that the mat- instability (Green et al., 2012; Mathews et al., 2008; McCormick
uration of the HPA axis occurs prior to maturation of the HPG axis. et al., 2008; Schmidt et al., 2007, 2010a,b; Sterlemann et al.,
Therefore, many of the studies focus on stressors in the pre- or 2008), social defeat procedures (Bourke and Neigh, 2011; Vidal
early pubertal periods. Additionally, once animals have progressed et al., 2007, 2011a,b; Weathington et al., 2012), and predator odors
through gonadarche, they seem to be less susceptible to stress-in- (Bazak et al., 2009; Cohen et al., 2007; Tsoory et al., 2007; Wright
duced changes, again demonstrating a vulnerable period to the et al., 2008, 2012a,b) increases anxiety-like behaviors in male and
enduring effects of stressors. female rats and mice in adulthood (Table 2; for further review see
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 99

Table 1
Experiments investigating the effects of stressors in puberty and adolescence on the HPA axis reactivity in adulthood.

Sex/strain/ Age at stress Stressor Age at test Test Result: HPA reactivity Source
species
# W rats 2132 Chronic variable stress 120 Cort ; Maslova et al. (2002)
# SD rats 2856 Chronic variable stress 77 Cort " Isgor et al. (2004)
GR mRNA ; in CA1, DG
# SD rats 23, 35, 60 Chronic variable stress 68 Basal ACTH Uys et al. (2006a)
Basal Cort ;
# SD rats 23, 35, 60 Chronic variable stress 61 Basal Cort (am) " Uys et al. (2006b)
GR " in hilus, DG
# SD rats 3058 Chronic variable stress 60 Basal Cort (am) Toth et al. (2008b)
Basal Cort (pm) ;
# SD rats 2729 Chronic variable stress 60 Basal Cort " Ilin and Richter-Levin (2009)
# SD rats 2729 Chronic variable stress 60 Cort " Jacobson-Pick and Richter-Levin (2010)
Mild/severe Pohl et al. (2007)
$ LE rats 2351 Chronic variable stress 72 Basal Cort /"
# LE rats 2233 Chronic variable stress 61 Cort ; Wilkin et al. (2012)
$ LE rats 2233 Chronic variable stress 61 Cort ;
# LE rats 3546 Chronic variable stress 73 Cort
$ LE rats 3546 Chronic variable stress 73 Cort
# W rats 2628 Repeated elevated platform 56 DHEA in hypothalamus, EC Avital et al. (2006)
DHEAS " in hypothalamus, EC
# LE rats 3348 Social instability 69 Cort McCormick et al. (2005)
$ LE rats 3348 Social instability 69 Cort
# LE rats 3345 Social instability 70 Cort Mathews et al. (2008)
$ LE rats 3345 Social instability 70 Cort
# LE rats 3345 Social instability 70 Cort McCormick et al. (2008)
$ LE rats 3345 Social instability 70 (estrus) Cort "
$ LE rats 3345 Social instability 70 (disestrus)
# CD-1 mice 2884 Social instability 92 Basal Cort Schmidt et al. (2007)
Basal ACTH (am) "
Basal ACTH (pm)
MR mRNA ; in CA2
GR mRNA ; in CA1
# CD-1 mice 2773 Social instability 450 Basal ACTH Sterlemann et al. (2008)
Basal Cort (am) "
Basal Cort (pm) ;
CRH mRNA
AVP mRNA " in PVN
MR mRNA ; in CA1, in DG
GR mRNA ; in CA1CA3, DG
Resilient/vulnerable Schmidt et al. (2010a,b)
# CD-1 mice 3080 Social instability 120 Basal Cort (am) /"
# W rats 3749 Social defeat 96 Cort Bourke and Neigh (2011)
# SD rats 3540 Social defeat 63 Basal Cort Watt et al. (2009)
# LE rats 2841 Social defeat 87 Basal Cort Weathington et al. (2012)
Cort
$ LE rats 2841 Social defeat 87 Basal Cort
Cort "
# W rats 2830 Predator odor 120 Cort Toledo-Rodriguez and Sandi (2007)
$ W rats 2830 120
# SD rats 28 Predator odor 60 Basal Cort " Bazak et al. (2009)
# LE rats 4048 Predator odor 62 Cort Wright et al. (2008)
$ LE rats Cort

LE, Long Evans; SD, SpragueDawley; W, Wistar; AVP, arginine vasopressin; CA1, Cornu Ammonis area 1 of the hippocampus; CA2, Cornu Ammonis area 2 of the hippo-
campus; CA3, Cornu Ammonis area 3 of the hippocampus; CRH, corticotropin-releasing hormone; DG; dentate gyrus of the hippocampus; EC, entorhinal cortex; GR,
glucocorticoid receptor; MR, mineralocorticoid receptor; PVN, paraventricular nucleus of the hypothalamus.

McCormick and Green, 2012). The increases in anxiety-like behav- social defeat also lead to a decrease in social interactions in
iors are typically indicated by decreases in the time spent in the adulthood (Green et al., 2012; Vidal et al., 2007, 2011a,b). While
open arm of the elevated plus maze (McCormick and Green, a few studies report lack of effect of stressors during pubertal
2012; McCormick et al., 2008; Schmidt et al., 2007; Sterlemann development on locomotor or exploratory behavior, these stressors
et al., 2008; Tsoory et al., 2007; Uys et al., 2006a). In one study, began on P30 and continued until well into adulthood (Peleg-
both female and male rats that experienced severe, variable phys- Raibstein and Feldon, 2011; Toth et al., 2008a,b), perhaps missing
ical stress exhibited exaggerated anxiety-like behavior in the an earlier vulnerable period.
elevated plus maze, as indicated by jumping to escape from the There may be sex differences in the effects of stressors during
maze (Pohl et al., 2007). An increase in anxiety-like behavior in the pubertal period on subsequent anxiety-like behavior in
adulthood is also displayed by decreases in locomotor and explor- adulthood. Following 10 days of foot-shock stress during the
atory behavior in the open eld following pre-pubertal (P28) peri-pubertal period, female, but not male mice, display an
stress (Avital and Richter-Levin, 2005; Schmidt et al., 2007; increase in startle reexes in adulthood (Table 2), suggestive of
Sterlemann et al., 2008; Tsoory et al., 2007). Social instability stress increased anxiety. If the mice are stressed with another foot-shock
in the early pubertal period also increases open eld anxiety and prior to anxiety testing, both male and female mice exhibit
decreases social interaction in adult male rats, while maintaining increases in anxiety-like behavior (Chester et al., 2008). The
exploratory behavior (Green et al., 2012). Social instability and reverse effects have also been reported: under similar conditions,
100 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Table 2
Experiments investigating the effects of stressors in puberty and adolescence on anxiety- and depression-like behaviors in adulthood.

Sex/strain/species Age at stress Stressor Age at test Test Result: Anxiety and depression Source
# W rats 2132 Chronic variable stress 120 EPM Maslova et al. (2002)
# SD rats 3058 Chronic variable stress 67 EPM Toth et al. (2008b)
FST
SC
# SD rats 2729 Chronic variable stress 60 EPM " Ilin and Richter-Levin (2009)
OFT "
# SD rats 2729 Chronic variable stress 60 EPM " Jacobson-Pick and Richter-Levin (2010)
OFT "
$ SD rats 2729 Chronic variable stress 60 EPM "
OFT "
# SD rats 2729 Chronic variable stress 60 EPM " Jacobson-Pick and Richter-Levin (2012)
# SD rats 3090 Chronic variable stress 90 OFT Toth et al. (2008a)
SI "
Mild/severe Pohl et al. (2007)
# LE rats 2351 Chronic variable stress 72 EPM /;
SC /
$ LE rats 2351 Chronic variable stress 72 EPM /;
SC "/"
# LE rats 2233 Chronic variable stress 61 EPM " Wilkin et al. (2012)
FST "
$ LE rats 2233 Chronic variable stress 61 EPM "
FST
# LE rats 3546 Chronic variable stress 73 EPM ;
FST
$ LE rats 3546 Chronic variable stress 73 EPM ;
FST
# C57Bl6 mice 2529 Chronic variable stress 81138 EPM Peleg-Raibstein and Feldon (2011)
OFT
# W rats 28 Repeated elevated platform 90 OFT Avital and Richter-Levin (2005)
SR
2628 Repeated elevated platform 60 OFT "
SR "
# W rats 2628 Repeated elevated platform 56 OFT " Avital et al. (2006)
# LE rats 3045 Social instability 70 FST Mathews et al. (2008)
$ LE rats 3045 Social instability 70 FST
# LE rats 3045 Social instability 70 EPM " McCormick et al. (2008)
$ LE rats 3045 Social instability 70 (estrus) EPM ;
$ LE rats 70 (diestrus) "
# LE rats 3045 Social instability 70 OFT " Green et al. (2012)
SI "
SA
# CD-1 mice 2884 Social instability 92 EPM " Schmidt et al. (2007)
NISF "
# CD-1 mice 2773 Social instability 77 EPM " Sterlemann et al. (2008)
NISF "
450 EPM "
NISF
Resilient/vulnerable Schmidt et al. (2010b)
# CD-1 mice 3080 Social instability 120 EPM /"
TST /"
$ CD-1 mice 3080 Social instability 120 EPM Schmidt et al. (2010a)
NISF "
# W rats 4557 Social defeat 78 SA " Vidal et al. (2007)
# W rats 4545 Social defeat 67 WCT " Vidal et al. (2011a)
# WT rats
# W rats 4558 Social defeat 78, 108 SA " Vidal et al. (2011b)
# WT rats
$ W rats "
$ WT rats
# W rats 3749 Social defeat 96 EPM Bourke and Neigh (2011)
SC
FST
$ W rats 3749 Social defeat EPM
SC "
FST "
# SD rats 3540 Social defeat 58 EPM ; Watt et al. (2009)
# LE rats 2841 Social defeat 63 EPM ; Weathington et al. (2012)
OFT
SI
FST
$ LE rats 2841 Social defeat 63 EPM "
OFT
SI "
FST "
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 101

Table 2 (continued)

Sex/strain/species Age at stress Stressor Age at test Test Result: Anxiety and depression Source
# SD rats 28 Predator odor 63 EPM " Tsoory et al. (2007)
# SD rats 28 Predator odor 60 EPM " Bazak et al. (2009)
# W rats 28 Predator odor 60 EPM " Cohen et al. (2007)
# W rats 3356 Predator odor 58 SI ; Kendig et al. (2011)
EPM
FST ;
# LE rats 4048 Predator odor 60 OFT " Wright et al. (2008)
$ LE rats OFT "
# LE rats 3846 Predator odor 68, 70 OFT " Wright et al. (2012a,b)
SI "
$ LE rats 3846 Predator odor 68, 70 OFT "
SI "

LE, Long Evans; SD, SpragueDawley; W, Wistar; EPM, elevated plus maze; FST, forced swim test; NISF, novelty-induced suppression of feeding; OFT, open eld test; SA, social
approach; SC, sucrose consumption; SI, social interaction; SR, startle response; WCT, water conict test.

