201
|
Neural mechanisms underlying sex-specific behaviors in vertebrates. Curr Opin Neurobiol 2008; 17:675-83. [PMID: 18343651 PMCID: PMC2483511 DOI: 10.1016/j.conb.2008.01.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 01/25/2008] [Accepted: 01/29/2008] [Indexed: 01/28/2023]
Abstract
From invertebrates to humans, males and females of a given species display identifiable differences in behaviors, mostly but not exclusively pertaining to sexual and social behaviors. Within a species, individuals preferentially exhibit the set of behaviors that is typical of their sex. These behaviors include a wide range of coordinated and genetically pre-programmed social and sexual displays that ensure successful reproductive strategies and the survival of the species. What are the mechanisms underlying sex-specific brain function? Although sexually dimorphic behaviors represent the most extreme examples of behavioral variability within a species, the basic principles underlying the sex specificity of brain activity are largely unknown. Moreover, with few exceptions, the quest for fundamental differences in male and female brain structures and circuits that would parallel that of sexual behaviors and peripheral organs has so far uncovered modest quantitative rather than the expected clear qualitative differences. As will be detailed in this review, recent advances have directly challenged the established notion of the unique role of steroid hormones in organizing and activating male- and female-specific brain circuits and have uncovered new mechanisms underlying the neural control of sex-specific behaviors.
Collapse
|
202
|
Jahagirdar V, Quadros PS, Wagner CK. Endogenous oestradiol regulates progesterone receptor expression in the brain of female rat fetuses: what is the source of oestradiol? J Neuroendocrinol 2008; 20:359-65. [PMID: 18208546 DOI: 10.1111/j.1365-2826.2008.01647.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Testosterone secreted by male testes during fetal development is aromatized to oestradiol (E(2)) or reduced to the androgen, dihydrotestosteorne (DHT), within specific tissues. The female brain is assumed to develop in the relative absence of gonadal steroid hormones, as the ovary is steroidogenically quiescent until later in postnatal life. However, the proximity of a female fetus to male littermates in utero can increase her exposure to testosterone, and thereby its metabolites. To date, it is has been difficult to dissociate the effects of male-derived E(2) from those of DHT on the developing female brain. In the present study, anogential distance (AGD) in females was used as an androgen-dependent bioassay, whereas progesterone receptor (PR) expression within the medial preoptic nucleus (MPN) was used as an E-dependent measure. Pregnant dams received the aromatase inhibitor, 1,4,6-androstatriene-3,17-dione (ATD), or vehicle from embryonic day 16 (ED16) to ED21. On ED22, AGD and PR-immunoreactivity (-ir) were measured in females that had zero, one, or two males (0-2M) or females that had three, four, or five males (3-5M) in the uterine horn. AGD was significantly greater in 3-5M females compared to 0-2M females, suggesting that male littermates are the source of androgenic exposure in the female fetus. ATD treatment significantly decreased PR-ir in the MPN, demonstrating E(2) regulation of PR. However, the total number of males in the uterine horn did not effect PR expression. There was no correlation between PR-ir and AGD, suggesting that these measures are influenced independently. Together, these results suggest that although male littermates provide a significant source of androgens to female fetuses, the amount of E(2) aromatized from male-derived testosterone may not be the only biologically relevant source of androgens or E(2). Alternative sources of E(2) may be essential in ensuring the normal development of the female brain.
Collapse
Affiliation(s)
- V Jahagirdar
- Department of Psychology and Centre for Neuroscience Research, University at Albany, SUNY, Albany, NY 12222, USA
| | | | | |
Collapse
|
203
|
Morissette M, Le Saux M, D'Astous M, Jourdain S, Al Sweidi S, Morin N, Estrada-Camarena E, Mendez P, Garcia-Segura LM, Di Paolo T. Contribution of estrogen receptors alpha and beta to the effects of estradiol in the brain. J Steroid Biochem Mol Biol 2008; 108:327-38. [PMID: 17936613 DOI: 10.1016/j.jsbmb.2007.09.011] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Clinical and experimental studies show a modulatory role of estrogens in the brain and suggest their beneficial action in mental and neurodegenerative diseases. The estrogen receptors ERalpha and ERbeta are present in the brain and their targeting could bring selectivity and reduced risk of cancer. Implication of ERs in the effect of estradiol on dopamine, opiate and glutamate neurotransmission is reviewed. The ERalpha agonist, PPT, is shown as estradiol to modulate hippocampal NMDA receptors and AMPA receptors in cortex and striatum of ovariectomized rats whereas the ERbeta agonist DPN is inactive. Striatal DPN activity suggests implication of ERbeta in estradiol modulation of D2 receptors and transporters in ovariectomized rats and is supported by the lack of effect of estradiol in ERbeta knockout (ERKObeta) mice. Both ERalpha and ERbeta agonists modulate striatal preproenkephalin (PPE) gene expression in ovariectomized rats. In male mice PPT protects against MPTP toxicity to striatal dopamine; this implicates Akt/GSK3beta signaling and the apoptotic regulators Bcl2 and Bad. This suggests a role for ERalpha in striatal dopamine neuroprotection. ERKOalpha mice are more susceptible to MPTP toxicity and not protected by estradiol; differences in ERKObeta mice are subtler. These results suggest therapeutic potential for the brain of ER specific agonists.
Collapse
Affiliation(s)
- M Morissette
- Molecular Endocrinology and Oncology Research Center, Medical Center and Faculty of Pharmacy, Laval University, 2705 Laurier Boulevard, Sainte-Foy, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Steroid abnormalities and the developing brain: declarative memory for emotionally arousing and neutral material in children with congenital adrenal hyperplasia. Psychoneuroendocrinology 2008; 33:238-45. [PMID: 18162329 PMCID: PMC2262834 DOI: 10.1016/j.psyneuen.2007.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 11/02/2007] [Accepted: 11/02/2007] [Indexed: 11/21/2022]
Abstract
Steroid hormones modulate memory in animals and human adults. Little is known on the developmental effects of these hormones on the neural networks underlying memory. Using Congenital adrenal hyperplasia (CAH) as a naturalistic model of early steroid abnormalities, this study examines the consequences of CAH on memory and its neural correlates for emotionally arousing and neutral material in children. Seventeen patients with CAH and 17 age- and sex-matched healthy children (ages 12-14 years) completed the study. Subjects were presented positive, negative and neutral pictures. Memory recall occurred about 30min after viewing the pictures. Children with CAH showed memory deficits for negative pictures compared to healthy children (p<0.01). There were no group differences on memory performance for either positive or neutral pictures (p>0.1). In patients, 24h urinary-free cortisol levels (reflecting glucocorticoid replacement therapy) and testosterone levels were not associated with memory performance. These findings suggest that early steroid imbalances affect memory for negative material in children with CAH. Such memory impairments may result from abnormal brain organization and function following hormonal dysfunction during critical periods of development.
Collapse
|
205
|
Luo H, Liu J, Kang D, Cui S. Ontogeny of estrogen receptor alpha, estrogen receptor beta and androgen receptor, and their co-localization with Islet-1 in the dorsal root ganglia of sheep fetuses during gestation. Histochem Cell Biol 2008; 129:525-33. [PMID: 18204852 DOI: 10.1007/s00418-008-0380-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2008] [Indexed: 02/02/2023]
Abstract
The aims of the present study were to detect the ontogeny of estrogen receptor (ERalpha and ERbeta) and androgen receptor (AR) expressions and their co-localization with Islet-1 in the developing dorsal root ganglia (DRG) of sheep fetuses by immunohistochemistry. From the single staining results, the ERalpha immunoreactivity (ERalpha-ir), ERbeta immunoreactivity (ERbeta-ir) and AR immunoreactivity (AR-ir) was first detected at days 90, 120 and 90 of gestation, respectively. From days 90 to 120, ERalpha and AR were consistently detected in the nuclei of DRG neurons and the relative percentage (approximately 60%) of ERalpha-ir or AR-ir cells did not change significantly. Moreover, there was no change in ERalpha expression, while a dramatic loss of AR expression was observed at birth. From day 120 of gestation to birth, very few neurons (approximately 8%) showed nuclear ERbeta immunoreactivity. The dual staining results showed that Islet-1 was co-localized with ERalpha, ERbeta or AR in the nuclei of DRG neurons with various frequencies, and over 70% ERalpha-ir, ERbeta-ir or AR-ir cells contained Islet-1. These results imply that ERs, AR and Islet-1 may be important in regulating the differentiation and functional maintenance of some phenotypes of DRG neurons after mid-gestation in the sheep fetus.
Collapse
Affiliation(s)
- Haoshu Luo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100094, People's Republic of China
| | | | | | | |
Collapse
|
206
|
Santos EM, Kille P, Workman VL, Paull GC, Tyler CR. Sexually dimorphic gene expression in the brains of mature zebrafish. Comp Biochem Physiol A Mol Integr Physiol 2008; 149:314-24. [PMID: 18289901 DOI: 10.1016/j.cbpa.2008.01.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 01/09/2008] [Accepted: 01/11/2008] [Indexed: 10/22/2022]
Abstract
The molecular signalling pathways mediating sexual dimorphism have principally been investigated in the gonads, and to a lesser extent in other organs. The brain plays a central role in coordinating sexual function, including the regulation of reproductive development, maturation and sexual behaviour in both sexes. In this study, we investigated sex-related differences in gene expression in the brains of breeding zebrafish (Danio rerio) to establish a greater understanding of the sex-specific physiology of the brain in lower vertebrates. The brain transcriptomic profiles of males and females were interrogated to identify the genes showing sexually dimorphic gene expression. 42 genes were differentially expressed between the sexes, from which 18 genes were over-expressed in males and 24 genes were over-expressed in females. In males, these included deiodinase, iodothyronine, type II and ribosomal protein S8, and in females, superoxide dismutase [Cu-Zn], sprouty-4, frizzled 10 and testis enhanced gene transcript. Estrogen responsive elements were found in the regulatory regions for 3 genes over-expressed in males and 7 genes over-expressed in females. We have demonstrated the existence of dimorphic patterns of gene expression in the brain of a sexually mature, non-mammalian, vertebrate model, with implications for studies into reproduction and chemical disruption of brain function.
