Summary
Males and females exhibit numerous anatomical and physiological differences in the brain which often underlie important sex differences in physiology or behavior, including aspects relating to reproduction. Neural sex differences are both region- and trait-specific and may consist of divergences in synapse morphology, neuron size and number, and specific gene expression levels. In most cases, sex differences are induced by the sex steroid hormonal milieu during early perinatal development. In rodents, the hypothalamic anteroventral periventricular nucleus (AVPV) is sexually differentiated as a result of postnatal sex steroids, and specific neuronal populations in this nucleus are also sexually dimorphic, with females possessing more kisspeptin, dopaminergic, and GABA/glutamate neurons than males. The ability of female rodents, but not males, to display an estrogen-induced luteinizing hormone (LH) surge is consistent with the higher levels of these neuropeptides in the AVPV of females. Of these AVPV populations, the recently-identified kisspeptin system has been most strongly implicated as a critical component of the sexually-dimorphic LH surge mechanism, though GABA and glutamate have also received some attention. New findings have suggested that the sexual differentiation and development of kisspeptin neurons in the AVPV is mediated by developmental estradiol signaling. Although apoptosis is the most common process implicated in neuronal sexual differentiation, it is currently unknown how developmental estradiol acts to differentiate specific neuronal populations in the AVPV, such as kisspeptin or dopaminergic neurons.
Keywords: sex difference, sexually dimorphic, sexual differentiation, hypothalamus, kisspeptin, Kiss1, tyrosine hydroxylase, development, reproduction, AVPV
Introduction
In mammals and many other vertebrates, both the brain and reproductive system are anatomically and physiologically differentiated between males and females. These sex differences range from disparities between men and women in the prevalence of certain reproductive health disorders to gender differences in normal reproductive physiology and reproductive circuits in the brain. In addition to girls typically entering puberty earlier than boys, girls are also more likely to present with early onset pubertal disease, whereas boys are more likely to present with diseases of delayed puberty or the inability to reach full sexual maturation. Often, these conditions are linked to defects in the secretion of reproductive hormones, which is controlled by forebrain and hypothalamic circuits. Although the key hypothalamic populations and neuronal mechanisms that underlie sex differences in reproductive disorders and physiology remain poorly understood, a specific region of the hypothalamus, the anteroventral periventricular nucleus (AVPV), is sexually differentiated in rodents, being larger in volume and containing more cells in females than males. Moreover, certain neuronal populations within the rodent AVPV are also sexually dimorphic, such as dopaminergic, GABA/glutamate, and Kiss1-expressing neurons, with females also having more of each of these specific cell types than males [1–3]. It has been postulated that these sexually dimorphic neuronal populations in the AVPV influence reproductive function, such as the ability of adult females, but not males, to produce the preovulatory gonadotropin surge, as well as possibly influencing sex differences in puberty onset. Although the Kiss1 sexual dimorphism in the AVPV is particularly compelling because kisspeptin signaling has been implicated in fertility and puberty in many species, including humans [4–6], it is currently unknown if the other sexually dimorphic AVPV populations, such as dopaminergic or GABA/glutamate neurons, also play similar or complementary roles. This brief review discusses the functional relevance of the sexual differentiation of these various AVPV neural circuits, with an emphasis on the newly identified Kiss1 system, and highlights some of the most recent findings in the field from the past few years. Additionally, based on known mechanisms of the sexual differentiation the brain, we also discuss some of the potential developmental mechanisms that could be underlying the sex-steroid mediated sexual differentiation of neuronal populations in the AVPV.
Sexual Differentiation of Neural Circuits and Reproductive Physiology
Mammals exhibit numerous sex differences in physiology and behavior, including several indices of reproductive biology [3,7,8]. Many of the physiological and anatomical differences between females and males reflect sex differences within the brain. In fact, there are many well-documented sex differences in the brains of many species, including insects, fish, birds, and mammals (reviewed in [9]). These neural sexual dimorphisms are present in many different areas of the brain, including the hypothalamus, hippocampus, and medial pre-optic nucleus [3,8,10,11]. Sex differences in the brain may include morphological differences in synapses, differential gene expression, and disparities in cell size or regional volume, including the number of neurons present (Table 1) [7,10,12]. For example, the medial preoptic nucleus (mPOA) and the principal bed nucleus of the stria terminalis (BNST) are both larger and possess more neurons in male rodents than in females [7,12,13]. In addition, the rodent BNST exhibits sexually dimorphic vasopressin (VP) gene expression, with adult males having more VP neurons in the BNST than females [14]. Interestingly, neuronal projections from the BNST to the AVPV are also sexually differentiated, with males having an abundance of projections compared to females [15]. Conversely, the AVPV of the rodent hypothalamus is greater in size and cell number in females than in males [16]. Particular neuronal populations in the AVPV are also sexually differentiated, with females having greater numbers of dopaminergic, GABA/glutamate, and Kiss1-expressing neurons [1,2,17]. In many cases, the functional significance of sexually dimorphic brain regions is currently unclear, and it remains unknown why neuron size or gene expression levels are higher in one sex than another. However, in a few cases, accumulating data has allowed for conjecture of potential functions of specific neural sex differences. For example, it has been postulated that the sexually dimorphic BNST is involved in modulating well-documented sex differences in anxiety and stress responses [18]. The AVPV, on the other hand, is known to be involved in reproduction and sex differences in this region may mediate numerous sex differences related to reproductive function, a possibility discussed in more detail below.