male rats displayed increases in anxiety in the shock probe burying hippocampal-dependent, spatial learning in which an animal must
task, while female rats expressed a decrease in anxiety-like behav- nd a hidden platform) in adulthood (Isgor et al., 2004). Moreover,
iors (Pohl et al., 2007). It should be noted that the female rats in the impaired cognition is not present during the pubertal period,
this study were intact and no mention was made regarding stage but it emerges during adulthood. Learning in the Morris water
of estrous cycle. Therefore, the possibility of hormonal status maze is also impaired following repeated exposures to the stress
affecting behavioral results remains. McCormick and colleagues of an elevated platform during the pre-pubertal period (P2628)
(2013)have shown that female rats increase anxiety in adulthood in adult male rats, but if the pre-pubertally-stressed rats are
following peri-pubertal stress (P3045) only during the diestrous re-exposed to a stressor in adulthood, learning is improved (Avital
phase of the estrous cycle (i.e., a period characterized by low levels and Richter-Levin, 2005). Taken together, these data suggest that
of estradiol and progesterone) (see Section 3.4.2). physical stressors experienced in puberty contribute to enduring
The evidence for effects of pubertal stress on depression-like decits in cognition in adulthood.
behavior in adulthood is not as strong. Female rats, exposed to The impaired performance on the Morris water maze in animals
chronic variable stressors during puberty (P2351) exhibit an in- exposed to variable physical stress in pubertal development is cor-
crease in anhedonia, as measured by a decrease in sucrose prefer- related with an inhibition of growth in the pyramidal cell layer of
ence in adulthood (Pohl et al., 2007). Social defeat stress during the CA1 and in the granular cell layer of the dentate gyrus of the
pubertal development (P2849) also increased depression-like hippocampus. Furthermore, the pre-pubertal stress also prevents
behavioral despair in adult female, but not male rats (Bourke and further growth of the pyramidal cell layer of the CA3 of the hippo-
Neigh, 2011; Weathington et al., 2012), reecting a fundamental campus. Decreases in the volume of CA1 and CA3 of the hippocam-
sex difference in the vulnerability to insults during the pubertal pus are also associated with down-regulation of hippocampal GR
period on adult behavior. However, other studies report no effects expression (Isgor et al., 2004). There is contradiction as to the
of chronic variable stress or social instability stress on either su- effects of peri-pubertal stress on brain-derived growth factor
crose preference (Toth et al., 2008b) or the forced swim test (Math- (BDNF) in the hippocampus with either no effect or increases in
ews et al., 2008; Toth et al., 2008b) in adulthood. Furthermore, in BDNF reported (Toth et al., 2008b; Uys et al., 2006a). Intriguingly,
one report the experience of predator odor during the peri-puber- in the work by Toth et al. (2008b), increases in BDNF were
tal period decreased depression-like behaviors in adult male rats correlated with increased neurogenesis, which may represent an
(Kendig et al., 2011). There also may be inherent sensitivities to attempt to recover from the enduring effects of adolescent
the pubertal stress on adult depression-like behaviors. For exam- stressors on the hippocampus.
ple, Schmidt and colleagues demonstrated that some outbred, Three days of stress (forced swim, elevated platform, and
CD1 male mice express increases in CRH and alterations in anxiety- foot-shock) during the pre-pubertal (P2629) or early pubertal
or depression-like behavior in adulthood, whereas other strains of (P3335) periods of male rats are sufcient to induce poor perfor-
mice do not (Schmidt et al., 2010b). The vulnerable mice also ex- mance in the shuttle avoidance task (in which a rat learns to move
press decreases in MR expression in the hippocampus, relative to to a different compartment to avoid an electric shock) in adulthood
the resilient mice, indicating a possible dysregulation in the tonic (Avital et al., 2006; Avital and Richter-Levin, 2005). In another
inhibitory tone on the HPA. study of shuttle avoidance learning, the experience of a multiple
stressor paradigm (food restriction, acute swim test, and elevated
4.4.3. Cognitive behavior in adulthood platforms from P30 until P90) was without effect (Toth et al.,
Areas such as the hippocampus are implicated in cognition, 2008a). It is possible that the prolonged exposure to a stressor from
learning and memory and continue to develop over the course of early puberty until well into adulthood resulted in a desensitiza-
puberty and adolescence (Andersen and Teicher, 2004; Yildirim tion or habituation to the stressors. This is illustrated by the habit-
et al., 2008). In addition to natural variations in performance of uation of adult rats do to repeated stressors (Girotti et al., 2006;
cognitive tasks due to age or developmental status (Altemus and Harris et al., 2004; Helmreich et al., 1997; Magarinos and McEwen,
Almli, 1997; Esmoris-Arranz et al., 2008; Schenk, 1985), acute 1995; Marti and Armario, 1997).
stressors experienced during puberty also contribute to disrup- The effect of pre-pubertal or pubertal stressors on learning and
tions and impairments in cognitive performances (Table 3) (Avital memory depends upon the specic neural development occurring
and Richter-Levin, 2005; Isgor et al., 2004; Tsoory et al., 2007; during the exposure of the stressors and on the particular type of
Tsoory and Richter-Levin, 2006). Variable physical, but not social, learning task. The use of predator odors are commonly used in fear
stress experienced from prior to the onset of puberty (P28) learning, which is mediated by the central and basolateral
into the late pubertal period (P56) in male rats results in amygdala and PFC (Farrell et al., 2013; Johansen et al., 2011), areas
impaired performance in the Morris water maze (a test of that continue to develop during the pubertal and adolescent
102 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Table 3
Experiments investigating the effects of stressors in puberty and adolescence on learning, memory, and cognition in adulthood.

Sex/strain/species Age at stress Stressor Age at test Test Result: Cognition Source
# SD rats 2856 Chronic variable stress 77 WM ; Isgor et al. (2004)
# SD rats 2729 Chronic variable stress 60 SA ; Tsoory and Richter-Levin (2006)
3335 Chronic variable stress 60 SA ;
# SD rats 2729 Chronic variable stress 6365 SA ; Tsoory et al. (2007)
# SD rats 3090 Chronic variable stress 90 SA Toth et al. (2008a)
# LE rats 2351 Chronic variable stress 72 SFCT / Pohl et al. (2007)
$ LE rats 2351 Chronic variable stress 72 SFCT /
# W rats 2628 Repeated elevated platform 60 WM ; Avital and Richter-Levin (2005)
28 Repeated elevated platform 90 WM ;
28, 90 Repeated elevated platform 90 "
2628, 60 Repeated elevated platform 90 WM "
# SD rats 2856 Social instability 77 WM Isgor et al. (2004)
# W rats 2830 Predator odor 83, 85, 87 Fear Conditioning Toledo-Rodriguez and Sandi (2007)
Extinction ;
$ W rats 2830 Predator odor 83.85.87 Fear Conditioning
Extinction

LE, Long Evans; SD, SpragueDawley; W, Wistar; SA, shuttle avoidance; SFCT, social food choice transmission; WM, water maze.

periods (Coulter et al., 1996; Cunningham et al., 2002; Farber et al., antibodies, the adaptive immune system provides stronger and
1995; Insel et al., 1990; Kalsbeek et al., 1988; Leslie et al., 1991). more rapid responses upon each exposure to a pathogen (Janeway,
Exposure to a predator odor during the pre-pubertal period, which 2001). Important for this review are the macrophages that phago-
does not affect corticosterone levels in adulthood (Toledo-Rodri- cytose the pathogens, produce cytokines, and recruit additional
guez and Sandi, 2007; Wright et al., 2008), also does not affect fear immune cells.
conditioning in either male or female adult rats (Toledo-Rodriguez
and Sandi, 2007). However, pre-pubertally-stressed male rats do 5.2. Immune signaling in the brain
not extinguish this fear conditioning (Toledo-Rodriguez and Sandi,
2007), suggesting abnormalities in PFC function. The PFC, which 5.2.1. The role of microglia in the healthy brain
continues to develop during the pubertal period (Coulter et al., The brain had been thought to be immune privileged and iso-
1996; Farber et al., 1995; Insel et al., 1990; Kalsbeek et al., 1988; lated from the immune system because of a lack of key immune
Leslie et al., 1991), also plays a critical role in extinction of fear proteins in neurons. Recently, immune molecules have been dem-
memories (Milad and Quirk, 2002; Santini et al., 2004). Therefore, onstrated to be present in the healthy brain and to play a critical
the experience of predator odor stress during puberty may disrupt role in brain development (Boulanger, 2009; Carpentier and Pal-
the ability of the PFC to suppress or extinguish fear learning. mer, 2009; Deverman and Patterson, 2009; Garay and McAllister,
2010).
Microglia are the primary immune cells in the brain. Microglia
5. Immunity and inammation
do not communicate by gap junctions, and they are not connected
like astrocytes (Eugenin et al., 2001). Rather, microglia maintain
During the last few decades, the importance of the intricate
fairly uniform distances in which their cell processes constantly
interactions of the nervous and immune systems has been realized,
scan the microenvironment in the healthy adult brain, surveying
especially with regard to the regulation of several physical and
for the presence of pathogens and monitoring the status of local
psychological pathologies, as will be discussed.
synapses (Hanisch and Kettenmann, 2007; Nimmerjahn et al.,
2005; Raivich, 2005; Tremblay et al., 2011; Wake et al., 2009). In
5.1. Immune system cases of either damage to the brain or presence of infectious
agents, the microglia migrate to the site of damage, up-regulate
The immune system is highly complex and has several special- immune molecules such as major histocompatibility complex class
ized components that protect an organism against disease. In order II (MHC II) receptor and complement receptors of cluster of differ-
to function properly, the immune system must recognize various entiation 11b (CD11b), and generate and maintain inammatory
types of pathogens as non-self and distinguish them from self responses (Graeber, 2010; Graeber and Streit, 2010; Streit et al.,
(for review Smith, 2012a,b). Vertebrates have both an innate and 1988, 1999, 2004).
an adaptive immune system. The innate immune system produces Microglia undergo substantial morphological restructuring in
rapid response to evolutionarily conserved pathogens, such as bac- response to free radicals or pro-inammatory cytokines (Graeber
terial, viral, fungal, or foreign proteins. Innate immune responses and Streit, 2010; Kettenmann et al., 2011; Ransohoff and Perry,
are not specic to the type of pathogen and consist of inamma- 2009; Streit et al., 1999). In the healthy brain, the microglia display
tory, phagocytic, and lytic responses to destroy the pathogens a highly ramied structure with a round or cell soma, from which
(for further review see Janeway and Medzhitov, 2002; Medzhitov emerge four to six primary branches (Hinwood et al., 2012). Fol-
and Janeway, 2000). The complement system is part of the innate lowing chronic stress, ramied microglia transition into hyper-
immune system; activation of complement proteases triggers a ramied microglia, with an increase in secondary but not primary
cascade of further cleavages, resulting in lysis of the target cell branching (Hinwood et al., 2012). Hyper-ramied microglia also
(for further information see Murphy, 2011). The other branch of reorient their processes in response to changes in neural activity
the immune system, the adaptive immune system, responds with (Tremblay et al., 2011). That is, in response to injury or inamma-
highly specialized cellular and highly targeted defenses to attack tory signals, microglia rst extend processes and reorient to the
and destroy a pathogen. The adaptive immune responses are based site of injury (Nimmerjahn et al., 2005), suggesting that the hy-
upon the ability of B and T-lymphocytes to recognize and remem- per-ramication may the rst step in the activation of microglia.
ber specic antigens found on the pathogens. Through the use of Next, the microglia transition into a reactive state, which is charac-
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 103

terized by shorter and thicker processes (Graeber and Streit, 2010; the rst regions populated by microglia (approximately embryonic
Kettenmann et al., 2011; Streit et al., 1999). Finally, the reactive day 17), whereas the PVN is not colonized until the early postnatal
microglia become phagocytic, amoeboid cells that have no pro- period (P4) (Schwarz et al., 2012). Developing microglia are largely
cesses and act like macrophages (Graeber and Streit, 2010; Streit in an activated or amoeboid state during the early postnatal period,
et al., 1999). which correlates with increases in cytokine levels. Moreover, envi-
Microglia are also involved in synapse maturation or elimina- ronmental factors, such as stressors, can increase the rate of colo-
tion via axonal guidance (Verney et al., 2010) and engulng synap- nization and the density of the microglia (Gomez-Gonzalez and
tic material (Graeber, 2010; Schafer et al., 2012; Stevens et al., Escobar, 2010). As microglia, particularly the amoeboid microglia,
2007; Tremblay et al., 2011). These processes are particularly play an important role in synaptic pruning through the clearing
important during synaptic maturation, when synaptic turnover is of dead and dying cells and processes, alterations in the coloniza-
high, or when synaptic pruning occurs (Paolicelli and Gross, tion or maturation rate could profoundly alter neural development
2011). This synaptic pruning by microglia is necessary for normal and subsequently adult brain and behavior. It has been postulated
brain development in the neonatal period. As pubertally-born that microglia may underlie the mechanisms by which insult in the
neurons and glia are functionally incorporated in neural circuits early postnatal period results in decits in cognition and disease
and contribute to the regulation of adult behaviors (Mohr and Sisk, states (Bilbo and Schwarz, 2009; Bilbo et al., 2011). Microglia are
2013), we speculate that microglia activity may play a critical role also excellent candidates for mediating the enduring effects of
in pubertal development as well. early insults due to their long life span and their ability to become
and remain activated (Town et al., 2005).
5.2.2. The Immune signals in development There is a sex difference in the time course of brain colonization
Cytokines are part of a large and diverse family of signaling by the microglia (Schwarz et al., 2012). Male rat pups in the early
molecules in the brain. They are typically described as either postnatal period (P4) have several-fold more microglia (especially
pro-inammatory, serving to promote inammation, and activated microglia) than female in the hippocampus, parietal cor-
anti-inammatory, limiting the effects of sustained or excessive tex and amygdala (Schwarz et al., 2012). Male rats are also partic-
inammatory reactions. The major pro-inammatory cytokines ularly sensitive to early infection-induced glial priming by which
responsible for the early responses to injury are also the most early life insults, challenges or injury lead to exaggerated re-
widely studied and include interleukin 1a (IL-1a), IL-1b, IL-6, and sponses to challenges in later life (Bilbo and Schwarz, 2009). That
tumor necrosis factor a (TNFa). The major anti-inammatory is, the morphology of the primed microglial cells resembles that
cytokines include IL-1 receptor antagonist, IL-4, IL-10, 1L-11, of the activated glial cells; however, primed microglia do not
IL-13, tumor growth factor b (TGFb) (Vilcek, 1998). Caution, chronically produce cytokines or other pro-inammatory media-
however, should be used in making generalizations about the tors (Perry et al., 2003). The primed microglia overreact to a subse-
pro- or anti- inammatory nature of the cytokines, as a given quent systemic challenge by overproduction of cytokines (Bilbo
cytokine can have both pro- and anti-inammatory actions (for et al., 2011; Perry et al., 2003). This overproduction of cytokines
review see Cavaillon, 2001). The action of a particular cytokine in turn may lead to the development of psychopathologies (Bilbo
depends upon the cytokine amount, timing and sequences of and Schwarz, 2009). Peri-pubertal female (P30) and adult (P60)
action, in addition to the nature of the target cell and of the activa- rats have more activated microglia than males of the same age in
tion signals. Further, the individual cytokines often act in concert the hippocampus, parietal cortex and amygdala, suggesting that
with other cytokines, resulting in synergistic or antagonistic effects females are more sensitive to immune dysregulation during
(reviewed in Hanisch, 2002; John et al., 2003). puberty/adolescence and early adulthood. We have recently
The major cytokines are expressed in the healthy brain (Bauer observed that female CD1 mice display more activated microglia
et al., 2007) and the expression patterns of cytokines and their in the pubertal period (P28 and P42) than in adulthood (P70;
receptors vary throughout development (Gadient and Otten, Holder and Blaustein, unpublished observations). Although
1994; Pousset, 1994; Takao et al., 1992). During development pubertal and adult mice show no apparent differences in sickness
cytokines play a role in neurogenesis, migration, differentiation, behavior in response to a bacterial endotoxin, the more activated
synapse formation, neural plasticity, and response to injury microglia in puberty may lead to enhanced cytokine and/or stress
(Boulanger, 2009; Carpentier and Palmer, 2009; Deverman and responses in the brain following LPS, but this has yet to be
Patterson, 2009; Garay and McAllister, 2010). Cytokines, such as empirically determined.
1L-1b and TNFa, modulate synaptic transmission. Whereas 1L-1b
inhibits excitatory synaptic transmission as measured by the 5.4. Working hypothesis of enduring effects of stress during puberty
frequency and/or amplitude of spontaneous excitatory postsynap-
tic currents (Yang et al., 2005), TNFa enhances inhibitory synaptic Neuroinammation refers to the chronic activation of microglia
transmission (Beattie et al., 2002; Stellwagen et al., 2005; and astrocytes (Aloisi, 2001; Kettenmann et al., 2011) and exagger-
Stellwagen and Malenka, 2006). Cytokines also are involved in ated expression of pro-inammatory mediators (e.g., cytokines,
synapse renement and plasticity in the adult cortex, hippocampus reactive oxygen species that can damage cellular structures) with-
and cerebellum (Boulanger, 2009). Studies examining the role of in the brain (Streit, 2010). A growing body of experimental and
cytokines in two models of activity-dependent plasticity, clinical research reveals a pivotal role of neuroinammation in
long-term plasticity (LTP) or long-term depression (LTD), indicate the etiology of mental disease, such as major depression (Capuron
that exogenous cytokines typically inhibit LTP and LTD (for further and Miller, 2011; Miller et al., 2009), anxiety (Hou and Baldwin,
review see Boulanger, 2009; Garay and McAllister, 2010). 2012; Jones and Thomsen, 2012), post-traumatic stress disorders
(Jones and Thomsen, 2012), and neurodegenerative diseases
5.3. Pubertal maturation of microglia associated with cognitive decline such as Alzheimers disease and
Parkinsons disease (Streit et al., 2004).
Microglia derive from primitive macrophages that originate in While boys are at higher risk for early-onset neurological
the extra-embryonic yolk sac (Ginhoux et al., 2010) and enter the disorders (e.g., autism spectrum disorders, attention decit
brain in around embryonic day 14 (Bilbo et al., 2011; Ling and hyperactivity disorder (ADHD)) and psychopathologies of brain
Wong, 1993). In rats, areas associated with cognition and emotion- organization (e.g., schizophrenia, Tourettes disorder), girls and
ality, such as the hippocampus, amygdala and cortex, are among women are more vulnerable to conditions that emerge during
104 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