Collapse
Affiliation(s)
- Eduarda M Santos
- School of Biosciences, University of Exeter, Prince of Wales Road, Exeter, EX4 4PS, UK
| | | | | | | | | |
Collapse
|
207
|
Endo D, Murakami S, Akazome Y, Park MK. Sex difference in Ad4BP/SF-1 mRNA expression in the chick-embryo brain before gonadal sexual differentiation. Zoolog Sci 2008; 24:877-82. [PMID: 17960991 DOI: 10.2108/zsj.24.877] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 05/09/2007] [Indexed: 11/17/2022]
Abstract
Sexual differentiation in the amniote brain is believed to be regulated by gonadal sex steroid hormones. Recently, however, the possibility of brain-autonomous sexual differentiation in avian and reptilian species has been reported. We conducted here an expressional analysis of genes related to sex steroid hormones in the chick-embryo brain before gonadal sexual differentiation. Female-specific P450 aromatase expression in the gonad was observed at day 6.5 of incubation, as previously reported, whereas the mRNAs of cholesterol side-chain cleavage enzyme, androgen receptor, and estrogen receptors alpha and beta were clearly expressed in all brain samples of both male and female embryos from day 4.5 of incubation. P450 aromatase was expressed in some brain samples before day 5.5 of incubation and in all brain samples after day 6 of incubation. The mRNA of Ad4BP/SF-1, a transcription factor that regulates steroidogenic enzymes, showed higher expression levels in the male brain than in the female brain at day 5.5 of incubation. This gene was expressed in the ventromedial hypothalamic nucleus, a region important for reproductive behavior. Embryonic Ad4BP/SF-1 expression is reported to play an important role in the formation of this region. These results therefore suggest the involvement of a sex steroid hormone signaling system in brain-autonomous sexual differentiation.
Collapse
Affiliation(s)
- Daisuke Endo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
208
|
Endo D, Kanaho YI, Park MK. Expression of sex steroid hormone-related genes in the embryo of the leopard gecko. Gen Comp Endocrinol 2008; 155:70-8. [PMID: 17543964 DOI: 10.1016/j.ygcen.2007.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Revised: 04/04/2007] [Accepted: 04/09/2007] [Indexed: 11/29/2022]
Abstract
Sex steroid hormones are known to play a central role in vertebrate sex determination and differentiation. However, the tissues in which they are produced or received during development, especially around the period of sex determination of the gonads, have rarely been investigated. In this study, we identified the cDNA sequence, including the full-length of the coding region of cholesterol side-chain cleavage enzyme (P450scc), from the leopard gecko; a lizard with temperature-dependent sex determination. Embryonic expression analysis of two steroidogenic enzymes, P450scc and P450 aromatase (P450arom), and four sex steroid hormone receptors, androgen receptor, estrogen receptor alpha and beta, and progesterone receptor, was subsequently conducted. mRNA expression of both steroidogenic enzymes was observed in the brain and gonads prior to the temperature-sensitive period of sex determination. The mRNAs of the four sex steroid hormone receptors were also detected in the brain and gonads at all stages examined. These results suggest the existence of a gonad-independent sex steroid hormone signaling system in the developing leopard gecko brain.
Collapse
Affiliation(s)
- Daisuke Endo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | | | | |
Collapse
|
209
|
Yuge K, Ikeo K, Gojobori T. Evolutionary origin of sex-related genes in the mouse brain. Gene 2007; 406:108-12. [PMID: 17728078 DOI: 10.1016/j.gene.2007.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 06/05/2007] [Accepted: 06/28/2007] [Indexed: 10/23/2022]
Abstract
With the aim of elucidating the evolutionary process of sexual dimorphism in the brain at the molecular level, we conducted genomic comparisons of a set of genes expressed in a sexually different manner in the mouse brain with all genes from other species of eukaryotes. First, seventeen protein-coding genes whose levels of mRNA expression in the brain differed between male and female mice have been known according to the currently available microarray data, and we designated these genes operationally as "sex-related genes in the mouse brain". Next, we estimated the time when these sex-related genes in the mouse brain emerged in the evolutionary process of eukaryotes by examining the presence or absence of the orthologues in the 26 eukaryotic species whose genome sequences are available. As a result, we found that the ten sex-related genes in the mouse brain emerged after the divergence of urochordates and mammals whereas the other seven sex-related genes in the mouse brain emerged before the divergence of urochordates and mammals. In particular, five sex-related genes out of the ten genes in the mouse brain emerged just before the appearance of bony fish which have phenotypic sexual dimorphism in the brain. Interestingly, three of these five sex-related genes that emerged during this period were classified into the "protein binding" function category. Moreover, all of these three genes were expected to have the functions that are related to cell-cell communications in the brain according to the gene expression patterns and/or functional information of these genes. These findings suggest that the orthologues of the sex-related genes in the mouse brain that emerged just before the divergence of bony fish might have essential roles in the evolution of the sexual dimorphism in the brain forming protein-protein interactions.
Collapse
Affiliation(s)
- Kazuya Yuge
- Center for Information Biology and DDBJ, National Institute of Genetics, Mishima, Japan
| | | | | |
Collapse
|
210
|
Steroid hormones alter neuroanatomy and aggression independently in the tree lizard. Physiol Behav 2007; 93:492-501. [PMID: 17996258 DOI: 10.1016/j.physbeh.2007.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 10/01/2007] [Accepted: 10/08/2007] [Indexed: 10/22/2022]
Abstract
Steroid hormones effect changes in both neuroanatomy and aggressive behavior in animals of various taxa. However, whether changes in neuroanatomy directly underlie changes in aggression is unknown. We investigate this relationship among steroid hormones, neuroanatomy, and aggression in a free-living vertebrate with a relatively simple nervous system, the tree lizard (Urosaurus ornatus). Weiss and Moore [1] manipulated testosterone and progesterone levels in adult male tree lizards and found that both hormones facilitated aggressive behavior toward a conspecific. In this study, we examined the brains of a subset of these animals to determine whether changes in limbic morphology were associated with hormone-induced changes in aggressive behavior. Specifically, we tested the hypothesis that testosterone and/or progesterone cause changes in neural morphology that are necessary for the expression of testosterone's effects on aggressive behavior. We found that both hormones increased aggression; however, only testosterone induced changes in neuroanatomy. Testosterone increased the size of both the amygdala and nucleus sphericus. However, we could detect no individual correlations between neuroanatomy and aggression levels suggesting that the observed large-scale changes in neuroanatomy are not precisely reflective of changes in mechanisms underlying aggression.
Collapse
|
211
|
Panzica GC, Viglietti-Panzica C, Mura E, Quinn MJ, Lavoie E, Palanza P, Ottinger MA. Effects of xenoestrogens on the differentiation of behaviorally-relevant neural circuits. Front Neuroendocrinol 2007; 28:179-200. [PMID: 17868795 DOI: 10.1016/j.yfrne.2007.07.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 06/11/2007] [Accepted: 07/12/2007] [Indexed: 11/18/2022]
Abstract
It has become increasingly clear that environmental chemicals have the capability of impacting endocrine function. Moreover, these endocrine disrupting chemicals (EDCs) have long term consequences on adult reproductive function, especially if exposure occurs during embryonic development thereby affecting sexual differentiation. Of the EDCs, most of the research has been conducted on the effects of estrogen active compounds. Although androgen active compounds are also present in the environment, much less information is available about their action. However, in the case of xenoestrogens, there is mounting evidence for long-term consequences of early exposure at a range of doses. In this review, we present data relative to two widely used animal models: the mouse and the Japanese quail. These two species long have been used to understand neural, neuroendocrine, and behavioral components of reproduction and are therefore optimal models to understand how these components are altered by precocious exposure to EDCs. In particular we discuss effects of bisphenol A and methoxychlor on the dopaminergic and noradrenergic systems in rodents and the impact of these alterations. In addition, the effects of embryonic exposure to diethylstilbestrol, genistein or ethylene,1,1-dichloro-2,2-bis(p-chlorophenyl) is reviewed relative to behavioral impairment and associated alterations in the sexually dimorphic parvocellular vasotocin system in quail. We point out how sexually dimorphic behaviors are particularly useful to verify adverse developmental consequences produced by chemicals with endocrine disrupting properties, by examining either reproductive or non-reproductive behaviors.
Collapse
|
212
|
Kauffman AS, Clifton DK, Steiner RA. Emerging ideas about kisspeptin- GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Neurosci 2007; 30:504-11. [PMID: 17904653 DOI: 10.1016/j.tins.2007.08.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Revised: 07/31/2007] [Accepted: 08/01/2007] [Indexed: 11/17/2022]
Abstract
Neurons that produce gonadotropin-releasing hormone (GnRH) drive the reproductive axis, but the molecular and cellular mechanisms by which hormonal and environmental signals regulate GnRH secretion remain poorly understood. Kisspeptins are products of the Kiss1 gene, and the interaction of kisspeptin and its receptor GPR54 plays a crucial role in governing the onset of puberty and adult reproductive function. This review discusses the latest ideas about kisspeptin-GPR54 signaling in the neuroendocrine regulation of reproduction, with special emphasis on the role of Kiss1 and kisspeptin in the negative and positive feedback control of gonadotropin secretion by sex steroids, timing of puberty onset, sexual differentiation of the brain and photoperiodic regulation of seasonal reproduction.
Collapse
|
213
|
Kauffman AS, Park JH, McPhie-Lalmansingh AA, Gottsch ML, Bodo C, Hohmann JG, Pavlova MN, Rohde AD, Clifton DK, Steiner RA, Rissman EF. The kisspeptin receptor GPR54 is required for sexual differentiation of the brain and behavior. J Neurosci 2007; 27:8826-35. [PMID: 17699664 PMCID: PMC6672184 DOI: 10.1523/jneurosci.2099-07.2007] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
GPR54 is a G-protein-coupled receptor, which binds kisspeptins and is widely expressed throughout the brain. Kisspeptin-GPR54 signaling has been implicated in the regulation of pubertal and adulthood gonadotropin-releasing hormone (GnRH) secretion, and mutations or deletions of GPR54 cause hypogonadotropic hypogonadism in humans and mice. Other reproductive roles for kisspeptin-GPR54 signaling, including the regulation of developmental GnRH secretion or sexual behavior in adults, have not yet been explored. Using adult wild-type (WT) and GPR54 knock-out (KO) mice, we first tested whether kisspeptin-GPR54 signaling is necessary for male and female sexual behaviors. We found that hormone-replaced gonadectomized GPR54 KO males and females displayed appropriate gender-specific adult sexual behaviors. Next, we examined whether GPR54 signaling is required for proper display of olfactory-mediated partner preference behavior. Testosterone-treated WT males preferred stimulus females rather than males, whereas similarly treated WT females and GPR54 KO males showed no preference for either sex. Because olfactory preference is sexually dimorphic and organized during development by androgens, we assessed whether GPR54 signaling is essential for sexual differentiation of other sexually dimorphic traits. Interestingly, adult testosterone-treated GPR54 KO males displayed "female-like" numbers of tyrosine hydroxylase-immunoreactive and Kiss1 mRNA-containing neurons in the anteroventral periventricular nucleus and likewise possessed fewer motoneurons in the spino-bulbocavernosus nucleus than did WT males. Our findings indicate that kisspeptin-GPR54 signaling is not required for male or female copulatory behavior, provided there is appropriate adulthood hormone replacement. However, GPR54 is necessary for proper male-like development of several sexually dimorphic traits, likely by regulating GnRH-mediated androgen secretion during "critical windows" in perinatal development.