Table 1.
A partial list of some neural sexual dimorphisms in the rodent brain. In some cases, the possible function of each sex difference is either unknown or only hypothesized based on available evidence. See text for more details.
| Brain Region | Phenotype | Sex Difference | Possible Function | References |
|---|---|---|---|---|
| AVPV | Cell number/size | females>males | unknown (perhaps reproduction) | [7,30] |
| AVPV | Kiss1 expression | females>males | puberty and/or reproduction | [1,32] |
| AVPV | TH expression | females>males | unknown | [3,31] |
| AVPV | GABA/glutamate | females>males | puberty and/or reproduction | [2] |
| BNST | Cell number/size | males>females | stress/anxiety/reproduction | [13,18] |
| BNST | VP expression | males>females | stress/anxiety/reproduction | [65] |
| BNST | Projections to AVPV | males>females | unknown | [15] |
| mPOA | Cell number/size | males>females | sexual behavior | [7] |
| Hippocampus | Cell number/size | males>females | memory formation | [7,58] |
In some cases, gender-biased genetic contributions by sex chromosomes lead to sexual dimorphisms in the brain [19]; however, the majority of known sex differences are induced by the sex steroid hormonal milieu during early postnatal development [7]. During the postnatal “critical period”, which is the first 7–10 days of life in rodents, males secrete gonadal testosterone (T) whereas females secrete negligible levels of sex steroids. Experiments manipulating the postnatal steroid milieu support the model that the presence or absence of postnatal T secretion determines whether sexually dimorphic traits develop to be male-like or female-like in adulthood (Figure 1). Thus, in newborn males, postnatal T [or its aromatized product, estradiol (E2)] organizes the developing brain to exhibit a male-like phenotype in adulthood [8]. In contrast, newborn females are not exposed to sufficient levels of sex steroids and therefore, their brains develop to be female-like in adulthood [3,7]. Supporting this model, castration of newborn males (which removes postnatal T secretion) results in the development of feminized neural populations, whereas sex steroid treatment to newborn females (to simulate T secretion in normal males) results in the development of masculinized neuronal circuitry (reviewed in [4]).
Figure 1.

Like the brain, the neuroendocrine reproductive axis also contains multiple discernible sexual dimorphisms. In rodents, these include sexually differentiated neural mechanisms controlling sex-specific reproductive behaviors [20], earlier pubertal maturation in females [21,22], and the presence of neural circuitry that generates preovulatory hormone surges in females but not males [1,23]. Besides sex differences in normal reproductive physiology and behavior, there are a number of reproductive health disorders and diseases in humans which present with disparate frequency between the sexes, such as idiopathic hypogonadotropic hypogonadism (more common in men), precocious puberty (more common in girls), and constitutional delayed puberty (more common in boys) [24,25]. Presumably, many of the sex differences in reproductive physiology, behavior, and health disorders reflect underlying sex differences in brain circuitries (Figure 1). For example, as mentioned earlier, if male rodents are castrated during the postnatal critical period, thereby preventing their ability to secrete postnatal T, they display female-like brain circuits and reproductive physiology in adulthood, including the ability to display an E2-induced luteinizing hormone (LH) surge, similar to normal adult females [7,8,26]. Conversely, if a female rodent is given a single injection of T or E2 during the postnatal critical period to mimic the male postnatal T secretion, she displays male-like brain phenotypes and reproductive physiology in adulthood, including her inability to produce a preovulatory LH surge or to display normal female sexual behaviors such as lordosis [7,8,26].
Sexually Differentiated Neurons of the AVPV and Their Role in Reproduction
Neurons that secrete gonadotropin releasing hormone (GnRH) are the final common pathway by which the brain controls reproduction. Interestingly, GnRH neurons are not themselves sexually dimorphic, suggesting that other neuronal populations influence sex differences in reproductive status. In rodents, the hypothalamic AVPV, which is responsible for conveying hormonal and environmental signals to GnRH neurons, does display sex differences in various parameters [7,27]. Importantly, the rodent AVPV is considered to be one of the main anatomical sites that drive the sexually differentiated preovulatory LH surge [28]. In accordance with this conjecture, the AVPV exhibits several robust sex differences [7,27,29]: both the overall size of the AVPV nucleus and the total number of cells in this region are greater in adult females than males [30]. Moreover, the AVPV contains a number of sexually dimorphic subpopulations which have been implicated to varying degrees in modulating or regulating GnRH cells, including dopaminergic neurons, which express the tyrosine hydroxylase (TH) enzyme [3,31], cells expressing both GABA and glutamate [2], and Kiss1 neurons [1,32,33]. In all three cases, the cell number of these specific AVPV neuronal populations is greater in females than males.
Tyrosine Hydroxylase and GABA/Glutamate Neurons
Dopaminergic neurons were the first sexually dimorphic neuronal population identified in the AVPV [17]. These sexually differentiated neurons, more prevalent in females than males, were originally detected by Simerly and colleagues in the rat AVPV with the use of immunohistochemical staining of the TH enzyme that synthesizes dopamine; the TH sex difference has since been confirmed in the mouse AVPV [17,30,31,34]. Although the higher amount of these dopaminergic AVPV neurons in females correlates with their ability to generate an LH surge, the evidence supporting the role of dopamine in the surge event is inconclusive [7]. There is evidence, however, that the second identified sexually dimorphic population in the AVPV, GABA/glutamate neurons, may contribute to signaling between the AVPV and GnRH neurons. Earlier work showed that antagonists to GABA and glutamate receptors blocked GnRH release and the proestrus LH surge in rats [2,35], and GABA treatment also affects the electrical activity of GnRH neurons [36,37]. More recently, it was discovered that the synaptic terminals of dual phenotype GABA/glutamate neurons in the AVPV directly contact GnRH neurons in rats, and that E2 inhibits GABA release and increases glutamate release during the LH surge [2]. Although these findings suggest that this particular neuronal population may be involved in the sexually dimorphic LH surge, at present, the role of GABA/glutamate AVPV neurons in eliciting the LH surge still remains poorly defined.