puberty and adolescence (e.g., anxiety, depression, eating 1996; Turner and Lloyd, 2004), which often use the rather nebu-
disorders) (Taylor and Rutter, 1985). Additionally, adolescent girls lous terms of childhood and adolescence. As the pubertal status
who experience a high rate of both sexual and physical abuse also and hormonal milieu are critical for the etiology of affective disor-
show an increased rate of major depression (Silverman et al., ders (Hayward and Sanborn, 2002; Patton et al., 1996), there may
1996). Paralleling the ability of stressors to chronically activate be sensitive periods in girls during which stressors contribute to
microglia in adulthood leading to increased levels of cytokines the development of psychopathologies. However, these sensitive
(Bilbo and Schwarz, 2009), stressors experienced in childhood or periods remain to be precisely dened in girls.
adolescence induce short and long-term alterations in immune Another important matter to consider is that, rather than one
function (Danese et al., 2007, 2009; Lemieux et al., 2008) and vulnerable period, there may be several sensitive windows during
higher levels of the inammatory biomarker, C-reactive protein which a particular stressor is more or less effective in altering the
(CRP), in adulthood (Danese et al., 2007). It is, therefore, tempting adult brain and behavior. In support of this notion, shipping or im-
to speculate that the vulnerability of pubertal and/or adolescent mune challenge in six, but not four or eight week old mice disrupts
rats, mice, and humans may be due, in part, to the development ovarian hormone signaling adulthood (Section 3.4). Chronic vari-
of the neural immune systems and induction of enduring able stressors during the early pubertal period (P2830) are effec-
neuroinammation. While these ideas have yet to be empirically tive in altering the brain and behavior, but they are not effective at
tested, evidence to date is suggestive of the possibility of a later time point (Section 4.4). We suggest that the reasons for the
neuroinammation as a common pathway by which pubertal maximally effective periods for each stressor might depend upon
stressors may alter the adult brain and behavior, especially as it the nature of the stressor and on the particular developmental
relates to mental illness, such as anxiety and depression, as well changes occurring in the brain regions that mediate the response
as learning, memory and cognition (Bilbo et al., 2011; Cribbs to the stressor and the hormonal milieu. That is, the experience
et al., 2012; Miller et al., 2009; Salim et al., 2012). of the stressor may fundamentally alter the ongoing neural devel-
opmental processes occurring in key regions, such as the hippo-
campus and prefrontal cortex, leading to altered trajectories of
6. Discussion and conclusions the development of these regions. Hormones inuence many
developmental processes; therefore, it is even more important to
The pubertal period is a vulnerable period for the emergence of carefully consider the hormonal status and stage of pubertal devel-
mental diseases, due in part to the neural developments occurring opment at which the stressor is experienced, as the stressors may
during this time (Andersen, 2003; Kessler et al., 2005; Paus et al., be disrupting these dynamic processes.
2008). Moreover, exposure to extreme stressors during puberty Finally, the enduring effects of stressors on mental illness-rele-
and adolescence increases the risk of developing a mental disease vant behaviors in rodent models are accompanied by perturbations
(Ge et al., 2001; Grant et al., 2003, 2004; Silverman et al., 1996; of the hormones and hormonal axes that modulate behaviors. To-
Turner and Lloyd, 2004). Overall, the work discussed in this review gether, this review provides a novel focus on pubertal hormones as
demonstrates that particular stressors, specically complex stress- being responsible for the vulnerability to stressors, and that the
ors or immune challenges, experienced during puberty and/or enduring effects of stressors during puberty may be, in large part,
early-to-mid adolescence inuence behaviors relevant to mental due to aberrant responses to hormones. Aberrant responses to hor-
health and mental processes. Stressors during pubertal and/or ado- mones have been recognized for specic mental diseases (Bloch
lescent development lead to increases in hormonally inuenced et al., 2003; Schmidt et al., 1998); however, it is possible that dis-
anxiety-like and depression-like behaviors, a decrease sexual per- ordered responses contribute to the etiology of other mental ill-
formance, and a decrease in learning, memory, and cognitive per- nesses, such as depression. These possibilities need to be more
formance in adulthood (Ismail and Blaustein, 2013; Ismail et al., fully studied in clinical populations in order to further elucidate
2011, 2012; Laroche et al., 2009a,b; Olesen et al., 2011). Ovarian the neurobiological mechanisms of mental illness and provide
hormones, particularly estradiol, modulate these behaviors in fe- more targeted therapeutic treatments.
male mice, and the experience of a stressor can also lead a block-
ade or reversal of the effects of estradiol (Ismail and Blaustein,
Acknowledgments
2013; Ismail et al., 2011, 2012; Laroche et al., 2009a,b; Olesen
et al., 2011). Furthermore, stressors during pubertal and adolescent
The work from the authors laboratory was supported by Grants
development cause alterations in stress reactivity, steroid hormone
NS 19327, MH093854 from the National Institutes of Health, IOS
receptor densities, and synaptic plasticity of key brain regions such
1050179 from the National Science Foundation, and an Isis grant
as the hippocampus (see Tables 13). What remains to be demon-
from the Society for Womens Health Research. We thank the late
strated are the mechanisms by which acute or short-term stressors
Dr. Steven Zalcman for his suggestion that we consider the use of
experienced during puberty remodel the developing brain, behav-
an immune challenge as a pubertal stressor, and the members of
ior, and physiology.
the Stress, Gender, Drugs and the Brain Network for Womens Re-
One of the biggest challenges to precisely determining the
search for many thoughtful discussions in the early stages of the
neurobiological mechanisms and behavioral consequences of
work. We also thank Bethany Rappleyea, Amarylis Velez-Perez,
stressors is the lack of precision in the timing of stressors in many
John Hernandez, and Christina Gagliardi on their comments on a
previous experiments. Hormones modulate many of the ongoing
previous version of the manuscript.
neurodevelopmental processes of puberty and adolescence. Re-
cently, we have demonstrated that the ovary and, presumably,
ovarian hormones are required for an immune challenge in the References
pubertal period to decrease sexual behavior in adulthood. (B. Rap-
Aloisi, F., 2001. Immune function of microglia. Glia 36, 165179.
pleyea, N. Ismail, H. King, and J. Blaustein, unpublished observa-
Altemus, K.L., Almli, C.R., 1997. Neonatal hippocampal damage in rats: long-term
tions). Therefore, we suggest that it is more appropriate to spatial memory decits and associations with magnitude of hippocampal
consider the pubertal or reproductive stage of the animal, rather damage. Hippocampus 7, 403415.
than chronological age. This consideration is especially important Andersen, S.L., 2003. Trajectories of brain development: point of vulnerability or
window of opportunity? Neurosci. Biobehav. Rev. 27, 318.
in studies examining the effects of extreme stress and trauma in Andersen, S.L., Teicher, M.H., 2004. Delayed effects of early stress on hippocampal
girls (Ge et al., 2001; Grant et al., 2003, 2004; Silverman et al., development. Neuropsychopharmacology 29, 19881993.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 105