Collapse
Affiliation(s)
- Alexander S Kauffman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Gioiosa L, Fissore E, Ghirardelli G, Parmigiani S, Palanza P. Developmental exposure to low-dose estrogenic endocrine disruptors alters sex differences in exploration and emotional responses in mice. Horm Behav 2007; 52:307-16. [PMID: 17568585 DOI: 10.1016/j.yhbeh.2007.05.006] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Revised: 05/04/2007] [Accepted: 05/04/2007] [Indexed: 11/30/2022]
Abstract
Estrogenic endocrine disruptors (EEDs) are naturally occurring or man-made compounds present in the environment that are able to bind to estrogen receptors and interfere with normal cellular development in target organs and tissues. There is mounting evidence that EEDs can interfere with the processes of sexual differentiation of brain and behavior in different animal models. We investigated the effects of maternal exposure to EEDs, at concentrations within the range of human exposure and not patently teratogenic, on behavioral responses of male and female house mice (Mus musculus domesticus) before and after puberty. Pregnant dams were trained to spontaneously drink daily doses of corn oil with or without the estrogenic plastic derivative, bisphenol A (BPA 10 microg/kg), or the estrogenic insecticide methoxychlor (MXC 20 microg/kg) from gestation day 11 to postpartum day 8. Their male and female offspring were examined at different ages to examine several components of explorative and emotional behaviors in 3 experimental paradigms: a novelty test before puberty and, as adults, a free-exploratory open-field test and the elevated plus maze test. The main results are sex differences in control mice on a number of behavioral responses at both ages and in all experimental paradigms, while perinatal exposure to BPA or MXC decreased or eliminated such sex differences. The present findings are evidence of long-term consequences of developmental exposure to BPA and MXC on neurobehavioral development and suggest a differential effect of low-dose exposure to these estrogenic chemicals in males and females.
Collapse
Affiliation(s)
- Laura Gioiosa
- Dipartimento di Biologia Evolutiva e Funzionale, Università di Parma, Viale Usberti 11A, 43100 Parma, Italy
| | | | | | | | | |
Collapse
|
215
|
Patisaul HB, Polston EK. Influence of endocrine active compounds on the developing rodent brain. ACTA ACUST UNITED AC 2007; 57:352-62. [PMID: 17822772 DOI: 10.1016/j.brainresrev.2007.06.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 06/11/2007] [Accepted: 06/12/2007] [Indexed: 11/25/2022]
Abstract
Changes in the volumes of sexually dimorphic brain nuclei are often used as a biomarker for developmental disruption by endocrine-active compounds (EACs). However, these gross, morphological analyses do not reliably predict disruption of cell phenotype or neuronal function. Therefore, an experimental approach that simultaneously assesses anatomical, physiological and behavioral endpoints is required when developing risk assessment models for EAC exposure. Using this more comprehensive approach we have demonstrated that the disruption of nuclear volume does not necessarily coincide with disruption of cellular phenotype or neuroendocrine function in two sexually dimorphic brain nuclei: the anteroventral periventricular nucleus of the hypothalamus (AVPV) and the sexually dimorphic nucleus of the preoptic area (SDN). These results demonstrate that nuclear volume is likely not an appropriate biomarker for EAC exposure. We further demonstrated that neonatal exposure to the EACs genistein (GEN) and Bisphenol-A (BPA) can affect sexually dimorphic brain morphology and neuronal phenotypes in adulthood with regional and cellular specificity suggesting that effects observed in one brain region may not be predictive of effects within neighboring regions. Finally, developmental EAC exposure has been shown to affect a variety of sexually dimorphic behaviors including reproductive behavior. These effects are likely to have a broad impact as maladaptive behavior could translate to decreased fitness of entire populations. Collectively, these findings emphasize the need to employ a comprehensive approach that addresses anatomical, functional and behavioral endpoints when evaluating the potential effects of EAC exposure.
Collapse
Affiliation(s)
- Heather B Patisaul
- Department of Zoology, North Carolina State University, 127 David Clark Labs, Raleigh, NC 27695, USA.
| | | |
Collapse
|
216
|
Ottem EN, Beck LA, Jordan CL, Breedlove SM. Androgen-dependent regulation of brain-derived neurotrophic factor and tyrosine kinase B in the sexually dimorphic spinal nucleus of the bulbocavernosus. Endocrinology 2007; 148:3655-65. [PMID: 17463054 DOI: 10.1210/en.2007-0308] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Castration of adult male rats causes the dendrites of androgen-sensitive motoneurons of the spinal nucleus of the bulbocavernosus (SNB) to retract. Brain-derived neurotrophic factor (BDNF), via activation of tyrosine receptor kinase B (trkB), has been implicated in mediating androgen effects on SNB dendrites. We used in situ hybridization to demonstrate that SNB motoneurons in gonadally intact adult male rats contain mRNA for both BDNF and trkB. Two weeks after gonadectomy, both transcripts were significantly decreased in SNB motoneurons but not in the non-androgen-responsive motoneurons of the adjacent retrodorsolateral nucleus (RDLN). In a second experiment, target perineal and foot muscles of SNB and RDLN motoneurons, respectively, were injected with the retrograde tracer Fluorogold, and then immunocytochemistry was performed to examine the distribution of BDNF and trkB proteins in SNB and RDLN motoneurons and their glutamatergic afferents. Confocal analysis revealed that gonadectomy induces a loss of BDNF protein in SNB dendrites but not in RDLN dendrites. Testosterone treatment of castrates prevented the loss of BDNF from SNB dendrites. Confocal analysis also revealed trkB protein in SNB and RDLN dendrites and in their glutamatergic afferents. Gonadectomy had no discernable effect on trkB protein in SNB or RDLN motoneurons or in their glutamatergic afferents. These results suggest that androgen maintains a BDNF-signaling pathway in SNB motoneurons that may underlie the maintenance of dendritic structure and synaptic signaling.
Collapse
Affiliation(s)
- Erich N Ottem
- Neuroscience Program, 108 Giltner Hall, Michigan State University, East Lansing, Michigan 48824, USA.
| | | | | | | |
Collapse
|
217
|
Abstract
The organization of the adult human medial preoptic nucleus was studied by using chemoarchitectonic markers for acetylcholinesterase, nonphosphorylated neurofilament protein (SMI-32), calbindin-D28k, neuropeptide Y (NPY), melanin-concentrating hormone (MCH), cocaine- and amphetamine-regulated transcript (CART), and 3-fucosyl-N-acetyl-lactosamine (CD15) to establish human homologs to the subnuclei making up MPO in the rat, where their connections and functional significance are better understood. The human MPO comprises three subnuclei, the medial MPO, the lateral MPO, and the dorsomedially positioned uncinate subnucleus. As in the rat, the human medial MPO is magnocellular and contains numerous NPY- and CART-immunoreactive fibers and terminals as well as calbindin-positive neurons. The human lateral MPO, like its homolog in the rat, distinctively features numerous MCH-positive fibers and terminals as well as SMI-32-immunoreactive fibers. The uncinate subnucleus is wedged between the lateral surface of the paraventricular nucleus and the medial MPO and is characterized by variable NPY- and CART-immunoreactive fibers and terminals, also seen in the rat central MPO, suggesting that the subnuclei are homologues. The intermediate nucleus was distinguished by CD15-positive neuronal staining, whereas the majority of its neurons also contained acetylcholinesterase. The human intermediate nucleus is positioned on the lateral surface of MPO and by its chemo- and cytoarchitecture constitutes a distinct nucleus of the preoptic area characterized by close structural association with the MPO complex. These findings demonstrate that the human MPO is organized similarly to the rat MPO, in chemo- and cytoarchitectonically distinct subnuclei, which implies differences in their functional specialization, as seen in the rat.
Collapse
Affiliation(s)
- Yuri Koutcherov
- The Prince of Wales Medical Research Institute, Randwick, New South Wales 2031, Australia
| | | | | |
Collapse
|
218
|
Purves-Tyson T, Arshi M, Handelsman DJ, Cheng Y, Keast JR. Androgen and estrogen receptor-mediated mechanisms of testosterone action in male rat pelvic autonomic ganglia. Neuroscience 2007; 148:92-104. [PMID: 17629410 PMCID: PMC2012365 DOI: 10.1016/j.neuroscience.2007.05.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Revised: 04/30/2007] [Accepted: 05/01/2007] [Indexed: 02/05/2023]
Abstract
Although male reproductive function is primarily androgen dependent, many studies suggest that estrogens have direct actions on the male reproductive organs. Pelvic autonomic neurons provide the motor control of the internal reproductive organs and the penis and various properties of these neurons are affected by endogenous androgens. However, the possible role of estrogens at this site has not been examined. Here we have investigated the significance of estrogens produced by aromatization of testosterone (T) in the physiological actions of androgens on adult male rat pelvic ganglion neurons. Reverse transcriptase polymerase chain reaction (RT-PCR) studies showed that aromatase and both estrogen receptors (ERalpha and ERbeta) are expressed in these ganglia. Western blotting also showed that aromatase is expressed in male pelvic ganglia. Using immunohistochemical visualization, ERalpha was predominantly expressed by nitric oxide synthase (NOS)-positive parasympathetic pelvic ganglion neurons. In vivo studies showed that the decrease in pelvic ganglion soma size caused by gonadectomy could be prevented by administration of T or dihydrotestosterone (DHT), but not 17beta-estradiol (E2), showing that this maintenance action of testosterone is mediated entirely by androgenic mechanisms. However, in vitro studies of cultured pelvic ganglion neurons revealed that T, DHT and E each stimulated the growth of longer and more complex neurites in both noradrenergic and cholinergic NOS-expressing neurons. The effects of T were attenuated by either androgen or estrogen receptor antagonists, or by inhibition of aromatase. Together these studies demonstrate that estrogens are likely to be synthesized in the male pelvic ganglia, produced from T by local aromatase. The effects of androgens on axonal growth are likely to be at least partly mediated by estrogenic mechanisms, which may be important for understanding disease-, aging- and injury-induced plasticity in this part of the nervous system.