Kiss1 Neurons
More recently, the neuropeptide kisspeptin has been implicated in the direct regulation of GnRH secretion and sexually dimorphic reproductive function (reviewed in [4]). Encoded by the Kiss1 gene, kisspeptin is a ligand for the G-protein-coupled receptor, Kiss1R, formerly known as GPR54 [38–40]. In mammals, Kiss1 mRNA expression and kisspeptin immunoreactivity have been detected in two discrete regions of the hypothalamus, the arcuate nucleus (ARC) and the AVPV [29]. Recent studies have identified the importance of Kiss1 for reproductive function in humans and other species, including mice. Deletions or mutations in the Kiss1 or kisspeptin receptor genes result in infertility and impaired pubertal maturation [41–43]. Exogenous treatment of rodents, sheep, monkeys, and humans with kisspeptin increases circulating LH and FSH levels [44,45]. The stimulation of gonadotropin secretion by kisspeptin is likely via activation of GnRH neurons since kisspeptin induces c-Fos expression in GnRH neurons, increases electrical activity of GnRH neurons, and causes the secretion of GnRH in hypothalamic explants [34,46–48]. Moreover, kisspeptin-containing fibers directly contact GnRH neurons, which express the kisspeptin receptor [32,46], suggesting that kisspeptin acts directly on GnRH neurons.
In 2007, Kiss1-expressing neurons in the AVPV were shown to be sexually differentiated, with adult females possessing more Kiss1 mRNA than males [1]. Similar observations have been reported for kisspeptin protein levels in the AVPV [32,33]. Thus, like TH and GABA/glutamate, the presence of numerous Kiss1 neurons in the AVPV correlates with the ability of an animal to generate an LH surge: adult females have high Kiss1 levels and can generate an LH surge, while adult males have little AVPV Kiss1 expression and cannot surge (even with E2 treatment). However, in contrast to TH and GABA/glutamate neurons, mounting evidence supports a critical involvement of kisspeptin in the sexually differentiated LH surge. For example, E2 dramatically stimulates Kiss1 expression in the AVPV [49] and Kiss1 neuronal activity in this region is upregulated in a circadian pattern in complete synchrony with the circadian timing of the LH surge [50]. Furthermore, Kiss1-null or Kiss1R-null mice are unable to produce an LH surge, even in the presence of E2 [5]. Most recently, it was shown that central infusion of a kisspeptin receptor antagonist in cycling female rats blocks the proestrus LH surge, emphasizing the requirement for kisspeptin signaling in this sexually dimorphic reproductive event [6]. In contrast to the AVPV population, Kiss1 cells in the ARC are not sexually dimorphic in adult rodents, regardless of adulthood sex steroid milieu [1,51]. However, it is noteworthy that sex differences in the regulation of Kiss1 expression in the ARC are present in mice during the prepubertal period, which may relate to sex differences in sexual maturation [51].
Regulation of Sexual Differentiation of the AVPV by Sex Steroids
Like many other sexually dimorphic brain traits, at least two of the AVPV populations, Kiss1 and TH, have been shown to be sexually differentiated by sex steroids during early postnatal development [1,31,33,52]. Postnatal T treatment to newborn female rats masculinizes the development of the AVPV TH population; the effects of T on the sexual differentiation of these TH neurons has been shown to be mediated by estrogen signaling (via aromatization of T to E2), likely via estrogen receptor α [31,52]. Although circulating sex steroids in adulthood can dramatically stimulate Kiss1 expression in the AVPV [49,53], the adult sex steroid milieu does not account for sex differences in AVPV Kiss1 expression [1]. Male and female rats that are gonadectomized as adults and treated with identical E2 levels still display sexually dimorphic Kiss1 expression in the AVPV [1,54]. Rather, the Kiss1 sex difference, like that of TH, appears to be permanently organized early in development by sex steroid signaling during the postnatal critical period (Figure 1). Indeed, castrating male rats at birth causes a permanent feminization of the developing AVPV Kiss1 system [33]. Conversely, postnatal female rats treated once with T or E2 exhibit a robust reduction of Kiss1- or kisspeptin-expressing cells in the AVPV in adulthood, similar to normal males [1,33,55].
The fact that postnatal E2 treatment can, like T, alter the development of the Kiss1 system suggests that masculinization of the AVPV Kiss1 system is likely mediated via aromatization of T to E2 during the critical period (as is also the case for the TH system). Importantly, the masculinzation (i.e., reduction) of AVPV Kiss1 expression in female rats treated at birth with sex steroids is consistent with their inability to generate an LH surge as adults [33], highlighting the link between the sexually dimorphic AVPV Kiss1 system and LH surge event. However, while the development of the sexually dimorphic Kiss1 system is dependent on the postnatal sex steroid milieu, it is unclear exactly how postnatal sex steroids, primarily E2, direct the sexual differentiation of this system.