Andersen, S.L., Thompson, A.P., Krenzel, E., Teicher, M.H., 2002. Pubertal changes Capuron, L., Miller, A.H., 2011. Immune system to brain signaling:
in gonadal hormones do not underlie adolescent dopamine receptor neuropsychopharmacological implications. Pharmacol. Ther. 130, 226238.
overproduction. Psychoneuroendocrinology 27, 683691. Carey, M.P., Deterd, C.H., de Koning, J., Helmerhorst, F., de Kloet, E.R., 1995. The
Angold, A., Costello, E.J., 2006. Puberty and depression. Child Adolesc. Psychiatric inuence of ovarian steroids on hypothalamicpituitaryadrenal regulation in
Clin. North Am. 15, 919937, ix. the female rat. J. Endocrinol. 144, 311321.
Anisman, H., 2009. Cascading effects of stressors and inammatory immune system Carpentier, P.A., Palmer, T.D., 2009. Immune inuence on adult neural stem cell
activation: implications for major depressive disorder. J. Psychiatry Neurosci. regulation and function. Neuron 64, 7992.
34, 420. Cavaillon, J.M., 2001. Pro- versus anti-inammatory cytokines: myth or reality. Cell
Apter, D., Pakarinen, A., Hammond, G.L., Vihko, R., 1979. Adrenocortical function in Mol. Biol. (Noisy-le-grand) 47, 695702.
puberty serum ACTH, cortisol and dehydroepiandrosterone in girls and boys. Chester, J.A., Barrenha, G.D., Hughes, M.L., Keuneke, K.J., 2008. Age- and sex-
Acta Paediatr. Scand. 68, 599604. dependent effects of footshock stress on subsequent alcohol drinking and
Araki, S., Toran-Allerand, C.D., Ferin, M., Vande Wiele, R.L., 1975. Immunoreactive acoustic startle behavior in mice selectively bred for high-alcohol preference.
gonadotropin-releasing hormone (Gn-RH) during maturation in the rat: Alcohol. Clin. Exp. Res. 32, 17821794.
ontogeny of regional hypothalamic differences. Endocrinology 97, 693697. Choleris, E., Gustafsson, J.A., Korach, K.S., Muglia, L.J., Pfaff, D.W., Ogawa, S., 2003. An
Arnett, J.J., 1996. Sensation seeking, aggressiveness, and adolescent reckless estrogen-dependent four-gene micronet regulating social recognition: a study
behavior. Personality Individ. Differ. 20, 693702. with oxytocin and estrogen receptor-alpha and -beta knockout mice. Proc. Natl.
Astbury, J., 2001. Gender Disparities in Mental Health. Mental Health. Ministerial Acad. Sci. USA 100, 61926197.
Round Tables 2001, 54th World Health Assemble. WHO, Geneva, Switzerland, Choleris, E., Ogawa, S., Kavaliers, M., Gustafsson, J.A., Korach, K.S., Muglia, L.J., Pfaff,
2001, pp. 7392. D.W., 2006. Involvement of estrogen receptor alpha, beta and oxytocin in social
Avital, A., Richter-Levin, G., 2005. Exposure to juvenile stress exacerbates the discrimination: a detailed behavioral analysis with knockout female mice.
behavioural consequences of exposure to stress in the adult rat. Int. J. Genes Brain Behav. 5, 528539.
Neuropsychopharmacol. 8, 163173. Chung, W.C., De Vries, G.J., Swaab, D.F., 2002. Sexual differentiation of the bed
Avital, A., Ram, E., Maayan, R., Weizman, A., Richter-Levin, G., 2006. Effects of early- nucleus of the stria terminalis in humans may extend into adulthood. J.
life stress on behavior and neurosteroid levels in the rat hypothalamus and Neurosci. 22, 10271033.
entorhinal cortex. Brain Res. Bull. 68, 419424. Cohen, H., Kaplan, Z., Matar, M.A., Loewenthal, U., Zohar, J., Richter-Levin, G., 2007.
Barnea-Goraly, N., Menon, V., Eckert, M., Tamm, L., Bammer, R., Karchemskiy, A., Long-lasting behavioral effects of juvenile trauma in an animal model of PTSD
Dant, C.C., Reiss, A.L., 2005. White matter development during childhood and associated with a failure of the autonomic nervous system to recover. Eur.
adolescence: a cross-sectional diffusion tensor imaging study. Cereb. Cortex 15, Neuropsychopharmacol. 17, 464477.
18481854. Commins, D., Yahr, P., 1985. Autoradiographic localization of estrogen and
Bauer, S., Kerr, B.J., Patterson, P.H., 2007. The neuropoietic cytokine family in androgen receptors in the sexually dimorphic area and other regions of the
development, plasticity, disease and injury. Nat. Rev. Neurosci. 8, 221232. gerbil brain. J. Comp. Neurol. 231, 473489.
Bazak, N., Kozlovsky, N., Kaplan, Z., Matar, M., Golan, H., Zohar, J., Richter-Levin, G., Conrad, K.L., Winder, D.G., 2011. Altered anxiety-like behavior and long-term
Cohen, H., 2009. Pre-pubertal stress exposure affects adult behavioral response potentiation in the bed nucleus of the stria terminalis in adult mice exposed to
in association with changes in circulating corticosterone and brain-derived chronic social isolation, unpredictable stress, and ethanol beginning in
neurotrophic factor. Psychoneuroendocrinology 34, 844858. adolescence. Alcohol 45, 585593.
Beattie, E.C., Stellwagen, D., Morishita, W., Bresnahan, J.C., Ha, B.K., Von Zastrow, M., Coulter, C.L., Happe, H.K., Murrin, L.C., 1996. Postnatal development of the
Beattie, M.S., Malenka, R.C., 2002. Control of synaptic strength by glial dopamine transporter: a quantitative autoradiographic study. Brain Res. Dev.
TNFalpha. Science 295, 22822285. Brain Res. 92, 172181.
Bekku, N., Yoshimura, H., 2005. Animal model of menopausal depressive-like state Crawford, A.M., Pentz, M.A., Chou, C.P., Li, C., Dwyer, J.H., 2003. Parallel
in female mice: prolongation of immobility time in the forced swimming test developmental trajectories of sensation seeking and regular substance use in
following ovariectomy. Psychopharmacology 183, 300307. adolescents. Psychol. Addict. Behav. 17, 179192.
Bekman, N.M., Cummins, K., Brown, S.A., 2010. Affective and personality risk and Cribbs, D.H., Berchtold, N.C., Perreau, V., Coleman, P.D., Rogers, J., Tenner, A.J.,
cognitive mediators of initial adolescent alcohol use. J. Stud. Alcohol Drugs 71, Cotman, C.W., 2012. Extensive innate immune gene activation accompanies
570580. brain aging, increasing vulnerability to cognitive decline and
Beltran-Campos, V., Prado-Alcala, R.A., Leon-Jacinto, U., Aguilar-Vazquez, A., neurodegeneration: a microarray study. J. Neuroinamm. 9, 179.
Quirarte, G.L., Ramirez-Amaya, V., Diaz-Cintra, S., 2011. Increase of mushroom Cunningham, M.G., Bhattacharyya, S., Benes, F.M., 2002. Amygdalo-cortical
spine density in CA1 apical dendrites produced by water maze training is sprouting continues into early adulthood: implications for the development
prevented by ovariectomy. Brain Res. 1369, 119130. of normal and abnormal function during adolescence. J. Comp. Neurol. 453,
Benes, F.M., Turtle, M., Khan, Y., Farol, P., 1994. Myelination of a key relay zone in 116130.
the hippocampal formation occurs in the human brain during childhood, Dahl, R.E., 2004. Adolescent brain development: a period of vulnerabilities and
adolescence, and adulthood. Arch. Gen. Psychiatry 51, 477484. opportunities. Keynote address. Ann. N. Y. Acad. Sci. 1021, 122.
Benes, F.M., Taylor, J.B., Cunningham, M.C., 2000. Convergence and plasticity of Dahl, R.E., Gunnar, M.R., 2009. Heightened stress responsiveness and emotional
monoaminergic systems in the medial prefrontal cortex during the postnatal reactivity during pubertal maturation: implications for psychopathology. Dev.
period: implications for the development of psychopathology. Cereb. Cortex 10, Psychopathol. 21, 16.
10141027. Dalla, C., Antoniou, K., Papadopoulou-Daifoti, Z., Balthazart, J., Bakker, J., 2004.
Bernardi, M., Vergoni, A.V., Sandrini, M., Tagliavini, S., Bertolini, A., 1989. Inuence Oestrogen-decient female aromatase knockout (ArKO) mice exhibit
of ovariectomy, estradiol and progesterone on the behavior of mice in an depressive-like symptomatology. Eur. J. Neurosci. 20, 217228.
experimental model of depression. Physiol. Behav. 45, 10671068. Danese, A., Pariante, C.M., Caspi, A., Taylor, A., Poulton, R., 2007. Childhood
Bilbo, S.D., Schwarz, J.M., 2009. Early-life programming of later-life brain and maltreatment predicts adult inammation in a life-course study. Proc. Natl.
behavior: a critical role for the immune system. Front. Behav. Neurosci. 3, 14. Acad. Sci. USA 104, 13191324.
Bilbo, S.D., Smith, S.H., Schwarz, J.M., 2011. A lifespan approach to Danese, A., Moftt, T.E., Harrington, H., Milne, B.J., Polanczyk, G., Pariante, C.M.,
neuroinammatory and cognitive disorders: a critical role for glia. J. Poulton, R., Caspi, A., 2009. Adverse childhood experiences and adult risk factors
Neuroimmune Pharmacol.. for age-related disease: depression, inammation, and clustering of metabolic
Blaustein, J.D., 2008. Neuroendocrine regulation of feminine sexual behavior: risk markers. Arch. Pediatr. Adolesc. Med. 163, 11351143.
lessons from rodent models and thoughts about humans. Ann. Rev. Psychol. 59, Daniel, J.M., Fader, A.J., Spencer, A.L., Dohanich, G.P., 1997. Estrogen enhances
93118. performance of female rats during acquisition of a radial arm maze. Horm.
Blaustein, J.D., Ismail, N., 2013. Enduring inuence of pubertal stressors on Behav. 32, 217225.
behavioral response to hormones in female mice. Horm. Behav. 64, 390398. Dantzer, R., 2001. Cytokine-induced sickness behavior: mechanisms and
Bloch, M., Daly, R.C., Rubinow, D.R., 2003. Endocrine factors in the etiology of implications. Ann. N. Y. Acad. Sci. 933, 222234.
postpartum depression. Compr. Psychiatry 44, 234246. Datson, N.A., Morsink, M.C., Meijer, O.C., de Kloet, E.R., 2008. Central corticosteroid
Boulanger, L.M., 2009. Immune proteins in brain development and synaptic actions: search for gene targets. Eur. J. Pharmacol. 583, 272289.
plasticity. Neuron 64, 93109. Davis, E.C., Shryne, J.E., Gorski, R.A., 1996. Structural sexual dimorphisms in the
Bourgeois, J.P., Rakic, P., 1993. Changes of synaptic density in the primary visual cortex of anteroventral periventricular nucleus of the rat hypothalamus are sensitive to
the macaque monkey from fetal to adult stage. J. Neurosci. 13, 28012820. gonadal steroids perinatally, but develop peripubertally. Neuroendocrinology
Bourgeois, J.P., Goldman-Rakic, P.S., Rakic, P., 1994. Synaptogenesis in the prefrontal 63, 142148.
cortex of rhesus monkeys. Cereb. Cortex 4, 7896. Davis, D.M., Jacobson, T.K., Aliakbari, S., Mizumori, S.J., 2005. Differential effects of
Bourke, C.H., Neigh, G.N., 2011. Behavioral effects of chronic adolescent stress are estrogen on hippocampal- and striatal-dependent learning. Neurobiol. Learn.
sustained and sexually dimorphic. Horm. Behav. 60, 112120. Mem. 84, 132137.
Brandi, L.S., Santoro, D., Natali, A., Altomonte, F., Baldi, S., Frascerra, S., Ferrannini, E., De Kloet, E.R., de Jong, I.E., Oitzi, M.S., 2008. Neuropharmacology of glucocorticoids:
1993. Insulin resistance of stress: sites and mechanisms. Clin. Sci. (Lond.) 85, focus on emotion, cognition and cocaine. Eur. J. Pharmacol. 585, 473482.
525535. Deverman, B.E., Patterson, P.H., 2009. Cytokines and CNS development. Neuron 64,
Broadbent, N.J., Squire, L.R., Clark, R.E., 2004. Spatial memory, recognition memory, 6178.
and the hippocampus. Proc. Natl. Acad. Sci. USA 101, 1451514520. Diaz-Veliz, G., Soto, V., Dussaubat, N., Mora, S., 1989. Inuence of the estrous cycle,
Brown, T.J., Hochberg, R.B., Zielinski, J.E., MacLusky, N.J., 1988. Regional sex ovariectomy and estradiol replacement upon the acquisition of conditioned
differences in cell nuclear estrogen-binding capacity in the rat hypothalamus avoidance responses in rats. Physiol. Behav. 46, 397401.
and preoptic area. Endocrinology 123, 17611770.
106 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Diaz-Veliz, G., Urresta, F., Dussaubat, N., Mora, S., 1991. Effects of estradiol Green, M.R., Barnes, B., McCormick, C.M., 2012. Social instability stress in
replacement in ovariectomized rats on conditioned avoidance responses and adolescence increases anxiety and reduces social interactions in adulthood in
other behaviors. Physiol. Behav. 50, 6165. male Long-Evans rats. Dev. Psychobiol..
Diaz-Veliz, G., Alarcon, T., Espinoza, C., Dussaubat, N., Mora, S., 1997. Ketanserin and Grippo, A.J., Beltz, T.G., Johnson, A.K., 2003. Behavioral and cardiovascular changes
anxiety levels: inuence of gender, estrous cycle, ovariectomy and ovarian in the chronic mild stress model of depression. Physiol. Behav. 78, 703710.
hormones in female rats. Pharmacol. Biochem. Behav. 58, 637642. Guillamon, A., de Blas, M.R., Segovia, S., 1988. Effects of sex steroids on the
Doremus-Fitzwater, T.L., Varlinskaya, E.I., Spear, L.P., 2009. Social and non-social development of the locus coeruleus in the rat. Brain Res. 468, 306310.
anxiety in adolescent and adult rats after repeated restraint. Physiol. Behav. 97, Handa, R.J., Nunley, K.M., Lorens, S.A., Louie, J.P., McGivern, R.F., Bollnow, M.R., 1994.
484494. Androgen regulation of adrenocorticotropin and corticosterone secretion in the
Eaton, N.R., Keyes, K.M., Krueger, R.F., Balsis, S., Skodol, A.E., Markon, K.E., Grant, B.F., male rat following novelty and foot shock stressors. Physiol. Behav. 55, 117
Hasin, D.S., 2012. An invariant dimensional liability model of gender differences 124.
in mental disorder prevalence: evidence from a national sample. J. Abnorm. Hanisch, U.K., 2002. Microglia as a source and target of cytokines. Glia 40, 140155.
Psychol. 121, 282288. Hanisch, U.K., Kettenmann, H., 2007. Microglia: active sensor and versatile effector
Ellison, P.T., Reiches, M.W., Shattuck-Faegre, H., Breakey, A., Konecna, M., Urlacher, cells in the normal and pathologic brain. Nat. Neurosci. 10, 13871394.
S., Wobber, V., 2012. Puberty as a life history transition. Ann. Hum. Biol. 39, Harris, R.B., Gu, H., Mitchell, T.D., Endale, L., Russo, M., Ryan, D.H., 2004. Increased
352360. glucocorticoid response to a novel stress in rats that have been restrained.
Esmoris-Arranz, F.J., Mendez, C., Spear, N.E., 2008. Contextual fear conditioning Physiol. Behav. 81, 557568.
differs for infant, adolescent, and adult rats. Behav. Processes 78, 340350. Hayward, C., Sanborn, K., 2002. Puberty and the emergence of gender differences in
Eugenin, E.A., Eckardt, D., Theis, M., Willecke, K., Bennett, M.V., Saez, J.C., 2001. psychopathology. J. Adolesc. Health 30, 4958.
Microglia at brain stab wounds express connexin 43 and in vitro form Helmreich, D.L., Morano, M.I., Akil, H., Watson, S.J., 1997. Correlation between
functional gap junctions after treatment with interferon-gamma and tumor changes in stress-induced corticosterone secretion and GR mRNA levels. Stress
necrosis factor-alpha. Proc. Natl. Acad. Sci. USA 98, 41904195. 2, 101112.
Farber, N.B., Wozniak, D.F., Price, M.T., Labruyere, J., Huss, J., St Peter, H., Olney, J.W., Herbison, A.E., 1995. Sexually dimorphic expression of androgen receptor
1995. Age-specic neurotoxicity in the rat associated with NMDA receptor immunoreactivity by somatostatin neurones in rat hypothalamic
blockade: potential relevance to schizophrenia? Biol. Psychiatry 38, 788796. periventricular nucleus and bed nucleus of the stria terminalis. J.
Farrell, M.R., Sengelaub, D.R., Wellman, C.L., 2013. Sex differences and chronic stress Neuroendocrinol. 7, 543553.
effects on the neural circuitry underlying fear conditioning and extinction. Hinwood, M., Tynan, R.J., Charnley, J.L., Beynon, S.B., Day, T.A., Walker, F.R., 2012.
Physiol. Behav.. Chronic stress induced remodeling of the prefrontal cortex: structural re-
Feder, H.H., Silver, R., 1974. Activation of lordosis in ovariectomized guinea pigs by organization of microglia and the inhibitory effect of minocycline. Cereb.
free and esteried forms of estrone, estradiol-17 beta and estriol. Physiol. Cortex.
Behav. 13, 251255. Hou, R., Baldwin, D.S., 2012. A neuroimmunological perspective on anxiety
Filardo, E.J., Thomas, P., 2012. Minireview: G protein-coupled estrogen receptor-1, disorders. Hum. Psychopharmacol. 27, 614.
GPER-1: its mechanism of action and role in female reproductive cancer, renal Huang, G.Z., Woolley, C.S., 2012. Estradiol acutely suppresses inhibition in the
and vascular physiology. Endocrinology 153, 29532962. hippocampus through a sex-specic endocannabinoid and mGluR-dependent
File, S.E., Seth, P., 2003. A review of 25 years of the social interaction test. Eur. J. mechanism. Neuron 74, 801808.
Pharmacol. 463, 3553. Huttenlocher, P.R., 1979. Synaptic density in human frontal cortex developmental
Gadient, R.A., Otten, U., 1994. Expression of interleukin-6 (IL-6) and interleukin-6 changes and effects of aging. Brain Res. 163, 195205.
receptor (IL-6R) mRNAs in rat brain during postnatal development. Brain Res. Huttenlocher, P.R., Dabholkar, A.S., 1997. Regional differences in synaptogenesis in
637, 1014. human cerebral cortex. J. Comp. Neurol. 387, 167178.
Galeeva, A., Tuohimaa, P., 2001. Analysis of mouse plus-maze behavior modulated Ilin, Y., Richter-Levin, G., 2009. Enriched environment experience overcomes
by ovarian steroids. Behav. Brain Res. 119, 4147. learning decits and depressive-like behavior induced by juvenile stress. PLoS
Garay, P.A., McAllister, A.K., 2010. Novel roles for immune molecules in neural ONE 4, e4329.
development: implications for neurodevelopmental disorders. Front. Synaptic Im-Bolter, N., Cohen, N.J., Farnia, F., 2013. I thought we were good: social cognition,
Neurosci. 2, 136. gurative language, and adolescent psychopathology. J. Child Psychol.
Ge, X., Conger, R.D., Elder Jr., G.H., 2001. Pubertal transition, stressful life events, and Psychiatry.
the emergence of gender differences in adolescent depressive symptoms. Dev. Imwalle, D.B., Gustafsson, J.A., Rissman, E.F., 2005. Lack of functional estrogen
Psychol. 37, 404417. receptor beta inuences anxiety behavior and serotonin content in female mice.
Gerocs, K., Rethelyi, M., Halasz, B., 1986. Quantitative analysis of dendritic Physiol. Behav. 84, 157163.
protrusions in the medial preoptic area during postnatal development. Brain Insel, T.R., Miller, L.P., Gelhard, R.E., 1990. The ontogeny of excitatory amino acid
Res. 391, 4957. receptors in rat forebrain I. N-methyl-D-aspartate and quisqualate receptors.
Giedd, J.N., Castellanos, F.X., Rajapakse, J.C., Vaituzis, A.C., Rapoport, J.L., 1997. Neuroscience 35, 3143.
Sexual dimorphism of the developing human brain. Prog. Intlekofer, K.A., Petersen, S.L., 2010. Distribution of mRNAs encoding classical
Neuropsychopharmacol. Biol. Psychiatry 21, 11851201. progestin receptor, progesterone membrane components 1 and 2, serpine
Giedd, J.N., Blumenthal, J., Jeffries, N.O., Castellanos, F.X., Liu, H., Zijdenbos, A., Paus, mRNA binding protein 1, and progestin and ADIPOQ receptor family members 7
T., Evans, A.C., Rapoport, J.L., 1999. Brain development during childhood and and 8 in rat forebrain. Neuroscience 172, 5565.
adolescence: a longitudinal MRI study. Nat. Neurosci. 2, 861863. Isgor, C., Kabbaj, M., Akil, H., Watson, S.J., 2004. Delayed effects of chronic variable
Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler, M.F., stress during peripubertal-juvenile period on hippocampal morphology and on
Conway, S.J., Ng, L.G., Stanley, E.R., Samokhvalov, I.M., Merad, M., 2010. Fate cognitive and stress axis functions in rats. Hippocampus 14, 636648.
mapping analysis reveals that adult microglia derive from primitive Ismail, N., Blaustein, J.D., 2013. Pubertal immune challenge blocks the ability of
macrophages. Science 330, 841845. estradiol to enhance performance on cognitive tasks in adult female mice.
Girotti, M., Pace, T.W., Gaylord, R.I., Rubin, B.A., Herman, J.P., Spencer, R.L., 2006. Psychoneuroendocrinology 38, 11701177.
Habituation to repeated restraint stress is associated with lack of stress-induced Ismail, N., Garas, P., Blaustein, J.D., 2011. Long-term effects of pubertal stressors on
c-fos expression in primary sensory processing areas of the rat brain. female sexual receptivity and estrogen receptor-alpha expression in CD-1
Neuroscience 138, 10671081. female mice. Horm. Behav. 59, 565571.
Goldstein, L.A., Kurz, E.M., Sengelaub, D.R., 1990. Androgen regulation of dendritic Ismail, N., Kumlin, A.M., Blaustein, J.D., 2012. A pubertal immune challenge alters
growth and retraction in the development of a sexually dimorphic spinal the antidepressant-like effects of chronic estradiol treatment in inbred and
nucleus. J. Neurosci. 10, 935946. outbred adult female mice. Neuroscience.
Gomez, F., Manalo, S., Dallman, M.F., 2004. Androgen-sensitive changes in Jacobson-Pick, S., Richter-Levin, G., 2010. Differential impact of juvenile stress and
regulation of restraint-induced adrenocorticotropin secretion between early corticosterone in juvenility and in adulthood, in male and female rats. Behav.
and late puberty in male rats. Endocrinology 145, 5970. Brain Res. 214, 268276.
Gomez-Gonzalez, B., Escobar, A., 2010. Prenatal stress alters microglial Jacobson-Pick, S., Richter-Levin, G., 2012. Short- and long-term effects of juvenile
development and distribution in postnatal rat brain. Acta Neuropathol. 119, stressor exposure on the expression of GABAA receptor subunits in rats. Stress
303315. 15, 416424.
Gottsch, M.L., Cunningham, M.J., Smith, J.T., Popa, S.M., Acohido, B.V., Crowley, W.F., Jakubowski, M., Blum, M., Roberts, J.L., 1991. Postnatal development of
Seminara, S., Clifton, D.K., Steiner, R.A., 2004. A role for kisspeptins in the gonadotropin-releasing hormone and cyclophilin gene expression in the
regulation of gonadotropin secretion in the mouse. Endocrinology 145, 4073 female and male rat brain. Endocrinology 128, 27022708.
4077. Janeway Jr., C.A., 2001. How the immune system works to protect the host from
Graeber, M.B., 2010. Changing face of microglia. Science 330, 783788. infection: a personal view. Proc. Natl. Acad. Sci. USA 98, 74617468.
Graeber, M.B., Streit, W.J., 2010. Microglia: biology and pathology. Acta Janeway Jr., C.A., Medzhitov, R., 2002. Innate immune recognition. Annu. Rev.
Neuropathol. 119, 89105. Immunol. 20, 197216.
Grant, K.E., Compas, B.E., Stuhlmacher, A.F., Thurm, A.E., McMahon, S.D., Halpert, Jedlicka, P., Vlachos, A., Schwarzacher, S.W., Deller, T., 2008. A role for the spine
J.A., 2003. Stressors and child and adolescent psychopathology: moving from apparatus in LTP and spatial learning. Behav. Brain Res. 192, 1219.
markers to mechanisms of risk. Psychol. Bull. 129, 447466. Johansen, J.P., Cain, C.K., Ostroff, L.E., LeDoux, J.E., 2011. Molecular mechanisms of
Grant, K.E., Compas, B.E., Thurm, A.E., McMahon, S.D., Gipson, P.Y., 2004. Stressors fear learning and memory. Cell 147, 509524.
and child and adolescent psychopathology: measurement issues and John, G.R., Lee, S.C., Brosnan, C.F., 2003. Cytokines: powerful regulators of glial cell
prospective effects. J. Clin. Child Adolesc. Psychol. 33, 412425. activation. Neuroscientist 9, 1022.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 107