Collapse
MESH Headings
- Androgen Antagonists/pharmacology
- Animals
- Aromatase/metabolism
- Cell Enlargement/drug effects
- Cells, Cultured
- Dihydrotestosterone/pharmacology
- Estrogen Antagonists/pharmacology
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor beta/metabolism
- Estrogens/biosynthesis
- Ganglia, Autonomic/drug effects
- Ganglia, Autonomic/metabolism
- Ganglia, Parasympathetic/drug effects
- Ganglia, Parasympathetic/metabolism
- Genitalia, Male/innervation
- Genitalia, Male/physiology
- Hypogastric Plexus/drug effects
- Hypogastric Plexus/metabolism
- Male
- Nitrergic Neurons/drug effects
- Nitrergic Neurons/metabolism
- Nitric Oxide Synthase/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Androgen/drug effects
- Receptors, Androgen/metabolism
- Receptors, Estrogen/drug effects
- Receptors, Estrogen/metabolism
- Testosterone/metabolism
Collapse
Affiliation(s)
- T.D. Purves-Tyson
- Pain Management Research Institute, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
- Prince of Wales Medical Research Institute and University of New South Wales, Barker Street, Randwick, NSW 2031 Australia
| | - M.S. Arshi
- Pain Management Research Institute, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | | | - Y. Cheng
- Pain Management Research Institute, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - J. R. Keast
- Pain Management Research Institute, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| |
Collapse
|
219
|
Olesen KM, Auger CJ, Auger AP. Regulation of progestin receptor expression in the developing rat brain by a dopamine d1 receptor antagonist. J Neuroendocrinol 2007; 19:481-8. [PMID: 17561880 DOI: 10.1111/j.1365-2826.2007.01554.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Steroid receptors within the developing brain influence a variety of cellular processes that endure into adulthood, altering both behaviour and physiology. Therefore, it is important to understand how steroid receptor expression is regulated during early brain development. Most studies indicate that oestradiol, by acting upon oestrogen receptors, increases the expression of progestin receptors in the developing brain. We have recently observed an additional mechanism by which dopamine can increase the expression of progestin receptors in developing female rat brain. That is, we found that a dopamine D1 receptor agonist can further increase progestin receptor expression by activating oestrogen receptors in a ligand-independent manner within restricted areas of female brain; however, it is unclear whether dopamine D1 receptors are involved in the normally occurring expression of progestin receptors in developing male and female brain. To investigate this, we examined whether a dopamine D1 receptor antagonist can disrupt the normal developmental expression of progestin receptors in both male and female rat brain. We report that treatment with a dopamine D1 receptor antagonist reduces progestin receptor expression within some, but not all, regions of the developing rat brain in a sex-specific manner. Some of the current findings also suggest that dopamine might be acting to prevent sex differences in progestin receptor expression in some areas while contributing to a sex difference in other areas.
Collapse
Affiliation(s)
- K M Olesen
- Department of Psychology, University of Wisconsin, Madison, WI 53706, USA
| | | | | |
Collapse
|
220
|
Honda S, Toda K, Tozuka Y, Yasuzawa S, Iwabuchi K, Tomooka Y. Migration and differentiation of neural cell lines transplanted into mouse brains. Neurosci Res 2007; 59:124-35. [PMID: 17651850 DOI: 10.1016/j.neures.2007.06.1467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 05/25/2007] [Accepted: 06/11/2007] [Indexed: 11/29/2022]
Abstract
In the past few years, the plasticity of the regional specification of the CNS has been widely debated on the results from in utero transplantation. Two different results are reported with this transplantation method. One is that the distribution of transplanted cells is dependent on the donor origin, and the other is that the distribution is independent on the donor cell origin. The present study attempted to examine closely the plasticity of the regional specification by in utero transplantation method with clonal neural cell lines, 2Y-3t and 2Y-5o2b. These lines were established from a cerebellum of an adult p53-deficient mouse. Our results showed that transplanted cells migrated into various regions of the CNS and supported the independent distribution. Moreover, different distribution patterns of transplanted cells were observed between host sexes. Labeled cells were localized around the ventricle of neonatal host brains, where they were undifferentiated. In 2-3 weeks after birth, labeled cells were found in the brain parenchyma and some of them took neuronal morphology. In the rostral migratory stream (RMS), cells with unipolar or bipolar morphology were still undifferentiated. In other regions, labeled cells were often associated with blood vessels; the soma were on the surface of vessels, extending processes or neurites into surrounding brain parenchyma. Time-lapse imaging demonstrated that they were migrating with blood vessels.
Collapse
Affiliation(s)
- Shinya Honda
- Department of Biological Science and Technology, and Tissue Engineering Research Center, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | |
Collapse
|
221
|
Holmes MM, Rosen GJ, Jordan CL, de Vries GJ, Goldman BD, Forger NG. Social control of brain morphology in a eusocial mammal. Proc Natl Acad Sci U S A 2007; 104:10548-52. [PMID: 17556547 PMCID: PMC1965550 DOI: 10.1073/pnas.0610344104] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Indexed: 11/18/2022] Open
Abstract
Social status impacts reproductive behavior in diverse vertebrate species, but little is known about how it affects brain morphology. We explore this in the naked mole-rat, a species with the most rigidly organized reproductive hierarchy among mammals. Naked mole-rats live in large, subterranean colonies where breeding is restricted to a single female and small number of males. All other members of the colony, known as subordinates, are reproductively suppressed. Subordinates can become breeders if removed from the colony and placed with an opposite sex partner, but in nature most individuals never attain reproductive status. We examined the brains of breeding and subordinate naked mole-rats of both sexes, including several regions linked to reproduction and shown to be sexually dimorphic in other mammals. Stereological analyses revealed that neural morphology depends on status, such that breeders, regardless of sex, had more cells than subordinates in the ventromedial nucleus of the hypothalamus and a larger volume of the bed nucleus of the stria terminalis, paraventricular nucleus, and medial amygdala. Several other brain regions examined were unaffected. Surprisingly, males and females did not differ on any measure. These findings provide evidence that a change in social status triggers considerable neural remodeling and indicate that status, rather than sex, has a predominant role in determining neural structure in this remarkably social mammal.
Collapse
Affiliation(s)
- Melissa M Holmes
- Center for Neuroendocrine Studies and Department of Psychology, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | | | | | | | |
Collapse
|
222
|
Tang YP, Peabody C, Tomaszycki ML, Wade J. Sexually dimorphic SCAMP1 expression in the forebrain motor pathway for song production of juvenile zebra finches. Dev Neurobiol 2007; 67:474-82. [PMID: 17443802 PMCID: PMC2878128 DOI: 10.1002/dneu.20354] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mechanisms regulating sexual differentiation of the zebra finch song system are not well understood. The present study was designed to more fully characterize secretory carrier membrane protein 1 (SCAMP1), which was identified in a cDNA microarray screen as showing increased expression in the forebrains of developing male compared with female zebra finches. We completed the sequence of the open reading frame and used in situ hybridization to compare mRNA in song control regions of juvenile (25-day-old) individuals. Expression was significantly greater in the HVC (used as a proper name) and robust nucleus of the arcopallium (RA) in males than in females. Immunohistochemistry revealed that SCAMP1 protein is also expressed in these two brain regions, and qualitatively appears greater in males. Western analysis confirmed that the protein is increased in the telencephalon of males when compared with females at 25 days of age. These results are consistent with the idea that SCAMP1 is involved in masculinization of these brain areas, perhaps facilitating the survival of cells within them.
Collapse
Affiliation(s)
- Yu Ping Tang
- Department of Psychology, Michigan State University, East Lansing, Michigan 48824-1101
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101
| | - Camilla Peabody
- Department of Psychology, Michigan State University, East Lansing, Michigan 48824-1101
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101
| | - Michelle L. Tomaszycki
- Department of Psychology, Michigan State University, East Lansing, Michigan 48824-1101
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101
| | - Juli Wade
- Department of Psychology, Michigan State University, East Lansing, Michigan 48824-1101
- Department of Zoology, Michigan State University, East Lansing, Michigan 48824-1101
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824-1101
- Correspondence to: J. Wade ()
| |
Collapse
|
223
|
Milner TA, Hernandez FJ, Herrick SP, Pierce JP, Iadecola C, Drake CT. Cellular and subcellular localization of androgen receptor immunoreactivity relative to C1 adrenergic neurons in the rostral ventrolateral medulla of male and female rats. Synapse 2007; 61:268-78. [PMID: 17318878 DOI: 10.1002/syn.20370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In male and female rats, high androgen levels can increase blood pressure. The C1 area of the rostral ventrolateral medulla (RVLM), which is crucial for blood pressure regulation, contains estrogen receptors (ERs) in pre- and postsynaptic neuronal compartments and is modulated by estrogens (Wang et al. [2006] Brain Res 1094:163-178). In this study, the cellular and subcellular localization of androgen receptors (ARs) in the C1 area was examined in sections from male, proestrus (high estrogen) and diestrus (low estrogen) female rat brains that were immunocytochemically labeled for AR and tyrosine hydroxylase (TH). By light and electron microscopy, AR-labeled nuclei were scattered among TH-labeled somata in the RVLM; significantly more AR-labeled nuclei were seen males compared to females. Electron microscopy revealed that extranuclear AR-immunoreactivity (ir) was in similar profile types in male and female rats. AR-ir was almost exclusively in myelinated and unmyelinated axons and in glia. Rarely, AR-ir was in axon terminals that contacted TH-containing dendrites. AR-labeled axon terminals had large diameters and contained numerous dense-core vesicles, resembling peptide-containing hypothalamic or solitary tract inputs. No nuclear or extranuclear AR-ir was found in TH-labeled perikarya and dendrites although a few non-TH- labeled dendrites contained AR-ir. Qualitatively, more axonal profiles appeared to be present in males compared to females. These studies suggest that, unlike ERs, ARs in male and female rats are almost exclusively positioned on afferents and glia, suggesting that androgens modulate RVLM C1 neurons, and thus blood pressure, through presynaptic and glial signaling.
Collapse
Affiliation(s)
- Teresa A Milner
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | | | | | | | | | |
Collapse
|
224
|
Cooke BM, Stokas MR, Woolley CS. Morphological sex differences and laterality in the prepubertal medial amygdala. J Comp Neurol 2007; 501:904-15. [PMID: 17311322 DOI: 10.1002/cne.21281] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The medial amygdala (MeA) is crucial in the expression of sex-specific social behaviors. In adult rats the regional volume of the MeA posterodorsal subnucleus (MeApd) is approximately 50% larger in males than in females. The MeApd is also sexually dimorphic in prepubertal rats. We have recently shown that the left MeApd is significantly larger in prepubertal males than females. In contrast with volumetric sex differences elsewhere in the brain, however, we found no sex difference in the number of left MeApd neurons. In the present study we investigated the cellular bases of the sex difference in MeApd regional volume by quantifying the volume occupied by dendrites, axons, synapses, or glia, and by measuring MeApd dendritic morphology in 26-29-day-old male and female rats. We find that the volume occupied by dendritic shafts and glia completely accounts for the sex difference in left MeApd regional volume. Dendritic length measurements in the left hemisphere confirm that males have greater overall dendritic length, which is due to greater branching rather than to longer dendrite segments. In the right hemisphere the pattern of sex differences was different: Males have more MeApd neurons than females, whereas the dendritic morphology of individual neurons is not sexually dimorphic. These results highlight the importance of evaluating laterality in the MeA and suggest that the left and right MeA could play different roles in neuroendocrine regulation and sexually dimorphic social behaviors.