Mechanisms of Sex Steroid-Mediated Sexual Differentiation of the Brain
How does T or E2 direct the sexual differentiation of the AVPV, and more specifically, subpopulations within the AVPV? Several hormone-dependent mechanisms, such as differential neurogenesis, migration, epigenetics, and apoptosis, have been implicated in the sexual differentiation and development of various neuronal populations (Figure 1) [7,11,56,57]. E2, for example, can promote neurogenesis in the olfactory bulb and dentate gyrus of the adult rat hippocampus, leading to more newly-formed neurons in females [7]. Likewise, in the developing rat hippocampus, sex steroids (which are higher in postnatal males than females) increase the number of new cells, leading to more neurons present in males [58]. Because the AVPV as a whole (i.e., total size of the nucleus) does not undergo differential neurogenesis in response to postnatal sex steroids [7], neurogenesis may not be a major contributor to the sexual differentiation of specific subpopulations, such as Kiss1 neurons, within the AVPV. However, this assertion has not yet been directly tested.
Many sexually dimorphic populations in the brain arise via apoptotic mechanisms (i.e., programmed cell death) [30,59,60]. In fact, sex differences in the overall size and total cell number of the AVPV, as well as other brain regions such as the BNST, are induced by apoptosis. Most of these apoptotic-induced sex differences are dependent on the pro-apoptotic gene, Bax [30,59,61]. BAX is a pro-apoptotic protein located primarily in the cytosol in a healthy cell. In response to cell death signals, BAX translocates to the mitochondria where it precipitates the release of cytochrome c. Cytochrome c release activates caspases, which degrade target substrates in the cell, resulting in cell death [62]. Interestingly, in the developing rat AVPV, postnatal males have higher Bax expression than females (and hence, potentially more cell death) [63], which correlates with the presence of fewer AVPV cells present in adult males than females. The differential postnatal expression of Bax also correlates with higher sex steroid levels in postnatal males than females, suggesting that sex steroids might affect Bax expression [63]. Indeed, neonatal E2 treatment of female rats increases the number of apoptotic AVPV neurons, supporting this conjecture [64]. Finally, a recent study determined that the sex difference in the total number of AVPV and BNST neurons is eliminated in Bax knockout mice [30]. Thus, at least one AVPV trait, total cell number, is sexually differentiated via Bax-dependent apoptotic mechanisms. Despite these findings, the sexual differentiation of TH neurons in the AVPV appears to occur via a Bax-independent mechanism, because Bax knockout mice do not have altered sexual differentiation of AVPV TH neurons [30]. Thus, this TH population is sexually differentiated by either other apoptotic pathways or non-apoptosis mechanisms. Likewise, although the sex difference in total cell number and volume of the BNST is due to Bax-dependent apoptosis, the mechanism of sexual differentiation of the BNST neuronal population expressing VP is not due to sexually differentiated cell death [65] Intriguingly, however, recent data indicate that sexual differentiation of AVPV GABA-ergic neurons may involve sex-specific apoptosis that is mediated via tumor necrosis factor α (TNFα) -dependent and -independent mechanisms [60], raising the possibility that TH and VP cells are also differentiated this way. The possibility that the AVPV Kiss1 system is also regulated via BAX- or TNFα- dependent apoptotic pathways has not yet been tested.
Epigenetic changes precipitated by sex hormones early in life are emerging as critical contributors to alterations in neuronal cell number and gene expression [56]. Modifications to histones (proteins that organize DNA into chromatin) or DNA methylation are thought to influence the transcriptional activity of specific genes, and histone acetylation in particular is associated with transcriptional activation [66]. Recently, histone acetylation has been implicated in the postnatal sex steroid-induced sexual differentiation of the total number of neurons and overall density of the BNST [57]. In rodents, the BNST contains more cells and is larger in volume in males than females. Pharmacological disruption of histone deacetylation during the postnatal critical period, thereby increasing the level of acetylated histones, resulted in the feminization of the BNST region [57]. In this case, it is likely that epigenetic changes directly or indirectly affected the process of apoptosis in the BNST, as sex differences in this region are known to be governed specifically by BAX-mediated apoptosis [59]. The concept that postnatal sex steroid milieu influences the sexual differentiation and phenotypic identity of numerous neuronal populations via epigenetic mechanisms is particularly attractive, especially in light of new studies implicating promoter DNA methylation (an epigenetic modification affecting transcriptional activity) in the sexual differentiation of estrogen receptor α expression in the developing rat pre-optic area [67]. Males had higher levels of estrogen receptor α methylation than females, which correlated with lower expression levels of the gene in the male pre-optic area [67]. Overall, these findings implicate the involvement of chromatin remodeling in the sexual differentiation of the brain, and suggests that other sexually dimorphic traits elsewhere in the brain, such as the AVPV, may arise in part due to epigenetic changes.
Conclusions
The hypothalamic AVPV region and particular neuronal populations within the AVPV are sexually differentiated under the influence of postnatal sex steroid milieu, with adult females possessing more AVPV neurons overall as well as more Kiss1, TH, and GABA/glutamate neurons than males. The sexual differentiation of AVPV neurons expressing GABA/glutamate, TH, and Kiss1, may underlie the ability of females, but not males, to display a GnRH/LH surge. However, of these sexually dimorphic AVPV populations, Kiss1 has been most strongly linked to both reproductive function and the regulation of sexually-dimorphic LH surge, though additional evidence supports a possible role of GABA/Glutamate as well. Although many advances have been made in the understanding of these sexually differentiated neuronal populations, there are important issues that have yet to be resolved. Such issues include defining the specific mechanisms for the sex steroid-induced sexual differentiation of these AVPV neuronal populations, and identifying if other sexually differentiated AVPV neurons, besides just Kiss1cells, play a critical role in sexually dimorphic reproductive function. Importantly, understanding the mechanisms underlying the development of sexually dimorphic AVPV populations and neuropeptide expression levels may have clinical relevance due to the abundance of sexually differentiated disorders, including some relating to reproductive function.