Johnston, A.L., File, S.E., 1991. Sex differences in animal tests of anxiety. Physiol. Magarinos, A.M., McEwen, B.S., 1995. Stress-induced atrophy of apical dendrites of
Behav. 49, 245250. hippocampal CA3c neurons: involvement of glucocorticoid secretion and
Jones, K.A., Thomsen, C., 2012. The role of the innate immune system in psychiatric excitatory amino acid receptors. Neuroscience 69, 8998.
disorders. Mol. Cell. Neurosci.. Mangelsdorf, D.J., Thummel, C., Beato, M., Herrlich, P., Schutz, G., Umesono, K.,
Kalsbeek, A., Voorn, P., Buijs, R.M., Pool, C.W., Uylings, H.B., 1988. Development of Blumberg, B., Kastner, P., Mark, M., Chambon, P., Evans, R.M., 1995. The nuclear
the dopaminergic innervation in the prefrontal cortex of the rat. J. Comp. receptor superfamily: the second decade. Cell 83, 835839.
Neurol. 269, 5872. Marcondes, F.K., Miguel, K.J., Melo, L.L., Spadari-Bratsch, R.C., 2001. Estrous cycle
Kauffman, A.S., Clifton, D.K., Steiner, R.A., 2007. Emerging ideas about kisspeptin inuences the response of female rats in the elevated plus-maze test. Physiol.
GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Behav. 74, 435440.
Neurosci. 30, 504511. Marshall, W.A., Tanner, J.M., 1969. Variations in pattern of pubertal changes in girls.
Kendig, M.D., Bowen, M.T., Kemp, A.H., McGregor, I.S., 2011. Predatory threat Arch. Dis. Child. 44, 291303.
induces huddling in adolescent rats and residual changes in early adulthood Marshall, W.A., Tanner, J.M., 1970. Variations in the pattern of pubertal changes in
suggestive of increased resilience. Behav. Brain Res. 225, 405414. boys. Arch. Dis. Child. 45, 1323.
Kessler, R.C., Berglund, P., Demler, O., Jin, R., Merikangas, K.R., Walters, E.E., 2005. Marti, O., Armario, A., 1997. Inuence of regularity of exposure to chronic stress on
Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the the pattern of habituation of pituitaryadrenal hormones, prolactin and
National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62, 593602. glucose. Stress 1, 179189.
Kettenmann, H., Hanisch, U.K., Noda, M., Verkhratsky, A., 2011. Physiology of Maslova, L.N., Bulygina, V.V., Markel, A.L., 2002. Chronic stress during prepubertal
microglia. Physiol. Rev. 91, 461553. development: immediate and long-lasting effects on arterial blood pressure and
Konkle, A.T., Baker, S.L., Kentner, A.C., Barbagallo, L.S., Merali, Z., Bielajew, C., 2003. anxiety-related behavior. Psychoneuroendocrinology 27, 549561.
Evaluation of the effects of chronic mild stressors on hedonic and physiological Mathews, I.Z., Wilton, A., Styles, A., McCormick, C.M., 2008. Increased depressive
responses: sex and strain compared. Brain Res. 992, 227238. behaviour in females and heightened corticosterone release in males to swim
Korenbrot, C.C., Huhtaniemi, I.T., Weiner, R.I., 1977. Preputial separation as an stress after adolescent social stress in rats. Behav. Brain Res. 190, 3340.
external sign of pubertal development in the male rat. Biol. Reprod. 17, 298 McCarthy, M.M., 2010. How its made: organisational effects of hormones on the
303. developing brain. J. Neuroendocrinol. 22, 736742.
Korol, D.L., 2004. Role of estrogen in balancing contributions from multiple memory McCormick, C.M., Green, M.R., 2012. From the stressed adolescent to the anxious
systems. Neurobiol. Learn. Mem. 82, 309323. and depressed adult: investigations in rodent models. Neuroscience.
Koss, W.A., Einat, H., Schloesser, R.J., Manji, H.K., Rubinow, D.R., 2012. Estrogen McCormick, C.M., Furey, B.F., Child, M., Sawyer, M.J., Donohue, S.M., 1998. Neonatal
effects on the forced swim test differ in two outbred rat strains. Physiol. Behav. sex hormones have organizational effects on the hypothalamicpituitary
106, 8186. adrenal axis of male rats. Brain Res. Dev. Brain Res. 105, 295307.
Krezel, W., Dupont, S., Krust, A., Chambon, P., Chapman, P.F., 2001. Increased anxiety McCormick, C.M., Linkroum, W., Sallinen, B.J., Miller, N.W., 2002. Peripheral and
and synaptic plasticity in estrogen receptor beta -decient mice. Proc. Natl. central sex steroids have differential effects on the HPA axis of male and female
Acad. Sci. USA 98, 1227812282. rats. Stress 5, 235247.
Kuhnemann, S., Brown, T.J., Hochberg, R.B., MacLusky, N.J., 1994. Sex differences in McCormick, C.M., Robarts, D., Kopeikina, K., Kelsey, J.E., 2005. Long-lasting, sex- and
the development of estrogen receptors in the rat brain. Horm. Behav. 28, 483 age-specic effects of social stressors on corticosterone responses to restraint
491. and on locomotor responses to psychostimulants in rats. Horm. Behav. 48, 64
Kvetnansky, R., Sabban, E.L., Palkovits, M., 2009. Catecholaminergic systems in 74.
stress: structural and molecular genetic approaches. Physiol. Rev. 89, 535606. McCormick, C.M., Smith, C., Mathews, I.Z., 2008. Effects of chronic social stress in
Laroche, J., Gasbarro, L., Herman, J.P., Blaustein, J.D., 2009a. Enduring inuences of adolescence on anxiety and neuroendocrine response to mild stress in male and
peripubertal/adolescent stressors on behavioral response to estradiol and female rats. Behav. Brain Res. 187, 228238.
progesterone in adult female mice. Endocrinology 150, 37173725. McCormick, C.M., Green, M.R., Cameron, N.M., Nixon, F., Levy, M.J., Clark, R.A., 2013.
Laroche, J., Gasbarro, L., Herman, J.P., Blaustein, J.D., 2009b. Reduced behavioral Decits in male sexual behavior in adulthood after social instability stress in
response to gonadal hormones in mice shipped during the peripubertal/ adolescence in rats. Horm. Behav. 63, 512.
adolescent period. Endocrinology 150, 23512358. McEwen, B.S., 2000. Allostasis, allostatic load, and the aging nervous system: role of
Laugero, K.D., Moberg, G.P., 2000. Summation of behavioral and immunological excitatory amino acids and excitotoxicity. Neurochem. Res. 25, 12191231.
stress: metabolic consequences to the growing mouse. Am. J. Physiol. McEwen, B.S., Akama, K.T., Spencer-Segal, J.L., Milner, T.A., Waters, E.M., 2012.
Endocrinol. Metab. 279, E44E49. Estrogen effects on the brain: actions beyond the hypothalamus via novel
Lemieux, A., Coe, C.L., Carnes, M., 2008. Symptom severity predicts degree of T cell mechanisms. Behav. Neurosci. 126, 416.
activation in adult women following childhood maltreatment. Brain Behav. Medzhitov, R., Janeway Jr., C., 2000. Innate immune recognition: mechanisms and
Immun. 22, 9941003. pathways. Immunol. Rev. 173, 8997.
Lenz, K.M., Nugent, B.M., McCarthy, M.M., 2012. Sexual differentiation of the rodent Meyer, G., Ferres-Torres, R., Mas, M., 1978. The effects of puberty and castration on
brain: dogma and beyond. Front. Neurosci. 6, 26. hippocampal dendritic spines of mice. A Golgi study. Brain Res. 155, 108112.
Leslie, C.A., Robertson, M.W., Cutler, A.J., Bennett Jr., J.P., 1991. Postnatal Milad, M.R., Quirk, G.J., 2002. Neurons in medial prefrontal cortex signal memory
development of D1 dopamine receptors in the medial prefrontal cortex, for fear extinction. Nature 420, 7074.
striatum and nucleus accumbens of normal and neonatal 6-hydroxydopamine Miller, A.H., Maletic, V., Raison, C.L., 2009. Inammation and its discontents: the
treated rats: a quantitative autoradiographic analysis. Brain Res. Dev. Brain Res. role of cytokines in the pathophysiology of major depression. Biol. Psychiatry
62, 109114. 65, 732741.
Leuner, B., Falduto, J., Shors, T.J., 2003. Associative memory formation increases the Mohr, M.A., Sisk, C.L., 2013. Pubertally born neurons and glia are functionally
observation of dendritic spines in the hippocampus. J. Neurosci. 23, 659665. integrated into limbic and hypothalamic circuits of the male Syrian hamster.
Lewis, D.A., Cruz, D., Eggan, S., Erickson, S., 2004. Postnatal development of Proc. Natl. Acad. Sci. USA 110, 47924797.
prefrontal inhibitory circuits and the pathophysiology of cognitive dysfunction Mora, S., Dussaubat, N., Diaz-Veliz, G., 1996. Effects of the estrous cycle and ovarian
in schizophrenia. Ann. N. Y. Acad. Sci. 1021, 6476. hormones on behavioral indices of anxiety in female rats.
Li, C., Brake, W.G., Romeo, R.D., Dunlop, J.C., Gordon, M., Buzescu, R., Magarinos, Psychoneuroendocrinology 21, 609620.
A.M., Allen, P.B., Greengard, P., Luine, V., McEwen, B.S., 2004. Estrogen alters Morgan, M.A., Pfaff, D.W., 2001. Effects of estrogen on activity and fear-related
hippocampal dendritic spine shape and enhances synaptic protein behaviors in mice. Horm. Behav. 40, 472482.
immunoreactivity and spatial memory in female mice. Proc. Natl. Acad. Sci. Morgan, M.A., Pfaff, D.W., 2002. Estrogens effects on activity, anxiety, and fear in
USA 101, 21852190. two mouse strains. Behav. Brain Res. 132, 8593.
Li, X., Morrow, D., Witkin, J.M., 2006. Decreases in nestlet shredding of mice by Murphy, K., 2011. Janeways Immunobiology. Garland Science, New York and
serotonin uptake inhibitors: comparison with marble burying. Life Sci. 78, London.
19331939. Navarro, V.M., Castellano, J.M., McConkey, S.M., Pineda, R., Ruiz-Pino, F., Pinilla, L.,
Ling, E.A., Wong, W.C., 1993. The origin and nature of ramied and amoeboid Clifton, D.K., Tena-Sempere, M., Steiner, R.A., 2011. Interactions between
microglia: a historical review and current concepts. Glia 7, 918. kisspeptin and neurokinin B in the control of GnRH secretion in the female
Lohmiller, J., Sonya, P., 2006. Reproduction and Breeding. Elsevier, San Diego. rat. Am. J. Physiol. Endocrinol. Metab. 300, E202E210.
Lu, S.F., McKenna, S.E., Cologer-Clifford, A., Nau, E.A., Simon, N.G., 1998. Androgen Nilsson, S., Makela, S., Treuter, E., Tujague, M., Thomsen, J., Andersson, G., Enmark,
receptor in mouse brain: sex differences and similarities in autoregulation. E., Pettersson, K., Warner, M., Gustafsson, J.A., 2001. Mechanisms of estrogen
Endocrinology 139, 15941601. action. Physiol. Rev. 81, 15351565.
Luine, V.N., 2008. Sex steroids and cognitive function. J. Neuroendocrinol. 20, 866 Nimmerjahn, A., Kirchhoff, F., Helmchen, F., 2005. Resting microglial cells are
872. highly dynamic surveillants of brain parenchyma in vivo. Science 308,
Luine, V.N., Frankfurt, M., 2012. Estrogens facilitate memory processing through 13141318.
membrane mediated mechanisms and alterations in spine density. Front. Novac, N., Heinzel, T., 2004. Nuclear receptors: overview and classication. Curr.
Neuroendocrinol. 33, 388402. Drug Targets Inamm. Allergy 3, 335346.
Luine, V.N., Richards, S.T., Wu, V.Y., Beck, K.D., 1998. Estradiol enhances learning Nunez, J.L., Jurgens, H.A., Juraska, J.M., 2000. Androgens reduce cell death in the
and memory in a spatial memory task and effects levels of monoaminergic developing rat visual cortex. Brain Res. Dev. Brain Res. 125, 8388.
neurotransmitters. Horm. Behav. 34, 149162. Nunez, J.L., Lauschke, D.M., Juraska, J.M., 2001. Cell death in the development of the
MacPherson, L., Magidson, J.F., Reynolds, E.K., Kahler, C.W., Lejuez, C.W., 2010. posterior cortex in male and female rats. J. Comp. Neurol. 436, 3241.
Changes in sensation seeking and risk-taking propensity predict increases in Oakley, A.E., Clifton, D.K., Steiner, R.A., 2009. Kisspeptin signaling in the brain.
alcohol use among early adolescents. Alcohol. Clin. Exp. Res. 34, 14001408. Endocr. Rev. 30, 713743.