Collapse
Affiliation(s)
- Bradley M Cooke
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208, USA.
| | | | | |
Collapse
|
225
|
Perrot-Sinal TS, Sinal CJ, Reader JC, Speert DB, McCarthy MM. Sex differences in the chloride cotransporters, NKCC1 and KCC2, in the developing hypothalamus. J Neuroendocrinol 2007; 19:302-8. [PMID: 17355320 DOI: 10.1111/j.1365-2826.2007.01530.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In immature neurones, high basal [Cl(-)](i) results in membrane depolarisation following GABA(A) receptor activation, which is critical for various developmental processes including steroid-mediated sexual differentiation of the hypothalamus. Previously, we demonstrated that oestradiol enhances GABA-mediated Ca(2+) influx in neonate hypothalamus and that Ca(2+) induced activation of the transcription factor, cyclicAMP response element binding protein (CREB), was higher in male (high oestradiol) relative to female neonate hypothalamus. Based on these results, we hypothesised that expression of developmentally regulated chloride cotransporters may be sexually dimorphic. Here, we investigate the expression of the chloride cotransporters, NKCC1 (Na-K-2Cl(-)) and KCC2 (K-Cl(-)) in neonate mediobasal hypothalamus of male and female rats. The NKCC1 transporter moves Cl(-) into cells and helps maintain depolarising GABA action while the KCC2 transporter has the opposite effect by moving Cl(-) out of cells. NKCC1 mRNA levels were higher in males than females on the day of birth (postnatal day 0; PND 0) and total NKCC1 protein levels were significantly higher in males than females on embryonic day (ED) 20 and PND0. Levels of activated phosphorylated NKCC1 (pNKCC1) were not sexually dimorphic. Females were treated with a masculinising dose of oestradiol benzoate (EB; 100 microg; EB-females) on PND0. Total NKCC1 protein levels in tissue processed on PND1 and PND2 were similar in EB-females and oil-treated PND0 males and females. However, pNKCC1 protein levels measured on PND2 (but not PND1) were significantly higher in EB-treated females relative to oil-treated males and females. By contrast, KCC2 mRNA levels were significantly lower in males relative to females on PND0. KCC2 protein was not detectable on ED20 or PND0 but was significantly lower in males relative to females on PND5. These results suggest a complex relationship between KCC2 and NKCC1 mRNA and protein in developing brain that is not easily linked to regulation by oestradiol.
Collapse
Affiliation(s)
- T S Perrot-Sinal
- Department of Psychology and Neuroscience Institute, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | | | |
Collapse
|
226
|
Gaumond I, Spooner MF, Marchand S. Sex differences in opioid-mediated pain inhibitory mechanisms during the interphase in the formalin test. Neuroscience 2007; 146:366-74. [PMID: 17306464 DOI: 10.1016/j.neuroscience.2007.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 12/16/2006] [Accepted: 01/04/2007] [Indexed: 10/23/2022]
Abstract
Many chronic pain conditions are more prevalent in women than men and both fundamental and clinical research supports the implication of endogenous pain inhibitory mechanisms. The goal of this study was to verify if sex differences on endogenous pain inhibitory mechanisms during the formalin test are opioidergic and modulated by sex hormones. Formalin tests were performed with naloxone hydrochloride, a non-selective opioid antagonist in intact and gonadectomized Sprague-Dawley rats of both sexes. Considering the sexual dimorphisms we found, where naloxone preferentially blocked the interphase in female rats, injections of all the possible combinations of mu- (naltrexone hydrochloride), delta- (naltrindole hydrochloride) and kappa-selective antagonists (norbinaltorphimine dihydrochloride) were given to evaluate the contribution of these opioid-receptor subtypes to the inhibitory mechanism during the interphase in intact females. Finally, the systemic administration of naloxone methiodide and intrathecal administration of naloxone hydrochloride in intact females allowed us to verify if the action of endogenous opioids that are liberated during the interphase takes place at the periphery or spinally, respectively. The results show that the interphase was almost completely inhibited by naloxone in females while it produced only a slight blockade in males. These results permitted us to conclude that opioids play a major role in the pain inhibitory mechanism of the interphase in females while a non-opioid mechanism seems to be responsible for this inhibitory pathway in males. Using gonadectomized animals of both sexes, we demonstrated the modulation of the opioidergic system of the interphase by sex hormones. The administration of different combinations of selective antagonists for mu-, kappa- and delta-opioid receptors in intact females permitted us to conclude that only the combination of kappa- and delta-selective antagonists significantly blocked the interphase. The same result was obtained with the combination of the three antagonists, confirming the results with systemic naloxone hydrochloride. Finally, intrathecal administration permitted us to support that the action of naloxone is primarily at the spinal level, even if a supraspinal action cannot be ruled out. These results are of particular interest in showing sexual dimorphisms in endogenous pain modulation mechanisms during the interphase of the formalin test. A clearer understanding of the difference between male and female endogenous pain inhibitory pathways should lead to a better understanding of the role of endogenous pain modulation deficits in certain chronic pain conditions.
Collapse
Affiliation(s)
- I Gaumond
- Département des Sciences de la Santé, Université du Québec en Abitibi-Témiscamingue, 445 Boulevard de l'Université, Rouyn-Noranda, Québec, Canada J9X 5E4
| | | | | |
Collapse
|
227
|
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; 148:1774-83. [PMID: 17204549 DOI: 10.1210/en.2006-1540] [Citation(s) in RCA: 366] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Kiss1 gene codes for kisspeptins, which have been implicated in the neuroendocrine regulation of reproduction. In the brain, Kiss1 mRNA-expressing neurons are located in the arcuate (ARC) and anteroventral periventricular (AVPV) nuclei. Kiss1 neurons in the AVPV appear to play a role in generating the preovulatory GnRH/LH surge, which occurs only in females and is organized perinatally by gonadal steroids. Because Kiss1 is involved in the sexually dimorphic GnRH/LH surge, we hypothesized that Kiss1 expression is sexually differentiated, with females having more Kiss1 neurons than either males or neonatally androgenized females. To test this, male and female rats were neonatally treated with androgen or vehicle; then, as adults, they were left intact or gonadectomized and implanted with capsules containing sex steroids or nothing. Kiss1 mRNA levels in the AVPV and ARC were determined by in situ hybridization. Normal females expressed significantly more Kiss1 mRNA in the AVPV than normal males, even under identical adult hormonal conditions. This Kiss1 sex difference was organized perinatally, as demonstrated by the observation that neonatally androgenized females displayed a male-like pattern of adulthood Kiss1 expression in the AVPV. In contrast, there was neither a sex difference nor an influence of neonatal treatment on Kiss1 expression in the ARC. Using double-labeling techniques, we determined that the sexually differentiated Kiss1 neurons in the AVPV are distinct from the sexually differentiated population of tyrosine hydroxylase (dopaminergic) neurons in this region. Our findings suggest that sex differences in kisspeptin signaling from the AVPV subserve the cellular mechanisms controlling the sexually differentiated GnRH/LH surge.
Collapse
Affiliation(s)
- Alexander S Kauffman
- Department of Physiology and Biophysics, University of Washington, Box 357290, Seattle, Washington 98195-7290, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Spring S, Lerch JP, Henkelman RM. Sexual dimorphism revealed in the structure of the mouse brain using three-dimensional magnetic resonance imaging. Neuroimage 2007; 35:1424-33. [PMID: 17408971 DOI: 10.1016/j.neuroimage.2007.02.023] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Revised: 02/12/2007] [Accepted: 02/16/2007] [Indexed: 11/15/2022] Open
Abstract
A large variety of sexual dimorphisms have been described in the brains of many vertebrate species, including humans. Naturally occurring sexual dimorphism has been implicated in the risk, progression and recovery from numerous neurological disorders, including head injury, multiple sclerosis and stroke. Genetically altered mice are a key tool in the study of structure-function relationships in the mammalian central nervous system and serve as models for human neuropsychiatric and neurological disorders. However, there are a limited number of quantitative three-dimensional analyses of the adult mouse brain structures. In order to address limitations in our knowledge of anatomical differences, a comprehensive study was undertaken using full 3D magnetic resonance imaging (MRI) to examine sexual dimorphisms in the C57BL/6J whole mouse brain. An expected difference in overall brain size between the sexes was found, where male brains were 2.5% larger in volume than female brains. Beyond the overall brain size differences in the sexes, the following significantly different regions were found: males were larger in the thalamus, primary motor cortex and posterior hippocampus, while females were larger in posterior hypothalamic area, entorhinal cortex and anterior hippocampus. Using high-definition 3D MRI on a normal inbred mouse strain, we have mapped in detail many sex-associated statistically significant differences in brain structures.
Collapse
Affiliation(s)
- Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8.
| | | | | |
Collapse
|
229
|
Viglietti-Panzica C, Mura E, Panzica G. Effects of early embryonic exposure to genistein on male copulatory behavior and vasotocin system of Japanese quail. Horm Behav 2007; 51:355-63. [PMID: 17274996 DOI: 10.1016/j.yhbeh.2006.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 11/14/2006] [Accepted: 12/04/2006] [Indexed: 11/19/2022]
Abstract
Genistein is a phytoestrogen, particularly abundant in soybeans that can bind estrogen receptors and sex hormone binding proteins, exerting both estrogenic and antiestrogenic activity. In this study we used the Japanese quail embryo as a test end-point to investigate the effects of early embryonic exposure to genistein on male copulatory behavior and on vasotocin parvocellular system. Both differentiate by the organizational effects of estradiol during development and may therefore represent an optimal model to study the effects of xenoestrogens. We injected two doses of genistein (100 and 1000 microg) into the yolk of 3-day-old Japanese quail eggs. Other eggs were treated with either 25 microg of estradiol benzoate or sesame oil as positive and negative controls. At the age of 6 weeks, behavioral tests revealed a significant decrease of all aspects of copulatory behavior (in comparison to the control group) in estradiol-treated birds. In contrast, genistein-treated animals demonstrated various degrees of decrease in the mean frequencies of some aspects of the sexual behavior. The computerized analysis of vasotocin innervation in medial preoptic, stria terminalis and lateral septum nuclei revealed a statistically significant decreased immunoreactivity in treated animals compared to control ones. These results demonstrate that genistein, similarly to estradiol, has an organizational effect on quail parvocellular vasotocin system and on copulatory behavior. In conclusion, present results confirm, in this avian model, that embryonic exposure to phytoestrogens may have life-long effects on sexual differentiation of brain structures and behaviors.
Collapse
Affiliation(s)
- Carla Viglietti-Panzica
- Laboratory of Neuroendocrinology, Neuroscience Institute of Torino, University of Torino, Torino, Italy
| | | | | |
Collapse
|
230
|
Kallivretaki E, Eggen RI, Neuhauss SC, Kah O, Segner H. The zebrafish, brain-specific, aromatasecyp19a2 is neither expressed nor distributed in a sexually dimorphic manner during sexual differentiation. Dev Dyn 2007; 236:3155-66. [DOI: 10.1002/dvdy.21344] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
231
|
Neal JK, Wade J. Courtship and copulation in the adult male green anole: effects of season, hormone and female contact on reproductive behavior and morphology. Behav Brain Res 2006; 177:177-85. [PMID: 17174414 PMCID: PMC2892282 DOI: 10.1016/j.bbr.2006.11.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 11/20/2006] [Accepted: 11/21/2006] [Indexed: 12/26/2022]
Abstract
Interactions among reproductive season, testosterone (T) and female presence were investigated on the structure and function of forebrain and neuromuscular systems controlling courtship and copulation in the green anole lizard. Under breeding (BS) or non-breeding (NBS) environmental conditions, male green anoles were implanted with either T or blank capsules and exposed to one of three female stimulus conditions: physical, visual or no female contact. T and at least visual exposure to females increased courtship displays (extension of a throat fan, or dewlap), and these effects were greater during the BS than NBS. T also facilitated copulation, and did so to a greater extent in the BS. The hormone increased soma size in the preoptic area (POA) and amygdala (AMY), and in the AMY the effects were greater in the BS than NBS. Cross-sectional areas of copulatory organs and associated muscle fibers were enhanced by T, and more so in the BS than NBS. However, no effects on morphology of dewlap motoneurons or muscles or copulatory motoneurons were detected. Thus, (1) changes in behavior and neural and/or muscular morphology are not always parallel and (2) differences in responsiveness to T exist across seasons and among tissues.