Acknowledgments
The authors are funded by the Eunice Kennedy Shriver National Institutes of Child Health and Human Development (NICHD) through grants R00 HD0561757 and T32 HD007203. Dr. Kauffman is also supported by the NICHD through cooperative agreement U54 HD012303 as part of the Specialized Cooperative Centers Program in Reproduction and Infertility Research.
Footnotes
Conflict of Interest: The authors declare no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References and Recommended Reading
Papers of particular interest, published within the period of the review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1••.Kauffman AS, Gottsch ML, Roa J, Byquist AC, Crown A, Clifton DK, Hoffman GE, Steiner RA, Tena-Sempere M. Sexual differentiation of Kiss1 gene expression in the brain of the rat. Endocrinology. 2007;148:1774–1783. doi: 10.1210/en.2006-1540. This study was the first to demonstrate the sexual differentiation of Kiss1 neurons in the rat AVPV and to show that sexual differentiation of Kiss1 neurons is influenced by postnatal sex steroid millieu. This study suggested that the sexual differentiation of AVPV Kiss1 neurons may underlie the sex-specific ability to produce an LH surge. [DOI] [PubMed] [Google Scholar]
- 2••.Ottem EN, Godwin JG, Krishnan S, Petersen SL. Dual-phenotype GABA/glutamate neurons in adult preoptic area: sexual dimorphism and function. J Neurosci. 2004;24:8097–8105. doi: 10.1523/JNEUROSCI.2267-04.2004. This article demonstrated sex differences in GABA/glutamate neurons in the rat AVPV. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Simerly RB. Organization and regulation of sexually dimorphic neuroendocrine pathways. Behav Brain Res. 1998;92:195–203. doi: 10.1016/s0166-4328(97)00191-5. [DOI] [PubMed] [Google Scholar]
- 4••.Kauffman AS. Coming of Age in the Kisspeptin Era: Sex differences, Development, and Puberty. Mol Cell Endocrinol. 2010 doi: 10.1016/j.mce.2010.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Clarkson J, d’Anglemont de Tassigny X, Moreno AS, Colledge WH, Herbison AE. Kisspeptin-GPR54 signaling is essential for preovulatory gonadotropin-releasing hormone neuron activation and the luteinizing hormone surge. J Neurosci. 2008;28:8691–8697. doi: 10.1523/JNEUROSCI.1775-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6•.Pineda R, Garcia-Galiano D, Roseweir A, Romero M, Sanchez-Garrido MA, Ruiz-Pino F, Morgan K, Pinilla L, Millar RP, Tena-Sempere M. Critical Roles of Kisspeptins in Female Puberty and Preovulatory Gonadotropin Surges as Revealed by a Novel Antagonist. Endocrinology. 2009 doi: 10.1210/en.2009-0803. This study found that administration of a kisspeptin antagonist delayed puberty and blocked the LH surge in female rats, providing convincing evidence directly linking kisspeptin to pubertal onset and the LH surge, two sexually dimorphic events. [DOI] [PubMed] [Google Scholar]
- 7.Simerly RB. Wired for reproduction: organization and development of sexually dimorphic circuits in the mammalian forebrain. Annu Rev Neurosci. 2002;25:507–536. doi: 10.1146/annurev.neuro.25.112701.142745. [DOI] [PubMed] [Google Scholar]
- 8.Morris JA, Jordan CL, Breedlove SM. Sexual differentiation of the vertebrate nervous system. Nat Neurosci. 2004;7:1034–1039. doi: 10.1038/nn1325. [DOI] [PubMed] [Google Scholar]
- 9.Jazin E, Cahill L. Sex differences in molecular neuroscience: from fruit flies to humans. Nat Rev Neurosci. 2010;11:9–17. doi: 10.1038/nrn2754. [DOI] [PubMed] [Google Scholar]
- 10.de Vries GJ, Sodersten P. Sex differences in the brain: the relation between structure and function. Horm Behav. 2009;55:589–596. doi: 10.1016/j.yhbeh.2009.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Forger NG. Control of cell number in the sexually dimorphic brain and spinal cord. J Neuroendocrinol. 2009;21:393–399. doi: 10.1111/j.1365-2826.2009.01825.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cooke B, Hegstrom CD, Villeneuve LS, Breedlove SM. Sexual differentiation of the vertebrate brain: principles and mechanisms. Front Neuroendocrinol. 1998;19:323–362. doi: 10.1006/frne.1998.0171. [DOI] [PubMed] [Google Scholar]
- 13.Hines M, Allen LS, Gorski RA. Sex differences in subregions of the medial nucleus of the amygdala and the bed nucleus of the stria terminalis of the rat. Brain Res. 1992;579:321–326. doi: 10.1016/0006-8993(92)90068-k. [DOI] [PubMed] [Google Scholar]
- 14.De Vries GJ, Panzica GC. Sexual differentiation of central vasopressin and vasotocin systems in vertebrates: different mechanisms, similar endpoints. Neuroscience. 2006;138:947–955. doi: 10.1016/j.neuroscience.2005.07.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hutton LA, Gu G, Simerly RB. Development of a sexually dimorphic projection from the bed nuclei of the stria terminalis to the anteroventral periventricular nucleus in the rat. J Neurosci. 1998;18:3003–3013. doi: 10.1523/JNEUROSCI.18-08-03003.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bleier R, Byne W, Siggelkow I. Cytoarchitectonic sexual dimorphisms of the medial preoptic and anterior hypothalamic areas in guinea pig, rat, hamster, and mouse. J Comp Neurol. 1982;212:118–130. doi: 10.1002/cne.902120203. [DOI] [PubMed] [Google Scholar]