108 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Ogawa, S., Eng, V., Taylor, J., Lubahn, D.B., Korach, K.S., Pfaff, D.W., 1998. Roles of Rachman, I.M., Unnerstall, J.R., Pfaff, D.W., Cohen, R.S., 1998. Estrogen alters
estrogen receptor-alpha gene expression in reproduction-related behaviors in behavior and forebrain c-fos expression in ovariectomized rats subjected to the
female mice. Endocrinology 139, 50705081. forced swim test. Proc. Natl. Acad. Sci. USA 95, 1394113946.
Ojeda, S.R., Urbanski, H.F., 1994. Puberty in the rat. In: Knobil, E., Neill, J. (Eds.), The Rainbow, T.C., Parsons, B., McEwen, B.S., 1982. Sex differences in rat brain oestrogen
Physiology of Reproduction. Raven Press, New York, pp. 363409. and progestin receptors. Nature 300, 648649.
Ojeda, S.R., Wheaton, J.E., Jameson, H.E., McCann, S.M., 1976. The onset of puberty in Raivich, G., 2005. Like cops on the beat: the active role of resting microglia. Trends
the female rat: changes in plasma prolactin, gonadotropins, luteinizing Neurosci. 28, 571573.
hormone-releasing hormone (LHRH), and hypothalamic LHRH content. Ramaswamy, S., Guerriero, K.A., Gibbs, R.B., Plant, T.M., 2008. Structural interactions
Endocrinology 98, 630638. between kisspeptin and GnRH neurons in the mediobasal hypothalamus of the
Ojeda, S.R., Lomniczi, A., Mastronardi, C., Heger, S., Roth, C., Parent, A.S., Matagne, V., male rhesus monkey (Macaca mulatta) as revealed by double
Mungenast, A.E., 2006. Minireview: the neuroendocrine regulation of puberty: immunouorescence and confocal microscopy. Endocrinology 149, 43874395.
is the time ripe for a systems biology approach? Endocrinology 147, 1166 Ransohoff, R.M., Perry, V.H., 2009. Microglial physiology: unique stimuli, specialized
1174. responses. Annu. Rev. Immunol. 27, 119145.
Okada, M., Hayashi, N., Kometani, M., Nakao, K., Inukai, T., 1997. Inuences of Redei, E., Li, L., Halasz, I., McGivern, R.F., Aird, F., 1994. Fast glucocorticoid feedback
ovariectomy and continuous replacement of 17beta-estradiol on the tail skin inhibition of ACTH secretion in the ovariectomized rat: effect of chronic
temperature and behavior in the forced swimming test in rats. Jpn. J. Pharmacol. estrogen and progesterone. Neuroendocrinology 60, 113123.
73, 9396. Reiss, A.L., Abrams, M.T., Singer, H.S., Ross, J.L., Denckla, M.B., 1996. Brain
Olesen, K.M., Ismail, N., Merchasin, E.D., Blaustein, J.D., 2011. Long-term alteration development, gender and IQ in children. A volumetric imaging study. Brain
of anxiolytic effects of ovarian hormones in female mice by a peripubertal 119 (Pt 5), 17631774.
immune challenge. Horm. Behav. 60, 318326. Rizza, R.A., Mandarino, L.J., Gerich, J.E., 1982. Cortisol-induced insulin resistance in
Olivera-Lopez, J.I., Molina-Hernandez, M., Tellez-Alcantara, N.P., Jaramillo, M.T., man: impaired suppression of glucose production and stimulation of glucose
2008. Estradiol and neuropeptide Y (intra-lateral septal) reduce anxiety-like utilization due to a postreceptor detect of insulin action. J. Clin. Endocrinol.
behavior in two animal models of anxiety. Peptides 29, 13961403. Metab. 54, 131138.
ONeil, M.F., Moore, N.A., 2003. Animal models of depression: are there any? Hum. Roberti, J.W., 2004. A review of behavioral and biological correlates of sensation
Psychopharmacol. 18, 239254. seeking. J. Res. Pers. 38, 256279.
Osterlund, M.K., 2010. Underlying mechanisms mediating the antidepressant Rocha, B.A., Fleischer, R., Schaeffer, J.M., Rohrer, S.P., Hickey, G.J., 2005. 17 Beta-
effects of estrogens. Biochim. Biophys. Acta 1800, 11361144. estradiol-induced antidepressant-like effect in the forced swim test is absent in
Palanza, P., Gioiosa, L., Parmigiani, S., 2001. Social stress in mice: gender differences estrogen receptor-beta knockout (BERKO) mice. Psychopharmacology 179,
and effects of estrous cycle and social dominance. Physiol. Behav. 73, 411420. 637643.
Pandaranandaka, J., Poonyachoti, S., Kalandakanond-Thongsong, S., 2006. Anxiolytic Romeo, R.D., Lee, S.J., Chhua, N., McPherson, C.R., McEwen, B.S., 2004a. Testosterone
property of estrogen related to the changes of the monoamine levels in various cannot activate an adult-like stress response in prepubertal male rats.
brain regions of ovariectomized rats. Physiol. Behav. 87, 828835. Neuroendocrinology 79, 125132.
Pandaranandaka, J., Poonyachoti, S., Kalandakanond-Thongsong, S., 2009. Romeo, R.D., Lee, S.J., McEwen, B.S., 2004b. Differential stress reactivity in intact and
Differential effects of exogenous and endogenous estrogen on anxiety as ovariectomized prepubertal and adult female rats. Neuroendocrinology 80,
measured by elevated T-maze in relation to the serotonergic system. Behav. 387393.
Brain Res. 198, 142148. Romeo, R.D., Bellani, R., McEwen, B.S., 2005. Stress-induced progesterone
Paolicelli, R.C., Gross, C.T., 2011. Microglia in development: linking brain wiring to secretion and progesterone receptor immunoreactivity in the
brain environment. Neuron Glia Biol. 7, 7783. paraventricular nucleus are modulated by pubertal development in male
Parent, A.S., Matagne, V., Bourguignon, J.P., 2005. Control of puberty by excitatory rats. Stress 8, 265271.
amino acid neurotransmitters and its clinical implications. Endocrine 28, 281 Romeo, R.D., Bellani, R., Karatsoreos, I.N., Chhua, N., Vernov, M., Conrad, C.D.,
286. McEwen, B.S., 2006. Stress history and pubertal development interact to shape
Pariante, C.M., Miller, A.H., 2001. Glucocorticoid receptors in major depression: hypothalamicpituitaryadrenal axis plasticity. Endocrinology 147, 1664
relevance to pathophysiology and treatment. Biol. Psychiatry 49, 391404. 1674.
Parker, L.N., 1991. Adrenarche. Endocrinol. Metab. Clin. North Am. 20, 7183. Romeo, R.D., Karatsoreos, I.N., Jasnow, A.M., McEwen, B.S., 2007. Age- and stress-
Parker Jr., C.R., Mahesh, V.B., 1976. Hormonal events surrounding the natural onset induced changes in corticotropin-releasing hormone mRNA expression in the
of puberty in female rats. Biol. Reprod. 14, 347353. paraventricular nucleus of the hypothalamus. Neuroendocrinology 85, 199
Parsons, B., Rainbow, T.C., MacLusky, N.J., McEwen, B.S., 1982. Progestin receptor 206.
levels in rat hypothalamic and limbic nuclei. J. Neurosci. 2, 14461452. Roy, V., Chapillon, P., 2004. Further evidences that risk assessment and object
Patton, G.C., Viner, R., 2007. Pubertal transitions in health. Lancet 369, 11301139. exploration behaviours are useful to evaluate emotional reactivity in rodents.
Patton, G.C., Hibbert, M.E., Carlin, J., Shao, Q., Rosier, M., Caust, J., Bowes, G., 1996. Behav. Brain Res. 154, 439448.
Menarche and the onset of depression and anxiety in Victoria, Australia. J. Salim, S., Chugh, G., Asghar, M., 2012. Inammation in anxiety. Adv. Protein Chem.
Epidemiol. Community Health 50, 661666. Struct. Biol. 88, 125.
Paus, T., Zijdenbos, A., Worsley, K., Collins, D.L., Blumenthal, J., Giedd, J.N., Rapoport, Sandstrom, N.J., Williams, C.L., 2004. Spatial memory retention is enhanced by acute
J.L., Evans, A.C., 1999. Structural maturation of neural pathways in children and and continuous estradiol replacement. Horm. Behav. 45, 128135.
adolescents: in vivo study. Science 283, 19081911. Santini, E., Ge, H., Ren, K., Pena de Ortiz, S., Quirk, G.J., 2004. Consolidation of fear
Paus, T., Keshavan, M., Giedd, J.N., 2008. Why do many psychiatric disorders emerge extinction requires protein synthesis in the medial prefrontal cortex. J.
during adolescence? Nat. Rev. Neurosci. 9, 947957. Neurosci. 24, 57045710.
Peleg-Raibstein, D., Feldon, J., 2011. Differential effects of post-weaning juvenile Sapolsky, R.M., Share, L.J., 1994. Rank-related differences in cardiovascular function
stress on the behaviour of C57BL/6 mice in adolescence and adulthood. among wild baboons role of sensitivity to glucocorticoids. Am. J. Primatol. 32,
Psychopharmacology 214, 339351. 261275.
Perry, V.H., Newman, T.A., Cunningham, C., 2003. The impact of systemic infection Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids inuence
on the progression of neurodegenerative disease. Nat. Rev. Neurosci. 4, 103 stress responses? Integrating permissive, suppressive, stimulatory, and
112. preparative actions. Endocr. Rev. 21, 5589.
Petanjek, Z., Judas, M., Simic, G., Rasin, M.R., Uylings, H.B., Rakic, P., Kostovic, I., Schafer, D.P., Lehrman, E.K., Stevens, B., 2012. The quad-partite synapse:
2011. Extraordinary neoteny of synaptic spines in the human prefrontal cortex. microglia-synapse interactions in the developing and mature CNS. Glia.
Proc. Natl. Acad. Sci. USA 108, 1328113286. Schenk, F., 1985. Development of place navigation in rats from weaning to puberty.
Phoenix, C.H., Goy, R.W., Gerall, A.A., Young, W.C., 1959. Organizing action of Behav. Neural Biol. 43, 6985.
prenatally administered testosterone propionate on the tissues mediating Schmidt, P.J., Nieman, L.K., Danaceau, M.A., Adams, L.F., Rubinow, D.R., 1998.
mating behavior in the female guinea pig. Endocrinology 65, 369382. Differential behavioral effects of gonadal steroids in women with and in those
Picazo, O., Estrada-Camarena, E., Hernandez-Aragon, A., 2006. Inuence of the post- without premenstrual syndrome. New Engl. J. Med. 338, 209216.
ovariectomy time frame on the experimental anxiety and the behavioural Schmidt, M.V., Sterlemann, V., Ganea, K., Liebl, C., Alam, S., Harbich, D., Greetfeld, M., Uhr,
actions of some anxiolytic agents. Eur. J. Pharmacol. 530, 8894. M., Holsboer, F., Muller, M.B., 2007. Persistent neuroendocrine and behavioral
Pignatelli, D., Xiao, F., Gouveia, A.M., Ferreira, J.G., Vinson, G.P., 2006. Adrenarche in effects of a novel, etiologically relevant mouse paradigm for chronic social stress
the rat. J. Endocrinol. 191, 301308. during adolescence. Psychoneuroendocrinology 32, 417429.
Pinos, H., Collado, P., Rodriguez-Zafra, M., Rodriguez, C., Segovia, S., Guillamon, A., Schmidt, M.V., Scharf, S.H., Liebl, C., Harbich, D., Mayer, B., Holsboer, F., Muller, M.B.,
2001. The development of sex differences in the locus coeruleus of the rat. Brain 2010a. A novel chronic social stress paradigm in female mice. Horm. Behav. 57,
Res. Bull. 56, 7378. 415420.
Plant, T.M., Barker-Gibb, M.L., 2004. Neurobiological mechanisms of puberty in Schmidt, M.V., Scharf, S.H., Sterlemann, V., Ganea, K., Liebl, C., Holsboer, F., Muller,
higher primates. Hum. Reprod. Update 10, 6777. M.B., 2010b. High susceptibility to chronic social stress is associated with a
Pohl, J., Olmstead, M.C., Wynne-Edwards, K.E., Harkness, K., Menard, J.L., 2007. depression-like phenotype. Psychoneuroendocrinology 35, 635643.
Repeated exposure to stress across the childhoodadolescent period alters rats Schommer, N.C., Hellhammer, D.H., Kirschbaum, C., 2003. Dissociation between
anxiety- and depression-like behaviors in adulthood: the importance of stressor reactivity of the hypothalamuspituitaryadrenal axis and the sympathetic
type and gender. Behav. Neurosci. 121, 462474. adrenalmedullary system to repeated psychosocial stress. Psychosom. Med.
Pousset, F., 1994. Developmental expression of cytokine genes in the cortex and 65, 450460.
hippocampus of the rat central nervous system. Brain Res. Dev. Brain Res. 81, Schwarz, J.M., Sholar, P.W., Bilbo, S.D., 2012. Sex differences in microglial
143146. colonization of the developing rat brain. J. Neurochem. 120, 948963.
M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110 109