Collapse
Affiliation(s)
- Jennifer K Neal
- Michigan State University, Neuroscience Program, East Lansing, MI 48824-1101, United States.
| | | |
Collapse
|
232
|
Abstract
Starting from fetal life, estrogens are crucial in determining central gender dimorphism, and an estrogen-induced synaptic plasticity is well evident during puberty and seasonal changes as well as during the ovarian cycle. Estrogens act on the central nervous system (CNS) both through genomic mechanisms, modulating synthesis, release and metabolism of neurotransmitters, neuropeptides and neurosteroids, and through non-genomic mechanisms, influencing electrical excitability, synaptic function and morphological features. Therefore, estrogen's neuroactive effects are multifaceted and encompass a system that ranges from the chemical to the biochemical to the genomic mechanisms, protecting against a wide range of neurotoxic insults. Clinical evidences show that, during the climacteric period, estrogen withdrawal in the limbic system gives rise to modifications in mood, behaviour and cognition and that estrogen administration is able to improve mood and cognitive efficiency in post-menopause. Many biological mechanisms support the hypothesis that estrogens might protect against Alzheimer's disease (AD) by influencing neurotransmission, increasing cerebral blood flow, modulating growth proteins associated with axonal elongation and blunting the neurotoxic effects of beta-amyloid. On the contrary, clinical studies of estrogen replacement therapy (ERT) and cognitive function have reported controversial results, indicating a lack of efficacy of estrogens on cognition in post-menopausal women aged >or=65 years. These findings suggest the presence of a critical period for HRT-related neuroprotection and underlie the potential importance of early initiation of therapy for cognitive benefit. In this review, we shall first describe the multiple effects of steroids in the nervous system, which may be significant in the ageing process. A critical update of HRT use in women and a discussion of possible prospectives for steroid use are subsequently proposed.
Collapse
Affiliation(s)
- Andrea Riccardo Genazzani
- Department of Reproductive Medicine and Child Development, Division of Obstetrics and Gynecology, University of Pisa, Pisa, Italy.
| | | | | | | |
Collapse
|
233
|
Schneider JE. Metabolic and hormonal control of the desire for food and sex: implications for obesity and eating disorders. Horm Behav 2006; 50:562-71. [PMID: 16875692 DOI: 10.1016/j.yhbeh.2006.06.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 06/26/2006] [Accepted: 06/26/2006] [Indexed: 01/08/2023]
Abstract
During evolution, the ability to overeat and store the extra energy as glycogen and lipids in specialized tissues must have conferred a reproductive advantage by releasing animals from the need to eat constantly, enabling them to engage in behaviors that improved reproductive success. Mechanisms that inhibited ingestive behavior might have been most adaptive when they caused individuals to stop foraging, hoarding and eating in order to find and court potential mates. Conversely, the ability to abstain from reproductive activities to engage in foraging and eating was probably critical for individual survival during severe energetic challenges because reproductive processes are energetically costly and can be delayed until the energetic conditions improve. The mechanisms that control ingestive behavior most likely evolved under conditions in which both food and mates were available, and thus, our understanding might be limited by our narrow focus on food intake in animals isolated from potential mates, and reproductive behaviors in the absence of food. Our understanding of obesity and eating disorders will be enriched by the study of the choice between ingestive and reproductive behaviors and by a renewed attention to "reproductive" hormones such as gonadal steroids and hypothalamic releasing hormones. Furthermore, leptin and reproductive hormones have both organizational and activational effects on the energy balancing system including those mechanisms that control appetite, body fat content and body fat distribution. Understanding these organizational and activational effects on body fat distribution might lead to a better understanding of sex differences in the propensity to develop obesity, type II diabetes and eating disorders.
Collapse
Affiliation(s)
- Jill E Schneider
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, USA.
| |
Collapse
|
234
|
Patisaul HB, Fortino AE, Polston EK. Differential disruption of nuclear volume and neuronal phenotype in the preoptic area by neonatal exposure to genistein and bisphenol-A. Neurotoxicology 2006; 28:1-12. [PMID: 17109964 DOI: 10.1016/j.neuro.2006.10.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 09/06/2006] [Accepted: 10/04/2006] [Indexed: 11/24/2022]
Abstract
Changes in the volumes of sexually dimorphic brain nuclei are often used as a biomarker for developmental disruption by endocrine-active compounds (EACs). However, these gross, morphological analyses do not reliably predict disruption of cell phenotype or neuronal function. In the present experiments, we used a more comprehensive approach to assess whether postnatal exposure to the EACs genistein (GEN) or bisphenol-A (BIS) affected the development of two sexually dimorphic brain regions in male rats: the anteroventral periventricular nucleus of the hypothalamus (AVPV) and the sexually dimorphic nucleus of the preoptic area (SDN). In addition to nuclear volumes, we also measured the number of immunopositive calbindin neurons in the SDN and the activational patterns of gonadotropin-releasing hormone (GnRH) neurons, a neuronal population that is functionally linked to the AVPV. In rats, exposure of the neonatal male brain to endogenous estrogen, aromatized from testicular testosterone, is essential for the proper sexual differentiation of these endpoints. Thus, we hypothesized that exposure to BIS and GEN during this critical period could disrupt brain sexual differentiation. Animals were given four subcutaneous injections of sesame oil (control), 250 microg GEN, or 250 microg BIS at 12 h intervals over postnatal days (PND) 1 and 2, gonadectomized on PND 85, and treated sequentially with estrogen and progesterone to stimulate Fos expression in GnRH neurons, a marker for their activation. A cohort of age-matched ovariectomized (OVX) females that were given the same hormone treatment in adulthood served as a positive control group. SDN volume was unchanged by treatment, but the number of calbindin neurons in the SDN was significantly increased by both BIS and GEN. GEN, but not BIS, demasculinized male AVPV volume, but patterns of GnRH neuronal activation were not affected by either compound. These results suggest that acute exposure to EACs during a critical developmental period can independently alter nuclear volumes of sexually dimorphic nuclei and their phenotypic profiles in a region specific manner.
Collapse
Affiliation(s)
- Heather B Patisaul
- CIIT Centers for Health Research, 6 Davis Drive, RTP, NC 27709, United States.
| | | | | |
Collapse
|
235
|
Giorgi FS, Velíšková J, Chudomel O, Kyrozis A, Moshé SL. The role of substantia nigra pars reticulata in modulating clonic seizures is determined by testosterone levels during the immediate postnatal period. Neurobiol Dis 2006; 25:73-9. [PMID: 17011203 PMCID: PMC1661598 DOI: 10.1016/j.nbd.2006.08.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 08/03/2006] [Accepted: 08/20/2006] [Indexed: 11/27/2022] Open
Abstract
GABAergic activation of substantia nigra pars reticulata (SNR) at postnatal day (PN) 15 has sex-specific features on seizure control in vivo and electrophysiological responses in vitro. In males, the GABA(A)-receptor agonist muscimol has proconvulsant effects and induces depolarizing responses. In females, muscimol has no effect on seizures and evokes hyperpolarizing responses. We determined the time period during which sex hormones must be present to produce the sex-specific muscimol effects on seizures and their influence on SNR GABA(A) receptor-mediated postsynaptic currents. Exposure to testosterone or its metabolites (estrogen or dihydrotestosterone) during PN0-2 in females or males castrated at PN0 was sufficient to produce proconvulsant muscimol effects but did not affect the in vitro GABA responses, which remained hyperpolarizing. The data suggest that the PN0-2 period is critical for the development of the seizure-controlling SNR system; the hormonal effect on seizure control is independent from their effect on GABA conductance.
Collapse
Affiliation(s)
- Filippo S. Giorgi
- The Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
- Department of Neurosciences, Section of Neurology, University of Pisa, Pisa, Italy
| | - Jana Velíšková
- The Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
- the Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
| | - Ondřej Chudomel
- The Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
| | - Andreas Kyrozis
- The Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
- Department of Neurology, University of Athens, Greece
| | - Solomon L. Moshé
- The Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
- the Dominick P. Purpura Department of Neuroscience, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
- Department of Pediatrics ,Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY, USA
| |
Collapse
|
236
|
Tang YP, Wade J. Sexually dimorphic expression of the genes encoding ribosomal proteins L17 and L37 in the song control nuclei of juvenile zebra finches. Brain Res 2006; 1126:102-8. [PMID: 16938280 PMCID: PMC2878125 DOI: 10.1016/j.brainres.2006.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 07/31/2006] [Accepted: 08/03/2006] [Indexed: 11/25/2022]
Abstract
Studies evaluating the role of steroid hormones in sexual differentiation of the zebra finch song system have produced complicated and at times paradoxical results, and indicate that additional factors may be critical. Therefore, in a previous study we initiated a screen for differential gene expression in the telencephalon of developing male and female zebra finches. The use of cDNA microarrays and real-time quantitative PCR revealed increased expression of the genes encoding ribosomal proteins L17 and L37 (RPL17 and RPL37) in the male forebrain as a whole. Preliminary in situ hybridization data then indicated enhanced expression of both these genes in song control regions. Two experiments in the present study quantified the mRNA expression. The first utilized 25-day-old male and female zebra finches. The second compared a separate set of juveniles to adults of both sexes to both re-confirm enhanced expression in juvenile males and to determine whether it is limited to developing animals. In Experiment 1, males exhibited increased expression of both RPL17 and RPL37 compared to females in Area X, the robust nucleus of the arcopallium (RA), and the ventral ventricular zone (VVZ), which may provide neurons to Area X. Experiment 2 replicated the sexually dimorphic expression of these genes at post-hatching day 25, and documented that the sex differences are eliminated or greatly reduced in adults. The results are consistent with the idea that these ribosomal proteins may influence sexual differentiation of Area X and RA, potentially regulating the genesis and/or survival of neurons.