- 17.Simerly RB, Swanson LW, Gorski RA. The distribution of monoaminergic cells and fibers in a periventricular preoptic nucleus involved in the control of gonadotropin release: Immunohistochemical evidence for a dopaminergic sexual dimorphism. Brain Research. 1985;330:55–64. doi: 10.1016/0006-8993(85)90007-1. [DOI] [PubMed] [Google Scholar]
- 18.Walker DL, Toufexis DJ, Davis M. Role of the bed nucleus of the stria terminalis versus the amygdala in fear, stress, and anxiety. European Journal of Pharmacology. 2003;463:199–216. doi: 10.1016/s0014-2999(03)01282-2. [DOI] [PubMed] [Google Scholar]
- 19.Arnold AP. The organizational-activational hypothesis as the foundation for a unified theory of sexual differentiation of all mammalian tissues. Hormones and Behavior. 2009;55:570–578. doi: 10.1016/j.yhbeh.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Burns-Cusato M, Scordalakes EM, Rissman EF. Of mice and missing data: what we know (and need to learn) about male sexual behavior. Physiology & Behavior. 2004;83:217–232. doi: 10.1016/j.physbeh.2004.08.015. [DOI] [PubMed] [Google Scholar]
- 21.Ojeda SRSMK. Puberty in the rat. In: Neill JD, editor. The Physiology of Reproduction. 3. San Diego: Academic Elsevier; 2006. pp. 2061–2126. [Google Scholar]
- 22.Plant TM, Witchel SM. Puberty in non-human primates and humans. In: Neill JD, editor. Knobil and Neill’s Physiology of Reproduction. Elsevier; 2006. pp. 2177–2230. [Google Scholar]
- 23.Clarkson J, Herbison AE. Oestrogen, Kisspeptin, GPR54 and the Pre-Ovulatory Luteinising Hormone Surge. Journal of Neuroendocrinology. 2009;21:305–311. doi: 10.1111/j.1365-2826.2009.01835.x. [DOI] [PubMed] [Google Scholar]
- 24.Cesario SK, Hughes LA. Precocious puberty: a comprehensive review of literature. J Obstet Gynecol Neonatal Nurs. 2007;36:263–274. doi: 10.1111/j.1552-6909.2007.00145.x. [DOI] [PubMed] [Google Scholar]
- 25.Fechner A, Fong S, McGovern P. A review of Kallmann syndrome: genetics, pathophysiology, and clinical management. Obstet Gynecol Surv. 2008;63:189–194. doi: 10.1097/OGX.0b013e3181641278. [DOI] [PubMed] [Google Scholar]
- 26.Goldman BD, Gorski RA. Effects of gonadal steroids on the secretion of LH and FSH in neonatal rats. Endocrinology. 1971;89:112–115. doi: 10.1210/endo-89-1-112. [DOI] [PubMed] [Google Scholar]
- 27.Kauffman AS. Sexual differentiation and the Kiss1 system: hormonal and developmental considerations. Peptides. 2009;30:83–93. doi: 10.1016/j.peptides.2008.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Herbison AE. Estrogen positive feedback to gonadotropin-releasing hormone (GnRH) neurons in the rodent: The case for the rostral periventricular area of the third ventricle (RP3V) Brain Research Reviews. 2008;57:277–287. doi: 10.1016/j.brainresrev.2007.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kauffman AS, Clifton DK, Steiner RA. Emerging ideas about kisspeptin- GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Neurosci. 2007;30:504–511. doi: 10.1016/j.tins.2007.08.001. [DOI] [PubMed] [Google Scholar]
- 30••.Forger NG, Rosen GJ, Waters EM, Jacob D, Simerly RB, de Vries GJ. Deletion of Bax eliminates sex differences in the mouse forebrain. Proc Natl Acad Sci U S A. 2004;101:13666–13671. doi: 10.1073/pnas.0404644101. This study implicated apoptosis in the sexual differentiation of the AVPV. The investigators showed that the sexual differentiation of cell number in the AVPV and BNST is eliminated in knockout mice lacking the pro-apoptotic gene Bax. whereas AVPV neurons expressing TH were not affected by Bax deletion. Thus, although total AVPV cell number may be dependent on developmental Bax-dependent cell death, the sexual differentiation of subsets of neurons within the AVPV may arise via different mechanisms. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Simerly RB, Zee MC, Pendleton JW, Lubahn DB, Korach KS. Estrogen receptor-dependent sexual differentiation of dopaminergic neurons in the preoptic region of the mouse. Proc Natl Acad Sci U S A. 1997;94:14077–14082. doi: 10.1073/pnas.94.25.14077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32•.Clarkson J, Herbison AE. Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology. 2006;147:5817–5825. doi: 10.1210/en.2006-0787. This study found that AVPV kisspeptin protein levels, as measured via immunohistochemistry, are higher in intact female than intact male mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33••.Homma T, Sakakibara M, Yamada S, Kinoshita M, Iwata K, Tomikawa J, Kanazawa T, Matsui H, Takatsu Y, Ohtaki T, et al. Significance of Neonatal Testicular Sex Steroids to Defeminize Anteroventral Periventricular Kisspeptin Neurons and the GnRH/LH Surge System in Male Rats. Biol Reprod. 2009 doi: 10.1095/biolreprod.109.078311. This study determined that the rat AVPV kisspeptin sex difference is induced postnatally by the aromatization of testosterone to estradiol in males, leading to less kisspeptin neurons in adult males than females. [DOI] [PubMed] [Google Scholar]