Scott, J.P., Stewart, J.M., De Ghett, V.J., 1974. Critical periods in the organization of Toledo-Rodriguez, M., Sandi, C., 2007. Stress before puberty exerts a sex- and age-
systems. Dev. Psychobiol. 7, 489513. related impact on auditory and contextual fear conditioning in the rat. Neural
Shaw, P., Kabani, N.J., Lerch, J.P., Eckstrand, K., Lenroot, R., Gogtay, N., Greenstein, D., Plast. 2007, 112.
Clasen, L., Evans, A., Rapoport, J.L., Giedd, J.N., Wise, S.P., 2008. Tomihara, K., Soga, T., Nomura, M., Korach, K.S., Gustafsson, J.A., Pfaff, D.W., Ogawa,
Neurodevelopmental trajectories of the human cerebral cortex. J. Neurosci. S., 2009. Effect of ER-beta gene disruption on estrogenic regulation of anxiety in
28, 35863594. female mice. Physiol. Behav. 96, 300306.
Sheridan, P.J., 1978. Localization of androgen- and estrogen-concentrating neurons Toth, E., Avital, A., Leshem, M., Richter-Levin, G., Braun, K., 2008a. Neonatal and
in the diencephalon and telencephalon of the mouse. Endocrinology 103, 1328 juvenile stress induces changes in adult social behavior without affecting
1334. cognitive function. Behav. Brain Res. 190, 135139.
Shughrue, P.J., Komm, B., Merchenthaler, I., 1996. The distribution of estrogen Toth, E., Gersner, R., Wilf-Yarkoni, A., Raizel, H., Dar, D.E., Richter-Levin, G., Levit, O.,
receptor-beta mRNA in the rat hypothalamus. Steroids 61, 678681. Zangen, A., 2008b. Age-dependent effects of chronic stress on brain plasticity
Shughrue, P.J., Lane, M.V., Merchenthaler, I., 1997. Comparative distribution of and depressive behavior. J. Neurochem. 107, 522532.
estrogen receptor-alpha and -beta mRNA in the rat central nervous system. J. Town, T., Nikolic, V., Tan, J., 2005. The microglial activation continuum: from
Comp. Neurol. 388, 507525. innate to adaptive responses. J. Neuroinamm. 2, 24.
Silverman, A.B., Reinherz, H.Z., Giaconia, R.M., 1996. The long-term sequelae of child Tremblay, M.E., Stevens, B., Sierra, A., Wake, H., Bessis, A., Nimmerjahn, A., 2011. The
and adolescent abuse: a longitudinal community study. Child Abuse Negl. 20, role of microglia in the healthy brain. J. Neurosci. 31, 1606416069.
709723. Tsoory, M., Richter-Levin, G., 2006. Learning under stress in the adult rat is
Simerly, R.B., Chang, C., Muramatsu, M., Swanson, L.W., 1990. Distribution of differentially affected by juvenile or adolescent stress. Int. J.
androgen and estrogen receptor mRNA-containing cells in the rat brain: an Neuropsychopharmacol. 9, 713728.
in situ hybridization study. J. Comp. Neurol. 294, 7695. Tsoory, M., Cohen, H., Richter-Levin, G., 2007. Juvenile stress induces a
Sisk, C.L., Foster, D.L., 2004. The neural basis of puberty and adolescence. Nat. predisposition to either anxiety or depressive-like symptoms following stress
Neurosci. 7, 10401047. in adulthood. Eur. Neuropsychopharmacol. 17, 245256.
Sisk, C.L., Zehr, J.L., 2005. Pubertal hormones organize the adolescent brain and Turner, R.J., Lloyd, D.A., 2004. Stress burden and the lifetime incidence of psychiatric
behavior. Front. Neuroendocrinol. 26, 163174. disorder in young adults: racial and ethnic contrasts. Arch. Gen. Psychiatry 61,
Sizonenko, P.C., 1989. Physiology of puberty. J. Endocrinol. Invest. 12, 5963. 481488.
Smejkalova, T., Woolley, C.S., 2010. Estradiol acutely potentiates hippocampal Ulrich-Lai, Y.M., Herman, J.P., 2009. Neural regulation of endocrine and autonomic
excitatory synaptic transmission through a presynaptic mechanism. J. Neurosci. stress responses. Nat. Rev. Neurosci. 10, 397409.
30, 1613716148. Urbanska, M., Swiech, L., Jaworski, J., 2012. Developmental plasticity of the dendritic
Smith, K.A., 2012a. Toward a molecular understanding of adaptive immunity: a compartment: focus on the cytoskeleton. Adv. Exp. Med. Biol. 970, 265284.
chronology, part II. Front. Immunol. 3, 364. Uys, J.D., Marais, L., Faure, J., Prevoo, D., Swart, P., Mohammed, A.H., Stein, D.J.,
Smith, K.A., 2012b. Toward a molecular understanding of adaptive immunity: a Daniels, W.M., 2006a. Developmental trauma is associated with behavioral
chronology, part I. Front. Immunol. 3, 369. hyperarousal, altered HPA axis activity, and decreased hippocampal
Sowell, E.R., Thompson, P.M., Tessner, K.D., Toga, A.W., 2001. Mapping continued neurotrophin expression in the adult rat. Ann. N. Y. Acad. Sci. 1071, 542546.
brain growth and gray matter density reduction in dorsal frontal cortex: inverse Uys, J.D., Muller, C.J., Marais, L., Harvey, B.H., Stein, D.J., Daniels, W.M., 2006b. Early
relationships during postadolescent brain maturation. J. Neurosci. 21, 8819 life trauma decreases glucocorticoid receptors in rat dentate gyrus upon adult
8829. re-stress: reversal by escitalopram. Neuroscience 137, 619625.
Spear, L.P., 2000. The adolescent brain and age-related behavioral manifestations. Vandenbergh, J.G., 1967. Effect of the presence of a male on the sexual maturation
Neurosci. Biobehav. Rev. 24, 417463. of female mice. Endocrinology 81, 345349.
Spinedi, E., Chisari, A., Pralong, F., Gaillard, R.C., 1997. Sexual dimorphism in Vandenbergh, J.G., 1969. Male odor accelerates female sexual maturation in mice.
the mouse hypothalamicpituitaryadrenal axis function after endotoxin Endocrinology 84, 658660.
and insulin stresses during development. NeuroImmunoModulation 4, 77 Vasudevan, N., Pfaff, D.W., 2008. Non-genomic actions of estrogens and their
83. interaction with genomic actions in the brain. Front. Neuroendocrinol. 29, 238
Stellwagen, D., Malenka, R.C., 2006. Synaptic scaling mediated by glial TNF-alpha. 257.
Nature 440, 10541059. Verney, C., Monier, A., Fallet-Bianco, C., Gressens, P., 2010. Early microglial
Stellwagen, D., Beattie, E.C., Seo, J.Y., Malenka, R.C., 2005. Differential regulation of colonization of the human forebrain and possible involvement in
AMPA receptor and GABA receptor trafcking by tumor necrosis factor-alpha. J. periventricular white-matter injury of preterm infants. J. Anat. 217, 436448.
Neurosci. 25, 32193228. Viau, V., 2002. Functional cross-talk between the hypothalamicpituitarygonadal
Sterlemann, V., Ganea, K., Liebl, C., Harbich, D., Alam, S., Holsboer, F., Muller, M.B., and adrenal axes. J. Neuroendocrinol. 14, 506513.
Schmidt, M.V., 2008. Long-term behavioral and neuroendocrine alterations Viau, V., Meaney, M.J., 1991. Variations in the hypothalamicpituitaryadrenal
following chronic social stress in mice: implications for stress-related disorders. response to stress during the estrous cycle in the rat. Endocrinology 129, 2503
Horm. Behav. 53, 386394. 2511.
Stevens, B., Allen, N.J., Vazquez, L.E., Howell, G.R., Christopherson, K.S., Nouri, N., Viau, V., Meaney, M.J., 1996. The inhibitory effect of testosterone on hypothalamic
Micheva, K.D., Mehalow, A.K., Huberman, A.D., Stafford, B., Sher, A., Litke, pituitaryadrenal responses to stress is mediated by the medial preoptic area. J.
A.M., Lambris, J.D., Smith, S.J., John, S.W., Barres, B.A., 2007. The classical Neurosci. 16, 18661876.
complement cascade mediates CNS synapse elimination. Cell 131, 1164 Vidal, J., Bie, J., Granneman, R.A., Wallinga, A.E., Koolhaas, J.M., Buwalda, B., 2007.
1178. Social stress during adolescence in Wistar rats induces social anxiety in
Straub, R.H., Buttgereit, F., Cutolo, M., 2011. Alterations of the hypothalamic adulthood without affecting brain monoaminergic content and activity. Physiol.
pituitaryadrenal axis in systemic immune diseases a role for misguided Behav. 92, 824830.
energy regulation. Clin. Exp. Rheumatol. 29, S23S31. Vidal, J., Buwalda, B., Koolhaas, J.M., 2011a. Differential long-term effects of social
Streit, W.J., 2010. Microglial activation and neuroinammation in Alzheimers stress during adolescence on anxiety in Wistar and wild-type rats. Behav.
disease: a critical examination of recent history. Front. Aging Neurosci. 2, 22. Processes 87, 176182.
Streit, W.J., Graeber, M.B., Kreutzberg, G.W., 1988. Functional plasticity of microglia: Vidal, J., Buwalda, B., Koolhaas, J.M., 2011b. Male Wistar rats are more susceptible to
a review. Glia 1, 301307. lasting social anxiety than Wild-type Groningen rats following social defeat
Streit, W.J., Walter, S.A., Pennell, N.A., 1999. Reactive microgliosis. Prog. Neurobiol. stress during adolescence. Behav. Processes 88, 7680.
57, 563581. Vilcek, J., 1998. The cytokines: an overview. In: Thomson, A. (Ed.), The Cytokine
Streit, W.J., Mrak, R.E., Grifn, W.S., 2004. Microglia and neuroinammation: a Handbook. Academic Press, San Diego, pp. 120.
pathological perspective. J. Neuroinamm. 1, 14. Vitaliano, P.P., Scanlan, J.M., Zhang, J., Savage, M.V., Hirsch, I.B., Siegler, I.C., 2002. A
Suda, S., Segi-Nishida, E., Newton, S.S., Duman, R.S., 2008. A postpartum model in path model of chronic stress, the metabolic syndrome, and coronary heart
rat: behavioral and gene expression changes induced by ovarian steroid disease. Psychosom. Med. 64, 418435.
deprivation. Biol. Psychiatry 64, 311319. Wagner, M.K., 2001. Behavioral characteristics related to substance abuse and risk-
Tafet, G.E., Bernardini, R., 2003. Psychoneuroendocrinological links between chronic taking, sensation-seeking, anxiety sensitivity, and self-reinforcement. Addict.
stress and depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 27, 893 Behav. 26, 115120.
903. Wake, H., Moorhouse, A.J., Jinno, S., Kohsaka, S., Nabekura, J., 2009. Resting
Takao, T., Culp, S.G., Newton, R.C., De Souza, E.B., 1992. Type I interleukin-1 microglia directly monitor the functional state of synapses in vivo and
receptors in the mouse brain-endocrine-immune axis labelled with determine the fate of ischemic terminals. J. Neurosci. 29, 39743980.
[125I]recombinant human interleukin-1 receptor antagonist. J. Walker, E.F., Walder, D.J., Reynolds, F., 2001. Developmental changes in cortisol
Neuroimmunol. 41, 5160. secretion in normal and at-risk youth. Dev. Psychopathol. 13, 721732.
Tanner, J.M., 1962. Growth at Adolescence. Blackwell Scientic Publications, Oxford. Wall, P.M., Messier, C., 2001. Methodological and conceptual issues in the use of the
Taylor, E., Rutter, M., 1985. Sex differences in neurodevelopmental and psychiatric elevated plus-maze as a psychological measurement instrument of animal
disorders: one explanation or many? Behav. Brain Sci. 8, 460. anxiety-like behavior. Neurosci. Biobehav. Rev. 25, 275286.
Tetel, M.J., Auger, A.P., Charlier, T.D., 2009. Whos in charge? Nuclear receptor Wallen, K., 2005. Hormonal inuences on sexually differentiated behavior in
coactivator and corepressor function in brain and behavior. Front. nonhuman primates. Front. Neuroendocrinol. 26, 726.
Neuroendocrinol. 30, 328342. Wang, H., Meng, X.H., Ning, H., Zhao, X.F., Wang, Q., Liu, P., Zhang, H., Zhang, C.,
Tirelli, E., Laviola, G., Adriani, W., 2003. Ontogenesis of behavioral sensitization and Chen, G.H., Xu, D.X., 2010. Age- and gender-dependent impairments of
conditioned place preference induced by psychostimulants in laboratory neurobehaviors in mice whose mothers were exposed to lipopolysaccharide
rodents. Neurosci. Biobehav. Rev. 27, 163178. during pregnancy. Toxicol. Lett. 192, 245251.
110 M.K. Holder, J.D. Blaustein / Frontiers in Neuroendocrinology 35 (2014) 89110