Collapse
Affiliation(s)
| | - Juli Wade
- Corresponding author. Fax: +1 517 432 2744. (J. Wade)
| |
Collapse
|
237
|
Lee G, Bahn JH, Park JH. Sex- and clock-controlled expression of the neuropeptide F gene in Drosophila. Proc Natl Acad Sci U S A 2006; 103:12580-5. [PMID: 16894172 PMCID: PMC1567921 DOI: 10.1073/pnas.0601171103] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Indexed: 11/18/2022] Open
Abstract
Drosophila neuropeptide F (NPF), a homolog of vertebrate neuropeptide Y, functions in feeding and coordination of behavioral changes in larvae and in modulation of alcohol sensitivity in adults, suggesting diverse roles for this peptide. To gain more insight into adult-specific NPF neuronal functions, we studied how npf expression is regulated in the adult brain. Here, we report that npf expression is regulated in both sex-nonspecific and male-specific manners. Our data show that male-specific npf (ms-npf) expression is controlled by the transformer (tra)-dependent sex-determination pathway. Furthermore, fruitless, one of the major genes functioning downstream of tra, is apparently an upstream regulator of ms-npf transcription. Males lacking ms-npf expression (through tra(F)-mediated feminization) or npf-ablated male flies display significantly reduced male courtship activity, suggesting that one function of ms-npf neurons is to modulate fruitless-regulated sexual behavior. Interestingly, one of the ms-npf neuronal groups belongs to the previously defined clock-controlling dorsolateral neurons. Such ms-npf expression in the dorsolateral neurons is absent in arrhythmic Clock(Jrk) and cycle(02) mutants, suggesting that npf is under dual regulation by circadian and sex-determining factors. Based on these data, we propose that NPF also plays a role in clock-controlled sexual dimorphism in adult Drosophila.
Collapse
Affiliation(s)
- Gyunghee Lee
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996
| | - Jae Hoon Bahn
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996
| | - Jae H. Park
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996
| |
Collapse
|
238
|
Trotter A, Ebsen M, Kiossis E, Meggle S, Kueppers E, Beyer C, Pohlandt F, Maier L, Thome UH. Prenatal estrogen and progesterone deprivation impairs alveolar formation and fluid clearance in newborn piglets. Pediatr Res 2006; 60:60-4. [PMID: 16690946 DOI: 10.1203/01.pdr.0000220360.77567.d8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Exposure to high levels of estradiol (E2) and progesterone (P) derived from the fetoplacentomaternal unit during the last trimester of pregnancy may play a crucial role in prenatal lung development and immediate postnatal alveolar fluid clearance (AFC). To measure prenatal alveolar formation and postnatal amiloride-sensitive AFC after pharmacological deprivation of E2 and P in utero, fetuses from five sows received an intramuscular depot injection of the E2 receptor blocker ICI 182.780 (ICI) and the P receptor blocker RTI 3021-022 (RTI) and fetuses of five other sows received a placebo injection (control group) during a laparotomy at 90 d of gestation (term gestation, 115 d). Piglets were delivered by cesarean section on d 114 of gestation. Of 95 live-born piglets, 35 were mechanically ventilated. The airways of the right lower lobe were isolated by a balloon catheter wedged in the bronchus and 5% albumin in 0.9% NaCl with or without 1 mmol/L amiloride was instilled. Amiloride-sensitive AFC was calculated from the protein concentration changes in fluid recovered after 120 min as the percentage of absorbed fluid. Lungs were removed under standardized conditions to perform alveolar counts. Prenatal treatment with ICI and RTI resulted in a significantly lower amiloride-sensitive AFC (median, 31%; min-max, -4-58) than placebo (74%, 18-231). Median alveolar counts per visual field were significantly lower in piglets that were exposed to ICI and RTI (38, 21-78) compared with placebo (56, 32-113). We conclude that prenatal E2 and P deprivation significantly impaired alveolar formation and amiloride-sensitive AFC.
Collapse
Affiliation(s)
- Andreas Trotter
- Division of Neonatology and Pediatric Critical Care Medicine, University of Ulm, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Miller JE, Levine RB. Steroid hormone activation of wandering in the isolated nervous system of Manduca sexta. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2006; 192:1049-62. [PMID: 16788816 DOI: 10.1007/s00359-006-0143-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Revised: 05/17/2006] [Accepted: 05/21/2006] [Indexed: 02/02/2023]
Abstract
Steroid hormones modulate motor circuits in both vertebrates and invertebrates. The insect Manduca sexta, with its well-characterized developmental and endocrinological history, is a useful model system in which to study these effects. Wandering is a stage-specific locomotor behavior triggered by the steroid hormone 20-hydroxyecdysone (20E), consisting of crawling and burrowing movements as the animal searches for a pupation site. This study was undertaken to determine whether the wandering motor pattern is activated by direct action of 20E on the CNS. 20E acts on the isolated larval nervous system to induce a fictive motor pattern showing features of crawling and burrowing. The latency of the response to 20E is long, suggestive of a genomic mechanism of action. The abdominal motoneurons or segmental pattern generating circuits are unlikely to be the primary targets of 20E action in inducing fictive wandering. Exposure of the segmental ganglia alone to hormone did not evoke fictive wandering. Therefore, as suggested by an earlier study, the likely site of 20E action is within the brain.
Collapse
Affiliation(s)
- Julie E Miller
- Division of Neurobiology, University of Arizona, Tucson, AZ 85721, USA.
| | | |
Collapse
|
240
|
Bolzán AG, Guimarey LM. Efecto del retardo de crecimiento intrauterino sobre el dimorfismo sexual en recien nacidos de termino: una adaptación prenatal en perspectiva evolutiva. REVISTA BRASILEIRA DE SAÚDE MATERNO INFANTIL 2006. [DOI: 10.1590/s1519-38292006000200006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJETIVOS: identificar si se modifica el dismorfismo sexual del crecimiento frente a retardo de crecimiento intrauterino. MÉTODOS: se compararon 4931 recién nacidos con (n=497) y sin (n=4434) retardo de crecimiento intrauterino. Todos fueron de término (para Argentina >37 semanas). La población se dividió en dos grupos de acuerdo a la condición de crecimiento fetal y el sexo. Los indicadores antropométricos del crecimiento prenatal incluyeron el peso corporal, la longitud y el perímetro cefálico. RESULTADOS: no hubo diferencias estadísticas significativas entre sexos en el peso al nacer y la longitud corporal en neonatos con retardo de crecimiento intrauterino. De forma contraria, los neonatos con crecimiento fetal normal evidenciaron diferencias significativas en ambos indicadores entre sexos siendo más grandes los varones. CONCLUSIONES: la inhibición del dismorfismo sexual está ya presente en etapas prenatales cuando ocurre una situación que afecte el crecimiento como es el caso del retardo de crecimiento intrauterino. Se considera la perspectiva evolutiva de dicho fenómeno inhibitorio.
Collapse
|
241
|
Bisenius ES, Veeramachaneni DNR, Sammonds GE, Tobet S. Sex differences and the development of the rabbit brain: effects of vinclozolin. Biol Reprod 2006; 75:469-76. [PMID: 16738224 DOI: 10.1095/biolreprod.106.052795] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The preoptic/anterior hypothalamic area (POA/AH) is one of the most sexually dimorphic areas of the vertebrate brain and plays a pivotal role in regulating male sexual behavior. Vinclozolin is a fungicide thought to be an environmental antiandrogen, which disrupts masculine sexual behavior when administered to rabbits during development. In this study, we examined several characteristics of the rabbit POA/AH for sexual dimorphism and endocrine disruption by vinclozolin. Pregnant rabbits were dosed orally with vinclozolin (10 mg/kg body weight) or carrot paste vehicle once daily for 6 wk beginning at midgestation and continuing through nursing until Postpartum Week 4. At 6 wk, offspring were perfused with 4% paraformaldehyde and brains processed for immunocytochemical localization of tyrosine hydroxylase, calbindin, gonadotropin-releasing hormone (GnRH), or Nissl stain. There were significant sex differences in the distribution of calbindin in the POA/AH and the size of cells in the dorsal POA/AH (values greater in females than in males), but not in the number or distribution of tyrosine hydroxylase or GnRH neurons. In both sexes, exposure to vinclozolin significantly increased calbindin expression in the ventral POA/AH and significantly decreased number of GnRH neurons selectively in the region of the organum vasculosum of the lamina terminalis (OVLT) but not more caudally in the POA/AH. This is the first documentation of a sexually dimorphic region in the rabbit brain, and further supports the use of this species as a model for studying the influence of vinclozolin on reproductive development with potential application to human systems.
Collapse
Affiliation(s)
- Erin S Bisenius
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | |
Collapse
|
242
|
Wade J. Relationships among hormones, brain and behavior: Exceptions in search of a rule? Horm Behav 2006; 49:577-9. [PMID: 16504185 DOI: 10.1016/j.yhbeh.2006.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Revised: 01/19/2006] [Accepted: 01/19/2006] [Indexed: 11/26/2022]
Affiliation(s)
- Juli Wade
- Michigan State University, Department of Psychology, 108 Giltner Hall, East Lansing, MI 48824-1101, USA.
| |
Collapse
|
243
|
Pinaud R, Fortes AF, Lovell P, Mello CV. Calbindin-positive neurons reveal a sexual dimorphism within the songbird analogue of the mammalian auditory cortex. ACTA ACUST UNITED AC 2006; 66:182-95. [PMID: 16288476 DOI: 10.1002/neu.20211] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The oscine song system, a set of interconnected brain nuclei involved in song production and learning, is one of the first and clearest examples of brain sexual dimorphism in a vertebrate, being typically well-developed in males, but not females. Here we present evidence for a sexual dimorphism in the caudomedial nidopallidum (NCM), an auditory area outside of the song system. NCM is thought to correspond to a portion of the auditory cortex of mammals and is involved in the perceptual processing of birdsong. We show that cells immunolabeled for the calcium-binding protein calbindin are primarily localized to caudal NCM and are almost twice as numerous in males as in females. We demonstrate that calbindin-positive cells constitute a subset of GABAergic cells in NCM, and show that the sex dimorphism in this cell population does not result from local gender differences in the overall density of neuronal or GABAergic cells. In addition, we demonstrate that calbindin-positive cells lack song-induced expression of the activity-dependent gene ZENK, and that song stimulation does not change the density or distribution of these cells in NCM. Finally, we show that the distribution of calbindin-positive cells in NCM is strikingly similar to the mRNA expression for the estrogen-generating enzyme aromatase. Together these results suggest that NCM is likely composed of neurochemically-distinct domains and presents a marked sex dimorphism in a specific subset of GABAergic neurons, which may confer sex-specific sensory processing capabilities to this auditory area. Our results also suggest that local sex steroid hormones may play a local role in auditory processing in the songbird telencephalon.
Collapse
Affiliation(s)
- Raphael Pinaud
- Laboratory of Auditory and Vocal Learning, Neurological Sciences Institute, Oregon Health & Science University, Portland, Oregon 97006, USA
| | | | | | | |
Collapse
|
244
|
Suzuki S, Brown CM, Wise PM. Mechanisms of neuroprotection by estrogen. Endocrine 2006; 29:209-15. [PMID: 16785597 DOI: 10.1385/endo:29:2:209] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 10/25/2005] [Accepted: 10/25/2005] [Indexed: 01/26/2023]
Abstract
Over the past decade our recognition that estrogens function as important neurotrophic and neuroprotective factors has grown rapidly. Accumulating evidence from basic science studies demonstrates that estrogens exert profound protective actions against various forms of neurodegenerative diseases and injury. Although a thorough understanding of the mechanisms underlying the protective effect of estrogens is far from complete, significant progress has been achieved through the use of in vivo as well as in vitro models. Here we review the results from our laboratory demonstrating that low physiological levels of estradiol therapy exert powerful protection against ischemic stroke-like injury. Using an animal model of cerebrovascular stroke and in vitro explant cultures, we have begun to decipher under what circumstances 17beta-estradiol protects against neuronal death and to uncover its mechanisms of action. In addition, we will review recent work demonstrating that estradiol may additionally enhance the ability of the adult brain to undergo repair by influencing the production of new neurons under neuropathological conditions, as well as by promoting an anti-inflammatory response. As we uncover the important protective roles of ovarian steroid hormones in brain disease and injury, we increasingly appreciate that the mechanisms by which estrogens achieve these effects are diverse and complex.