- 34.Kauffman AS, Park JH, McPhie-Lalmansingh AA, Gottsch ML, Bodo C, Hohmann JG, Pavlova MN, Rohde AD, Clifton DK, Steiner RA, et al. The kisspeptin receptor GPR54 is required for sexual differentiation of the brain and behavior. J Neurosci. 2007;27:8826–8835. doi: 10.1523/JNEUROSCI.2099-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brann DW, Mahesh VB. Glutamate: a major neuroendocrine excitatory signal mediating steroid effects on gonadotropin secretion. J Steroid Biochem Mol Biol. 1995;53:325–329. doi: 10.1016/0960-0760(95)00070-g. [DOI] [PubMed] [Google Scholar]
- 36.Han SK, Todman MG, Herbison AE. Endogenous GABA release inhibits the firing of adult gonadotropin-releasing hormone neurons. Endocrinology. 2004;145:495–499. doi: 10.1210/en.2003-1333. [DOI] [PubMed] [Google Scholar]
- 37.DeFazio RA, Heger S, Ojeda SR, Moenter SM. Activation of A-type gamma-aminobutyric acid receptors excites gonadotropin-releasing hormone neurons. Mol Endocrinol. 2002;16:2872–2891. doi: 10.1210/me.2002-0163. [DOI] [PubMed] [Google Scholar]
- 38.Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, Brezillon S, Tyldesley R, Suarez-Huerta N, Vandeput F, et al. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276:34631–34636. doi: 10.1074/jbc.M104847200. [DOI] [PubMed] [Google Scholar]
- 39.Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ, Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P, et al. AXOR12, a novel human G protein-coupled receptor, activated by the peptide KiSS-1. J Biol Chem. 2001;276:28969–28975. doi: 10.1074/jbc.M102743200. [DOI] [PubMed] [Google Scholar]
- 40.Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A, Kanehashi K, Terao Y, Kumano S, Takatsu Y, Masuda Y, et al. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature. 2001;411:613–617. doi: 10.1038/35079135. [DOI] [PubMed] [Google Scholar]
- 41.Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr, Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, et al. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349:1614–1627. doi: 10.1056/NEJMoa035322. [DOI] [PubMed] [Google Scholar]
- 42.Funes S, Hedrick JA, Vassileva G, Markowitz L, Abbondanzo S, Golovko A, Yang S, Monsma FJ, Gustafson EL. The KiSS-1 receptor GPR54 is essential for the development of the murine reproductive system. Biochem Biophys Res Commun. 2003;312:1357–1363. doi: 10.1016/j.bbrc.2003.11.066. [DOI] [PubMed] [Google Scholar]
- 43.de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100:10972–10976. doi: 10.1073/pnas.1834399100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Navarro VM, Castellano JM, Fernandez-Fernandez R, Tovar S, Roa J, Mayen A, Nogueiras R, Vazquez MJ, Barreiro ML, Magni P, et al. Characterization of the potent luteinizing hormone-releasing activity of KiSS-1 peptide, the natural ligand of GPR54. Endocrinology. 2005;146:156–163. doi: 10.1210/en.2004-0836. [DOI] [PubMed] [Google Scholar]
- 45.Navarro VM, Castellano JM, Fernandez-Fernandez R, Tovar S, Roa J, Mayen A, Barreiro ML, Casanueva FF, Aguilar E, Dieguez C, et al. Effects of KiSS-1 peptide, the natural ligand of GPR54, on follicle-stimulating hormone secretion in the rat. Endocrinology. 2005;146:1689–1697. doi: 10.1210/en.2004-1353. [DOI] [PubMed] [Google Scholar]
- 46.Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM, Cunningham MJ, Gottsch ML, Clifton DK, Steiner RA. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology. 2004;80:264–272. doi: 10.1159/000083140. [DOI] [PubMed] [Google Scholar]
- 47.Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich SK, Clifton DK, Steiner RA, Herbison AE. Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci. 2005;25:11349–11356. doi: 10.1523/JNEUROSCI.3328-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, Thresher RR, Malinge I, Lomet D, Carlton MB, et al. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A. 2005;102:1761–1766. doi: 10.1073/pnas.0409330102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Smith JT, Popa SM, Clifton DK, Hoffman GE, Steiner RA. Kiss1 neurons in the forebrain as central processors for generating the preovulatory luteinizing hormone surge. J Neurosci. 2006;26:6687–6694. doi: 10.1523/JNEUROSCI.1618-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Robertson JL, Clifton DK, de la Iglesia HO, Steiner RA, Kauffman AS. Circadian regulation of Kiss1 neurons: implications for timing the preovulatory gonadotropin-releasing hormone/luteinizing hormone surge. Endocrinology. 2009;150:3664–3671. doi: 10.1210/en.2009-0247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kauffman AS, Navarro VM, Kim J, Clifton DK, Steiner RA. Sex differences in the regulation of Kiss1/NKB neurons in juvenile mice: implications for the timing of puberty. Am J Physiol Endocrinol Metab. 2009;297:E1212–1221. doi: 10.1152/ajpendo.00461.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Simerly RB, Swanson LW, Handa RJ, Gorski RA. Influence of perinatal androgen on the sexually dimorphic distribution of tyrosine hydroxylase-immunoreactive cells and fibers in the anteroventral periventricular nucleus of the rat. Neuroendocrinology. 1985;40:501–510. doi: 10.1159/000124122. [DOI] [PubMed] [Google Scholar]