Watt, M.J., Burke, A.R., Renner, K.J., Forster, G.L., 2009. Adolescent male rats exposed Wright, L.D., Muir, K.E., Perrot, T.S., 2012b. Stress responses of adolescent male and
to social defeat exhibit altered anxiety behavior and limbic monoamines as female rats exposed repeatedly to cat odor stimuli, and long-term enhancement
adults. Behav. Neurosci. 123, 564576. of adult defensive behaviors. Dev. Psychobiol..
Waylen, A., Wolke, D., 2004. Sex n drugs n rock n roll: the meaning and Xiao, L., Jordan, C.L., 2002. Sex differences, laterality, and hormonal regulation of
social consequences of pubertal timing. Eur. J. Endocrinol. 151 (Suppl. 3), androgen receptor immunoreactivity in rat hippocampus. Horm. Behav. 42,
U151U159. 327336.
Weathington, J.M., Arnold, A.R., Cooke, B.M., 2012. Juvenile social subjugation Xu, X., Zhang, Z., 2006. Effects of estradiol benzoate on learning-memory behavior
induces a sex-specic pattern of anxiety and depression-like behaviors in adult and synaptic structure in ovariectomized mice. Life Sci. 79, 15531560.
rats. Horm. Behav. 61, 9199. Yamada, S., Noguchi, D., Ito, H., Yamanouchi, K., 2009. Sex and regional differences
Whitworth, J.A., Brown, M.A., Kelly, J.J., Williamson, P.M., 1995. Mechanisms of in decrease of estrogen receptor alpha-immunoreactive cells by estrogen in rat
cortisol-induced hypertension in humans. Steroids 60, 7680. hypothalamus and midbrain. Neurosci. Lett. 463, 135139.
Wiemann, J.N., Clifton, D.K., Steiner, R.A., 1989. Pubertal changes in gonadotropin- Yang, S., Liu, Z.W., Wen, L., Qiao, H.F., Zhou, W.X., Zhang, Y.X., 2005. Interleukin-
releasing hormone and proopiomelanocortin gene expression in the brain of the 1beta enhances NMDA receptor-mediated current but inhibits excitatory
male rat. Endocrinology 124, 17601767. synaptic transmission. Brain Res. 1034, 172179.
Wilkin, M.M., Waters, P., McCormick, C.M., Menard, J.L., 2012. Intermittent Yankova, M., Hart, S.A., Woolley, C.S., 2001. Estrogen increases synaptic connectivity
physical stress during early- and mid-adolescence differentially alters rats between single presynaptic inputs and multiple postsynaptic CA1 pyramidal cells: a
anxiety- and depression-like behaviors in adulthood. Behav. Neurosci. 126, serial electron-microscopic study. Proc. Natl. Acad. Sci. USA 98, 35253530.
344360. Yildirim, M., Mapp, O.M., Janssen, W.G., Yin, W., Morrison, J.H., Gore, A.C., 2008.
Woo, T.U., Pucak, M.L., Kye, C.H., Matus, C.V., Lewis, D.A., 1997. Peripubertal Postpubertal decrease in hippocampal dendritic spines of female rats. Exp.
renement of the intrinsic and associational circuitry in monkey prefrontal Neurol. 210, 339348.
cortex. Neuroscience 80, 11491158. Yokosuka, M., Okamura, H., Hayashi, S., 1997. Postnatal development and sex
Woolley, C.S., 2000. Effects of oestradiol on hippocampal circuitry. Novartis Found. difference in neurons containing estrogen receptor-alpha immunoreactivity in
Symp. 230, 173180, discussion 181-7. the preoptic brain, the diencephalon, and the amygdala in the rat. J. Comp.
Wright, L.D., Hebert, K.E., Perrot-Sinal, T.S., 2008. Periadolescent stress exposure Neurol. 389, 8193.
exerts long-term effects on adult stress responding and expression of prefrontal Young, E.A., Altemus, M., Parkison, V., Shastry, S., 2001. Effects of estrogen
dopamine receptors in male and female rats. Psychoneuroendocrinology 33, antagonists and agonists on the ACTH response to restraint stress in female
130142. rats. Neuropsychopharmacology 25, 881891.
Wright, L.D., Muir, K.E., Perrot, T.S., 2012a. Enhanced stress responses in adolescent Zhang, J.Q., Cai, W.Q., Zhou, D.S., Su, B.Y., 2002. Distribution and differences of
versus adult rats exposed to cues of predation threat, and peer interaction as a estrogen receptor beta immunoreactivity in the brain of adult male and female
predictor of adult defensiveness. Dev. Psychobiol. 54, 4769. rats. Brain Res. 935, 7380.

Вам также может понравиться