Collapse
Affiliation(s)
- Shotaro Suzuki
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | | | | |
Collapse
|
245
|
Schaevitz LR, Berger-Sweeney J. Neurogenesis of the cholinergic medial septum in female and male C57BL/6J mice. ACTA ACUST UNITED AC 2006; 65:294-303. [PMID: 16187329 DOI: 10.1002/neu.20188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Sex differences exist in the structure and function of the cholinergic septo-hippocampal system throughout the lifespan of mammals. How and when these sex differences originate is unclear. Because estrogen modulates sexual differentiation of several brain regions during development and influences neurogenesis in adult mammals, we hypothesized that sexual dimorphism of the cholinergic septo-hippocampal system would extend to its neurogenesis. A birthdating agent 5'-bromo-2'-deoxyuridine (BrdU) was injected into pregnant dams on one of eight gestational days, ranging from embryonic day (E)10 to E17. The offspring were euthanized at 2 months of age, and brains were processed for BrdU and choline acetyltransferase (ChAT) immunoreactivity to label cholinergic neurons that became postmitotic on a given embryonic day and survived to adulthood. Unbiased stereology was used to compare the number of double-labeled neurons in the medial septum (MS) of female and male offspring. Cholinergic neurons in the MS were generated primarily between E11 and E14, similar to other published reports. We found sex differences in the pattern of peak neurogenesis but not in the length of neurogenesis, or in total number of neurons generated in the MS. Additionally, in adult female and male mice, we estimated the total number of cholinergic neurons using unbiased stereology and found no sex differences in the number of cholinergic neurons or in the volume of the MS in adulthood. These results suggest that sex differences noted in the function of the postnatal cholinergic septo-hippocampal system may originate from its neurogenesis.
Collapse
Affiliation(s)
- Laura R Schaevitz
- Department of Biological Sciences, Wellesley College, Massachusetts 02481, USA
| | | |
Collapse
|
246
|
Abstract
The medial amygdala is important in social behaviors, many of which differ between males and females. The posterodorsal subnucleus of the medial amygdala (MeApd) is particularly sensitive to gonadal steroid hormones and is a likely site for gonadal hormone regulation of sexually dimorphic social behavior. Here we show that the synaptic organization of the MeApd in the rat is sexually dimorphic and lateralized before puberty. With the use of whole-cell voltage-clamp recording and quantitative electron microscopy, we found that, specifically in the left hemisphere, prepubertal males have approximately 80% more excitatory synapses per MeApd neuron than females. In the left but not the right MeApd, miniature EPSC (mEPSC) frequency was significantly greater in males than in females; mEPSC amplitude was not sexually dimorphic. Paired-pulse facilitation of EPSCs, an index of release probability, also was not sexually dimorphic, suggesting that greater mEPSC frequency is caused by a difference in excitatory synapse number. Electron microscopy confirmed that the asymmetric synapse-to-neuron ratio and the total asymmetric synapse number were significantly greater in the left MeApd of males than of females. In contrast to results for excitatory synapses, we found no evidence of sexual dimorphism or laterality in inhibitory synapses. Neither the frequency nor the amplitude of mIPSCs was sexually dimorphic or lateralized. Likewise, the number of symmetric synapses measured with electron microscopy was not sexually dimorphic. These findings show that the excitatory synaptic organization of the left MeApd is sexually differentiated before puberty, which could provide a sexually dimorphic neural substrate for the effects of hormones on adult social behavior.
Collapse
Affiliation(s)
- Bradley M Cooke
- Department of Neurobiology and Physiology and Institute for Neuroscience, Northwestern University, Evanston, Illinois 60208, USA.
| | | |
Collapse
|
247
|
Chung WCJ, Pak TR, Weiser MJ, Hinds LR, Andersen ME, Handa RJ. Progestin receptor expression in the developing rat brain depends upon activation of estrogen receptor alpha and not estrogen receptor beta. Brain Res 2006; 1082:50-60. [PMID: 16513095 DOI: 10.1016/j.brainres.2006.01.109] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 01/25/2006] [Accepted: 01/27/2006] [Indexed: 01/12/2023]
Abstract
Perinatal 17beta-estradiol (E2) rapidly and markedly affects the morphological and neurochemical organization of the vertebrate brain. For instance, the sex difference in perinatal progestin receptor (PR) immunoreactivity in the medial preoptic nucleus (MPN) of the rat brain is due to the intracellular conversion of testosterone into E2 in males. Neonatal alpha-fetoprotein prevents circulating estrogens from accessing the brain, therefore, to overcome alpha-fetoprotein sequestration of E2, estrogen replacement studies during development have used natural and synthetic estrogen dosages in the milligram to microgram range. These levels could be considered as supraphysiological. Moreover, it is not clear through which ER subtype E2 acts to induce PR expression in the neonatal rat MPN because E2 binds similarly to estrogen receptor (ER)alpha and ERbeta. Consequently, we investigated whether nanogram levels of E2 affected PR protein and mRNA levels in the neonatal MPN. Furthermore, propylpyrazole-triol (PPT), a highly selective agonist for ERalpha, and diarylpropionitrile (DPN), a highly selective agonist for ERbeta, were used to determine if E2-dependent PR expression in the neonatal rat is mediated through ERalpha and/or ERbeta. Immunocytochemistry and quantitative real-time RT-PCR determined that as little as 100 ng E2 significantly induced PR protein and mRNA in the female and neonatally castrated male MPN on PN 4, indicating that the neonatal rat brain is highly sensitive to circulating estrogens. PPT, but not DPN, induced PR expression in the neonatal MPN and arcuate nucleus (Arc), demonstrating that PR expression in the neonatal rat brain depends solely on E2 activated ERalpha. In the lateral bed nucleus of the stria terminalis (BSTL), neither PPT nor DPN affected PR expression, suggesting the presence of a gonadal hormone-independent PR regulatory mechanism.
Collapse
Affiliation(s)
- Wilson C J Chung
- Department of Biomedical Sciences, Colorado State University, Fort Collins, 80523, USA.
| | | | | | | | | | | |
Collapse
|
248
|
Lorenz B, Garcia-Segura LM, DonCarlos LL. Cellular phenotype of androgen receptor-immunoreactive nuclei in the developing and adult rat brain. J Comp Neurol 2006; 492:456-68. [PMID: 16228996 DOI: 10.1002/cne.20763] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Androgen exposure during development and adulthood promotes cell-to-cell communication, modulates the size of specific brain nuclei, and influences hormone-dependent behavioral and neuroendocrine functions. Androgen action involves the activation of androgen receptors (AR). To elucidate the mechanisms involved in AR-mediated effects on forebrain development, double-label fluorescent immunohistochemistry and confocal microscopy were employed to identify the cellular phenotype of AR-immunoreactive (AR(+)) cells in the developing (embryonic day 20, postnatal days 0, 4, 10) and adult male rat forebrain. Sections were doubly labeled with antibodies directed against AR and one of the following: neurons (immature, nestin; mature, NeuN) or astrocytes [immature, vimentin; mature, glial fibrillary acidic protein (GFAP)] or mature oligodendrocytes (mGalC). In all brain regions examined, by far the majority of AR(+) cells were neurons. In addition, small subsets of AR(+) cells were identified as mature astrocytes (GFAP(+)) but only in specific brain regions at specific ages. AR(+)/GFAP(+) cells were observed in the cerebral cortex but only in postnatal day 10 rats and in the arcuate nucleus of the hypothalamus but only in adult rats. Immature neurons, immature astrocytes, and oligodendrocytes were not AR(+) at any age, in any region. Thus, both neurons and astrocytes in the male rat forebrain contain ARs, suggesting that androgens, via ARs, may exert effects on both cell types in an age- and region-dependent manner.
Collapse
Affiliation(s)
- Betty Lorenz
- Neuroscience Program, Loyola University Chicago, Maywood, IL 60153, USA
| | | | | |
Collapse
|
249
|
Keast JR. Plasticity of pelvic autonomic ganglia and urogenital innervation. INTERNATIONAL REVIEW OF CYTOLOGY 2006; 248:141-208. [PMID: 16487791 DOI: 10.1016/s0074-7696(06)48003-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pelvic ganglia contain a mixture of sympathetic and parasympathetic neurons and provide most of the motor innervation of the urogenital organs. They show a remarkable sensitivity to androgens and estrogens, which impacts on their development into sexually dimorphic structures and provide an array of mechanisms by which plasticity of these neurons can occur during puberty and adulthood. The structure of pelvic ganglia varies widely among species, ranging from rodents, which have a pair of large ganglia, to humans, in whom pelvic ganglion neurons are distributed in a large, complex plexus. This plexus is frequently injured during pelvic surgical procedures, yet strategies for its repair have yet to be developed. Advances in this area will come from a better understanding of the effects of injury on the cellular signaling process in pelvic neurons and also the role of neurotrophic factors during development, maintenance, and repair of these axons.
Collapse
Affiliation(s)
- Janet R Keast
- Pain Management Research Institute, University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
250
|
Lippa RA. Finger lengths, 2D:4D ratios, and their relation to gender-related personality traits and the Big Five. Biol Psychol 2006; 71:116-21. [PMID: 16360883 DOI: 10.1016/j.biopsycho.2005.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Accepted: 02/19/2005] [Indexed: 10/25/2022]
Abstract
Finger lengths and the ratio of index finger to ring finger length (2D:4D) may be markers of gonadal hormone exposure. The current study investigated possible associations between absolute finger lengths, 2D:4D ratios, and gender-related personality traits in over 2000 participants. Regression analyses showed no associations between men's 2D:4D ratios and gender-related personality traits and weak associations between women's 2D:4D ratios and gender-related occupational preferences. Men's absolute finger lengths were weakly associated with self-ascribed masculinity, and women's absolute finger lengths were weakly associated with masculine occupational preferences. Big Five personality traits were assessed in a subsample of over 1000 participants. Analyses showed a weak positive association between 2D:4D and extraversion and a weak negative association between 2D:4D and openness to experience. Absolute finger lengths showed a weak negative association with agreeableness and a tendency to be associated with women's but not men's openness. Overall, associations between finger-length measures and personality were weak and inconsistent.
Collapse
Affiliation(s)
- Richard A Lippa
- Psychology Department, California State University, Fullerton, CA 92834, USA.
| |
Collapse
|