- 53.Smith JT, Clifton DK, Steiner RA. Regulation of the neuroendocrine reproductive axis by kisspeptin-GPR54 signaling. Reproduction. 2006;131:623–630. doi: 10.1530/rep.1.00368. [DOI] [PubMed] [Google Scholar]
- 54.Adachi S, Yamada S, Takatsu Y, Matsui H, Kinoshita M, Takase K, Sugiura H, Ohtaki T, Matsumoto H, Uenoyama Y, et al. Involvement of anteroventral periventricular metastin/kisspeptin neurons in estrogen positive feedback action on luteinizing hormone release in female rats. J Reprod Dev. 2007;53:367–378. doi: 10.1262/jrd.18146. [DOI] [PubMed] [Google Scholar]
- 55.Bateman HL, Patisaul HB. Disrupted female reproductive physiology following neonatal exposure to phytoestrogens or estrogen specific ligands is associated with decreased GnRH activation and kisspeptin fiber density in the hypothalamus. Neurotoxicology. 2008;29:988–997. doi: 10.1016/j.neuro.2008.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.McCarthy MM, Auger AP, Bale TL, De Vries GJ, Dunn GA, Forger NG, Murray EK, Nugent BM, Schwarz JM, Wilson ME. The epigenetics of sex differences in the brain. J Neurosci. 2009;29:12815–12823. doi: 10.1523/JNEUROSCI.3331-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57••.Murray EK, Hien A, de Vries GJ, Forger NG. Epigenetic control of sexual differentiation of the bed nucleus of the stria terminalis. Endocrinology. 2009;150:4241–4247. doi: 10.1210/en.2009-0458. This study showed that altering histone deacetylation during the postnatal critical period was sufficient to block the masculinization of the rodent BNST. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhang JM, Konkle AT, Zup SL, McCarthy MM. Impact of sex and hormones on new cells in the developing rat hippocampus: a novel source of sex dimorphism? Eur J Neurosci. 2008;27:791–800. doi: 10.1111/j.1460-9568.2008.06073.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gotsiridze T, Kang N, Jacob D, Forger NG. Development of sex differences in the principal nucleus of the bed nucleus of the stria terminalis of mice: role of Bax-dependent cell death. Dev Neurobiol. 2007;67:355–362. doi: 10.1002/dneu.20353. [DOI] [PubMed] [Google Scholar]
- 60.Krishnan S, Intlekofer KA, Aggison LK, Petersen SL. Central role of TRAF-interacting protein in a new model of brain sexual differentiation. Proc Natl Acad Sci U S A. 2009;106:16692–16697. doi: 10.1073/pnas.0906293106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Holmes MM, McCutcheon J, Forger NG. Sex differences in NeuN- and androgen receptor-positive cells in the bed nucleus of the stria terminalis are due to Bax-dependent cell death. Neuroscience. 2009;158:1251–1256. doi: 10.1016/j.neuroscience.2008.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007;26:1324–1337. doi: 10.1038/sj.onc.1210220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tsukahara S, Kakeyama M, Toyofuku Y. Sex differences in the level of Bcl-2 family proteins and caspase-3 activation in the sexually dimorphic nuclei of the preoptic area in postnatal rats. J Neurobiol. 2006;66:1411–1419. doi: 10.1002/neu.20276. [DOI] [PubMed] [Google Scholar]
- 64•.Waters EM, Simerly RB. Estrogen induces caspase-dependent cell death during hypothalamic development. J Neurosci. 2009;29:9714–9718. doi: 10.1523/JNEUROSCI.0135-09.2009. This work provided evidence that estradiol can induce mitochondrial-dependent cell death in the AVPV, correlating with a reduction in TH neurons. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.de Vries GJ, Jardon M, Reza M, Rosen GJ, Immerman E, Forger NG. Sexual differentiation of vasopressin innervation of the brain: cell death versus phenotypic differentiation. Endocrinology. 2008;149:4632–4637. doi: 10.1210/en.2008-0448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007;128:635–638. doi: 10.1016/j.cell.2007.02.006. [DOI] [PubMed] [Google Scholar]
- 67•.Kurian JR, Olesen KM, Auger AP. Sex Differences in Epigenetic Regulation of the Estrogen Receptor-α Promoter within the Developing Preoptic Area. Endocrinology. 2010 doi: 10.1210/en.2009-0649. (In Press) This study identified a sex difference in DNA methylation of the estrogen receptor-α (ERα) promoter in the pre-optic area of rats (higher methylation in males than females), and this was associated with sexually dimorphic ERα gene expression. Additionally, variations in maternal care and early estradiol exposure influenced sexually dimorphic ERα promoter methylation. [DOI] [PMC free article] [PubMed] [Google Scholar]
