1
|
McCarthy MM. Neural Control of Sexually Dimorphic Social Behavior: Connecting Development to Adulthood. Annu Rev Neurosci 2023; 46:321-339. [PMID: 37001242 DOI: 10.1146/annurev-neuro-121522-110856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Rapid advances in the neural control of social behavior highlight the role of interconnected nodes engaged in differential information processing to generate behavior. Many innate social behaviors are essential to reproductive fitness and therefore fundamentally different in males and females. Programming these differences occurs early in development in mammals, following gonadal differentiation and copious androgen production by the fetal testis during a critical period. Early-life programming of social behavior and its adult manifestation are separate but yoked processes, yet how they are linked is unknown. This review seeks to highlight that gap by identifying four core mechanisms (epigenetics, cell death, circuit formation, and adult hormonal modulation) that could connect developmental changes to the adult behaviors of mating and aggression. We further propose that a unique social behavior, adolescent play, bridges the preweaning to the postpubertal brain by engaging the same neural networks underpinning adult reproductive and aggressive behaviors.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
2
|
Chiang VSC, DeRosa H, Park JH, Hunter RG. The Role of Transposable Elements in Sexual Development. Front Behav Neurosci 2022; 16:923732. [PMID: 35874645 PMCID: PMC9301316 DOI: 10.3389/fnbeh.2022.923732] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022] Open
Abstract
Up to 50% of most mammalian genomes are made up of transposable elements (TEs) that have the potential to mobilize around the genome. Despite this prevalence, research on TEs is only beginning to gain traction within the field of neuroscience. While TEs have long been regarded as "junk" or parasitic DNA, it has become evident that they are adaptive DNA and RNA regulatory elements. In addition to their vital role in normal development, TEs can also interact with steroid receptors, which are key elements to sexual development. In this review, we provide an overview of the involvement of TEs in processes related to sexual development- from TE activity in the germline to TE accumulation in sex chromosomes. Moreover, we highlight sex differences in TE activity and their regulation of genes related to sexual development. Finally, we speculate on the epigenetic mechanisms that may govern TEs' role in sexual development. In this context, we emphasize the need to further the understanding of sexual development through the lens of TEs including in a variety of organs at different developmental stages, their molecular networks, and evolution.
Collapse
Affiliation(s)
| | | | | | - Richard G. Hunter
- College of Liberal Arts, Department of Psychology, Developmental and Brain Sciences Program, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
3
|
Tsukahara S, Morishita M. Sexually Dimorphic Formation of the Preoptic Area and the Bed Nucleus of the Stria Terminalis by Neuroestrogens. Front Neurosci 2020; 14:797. [PMID: 32848568 PMCID: PMC7403479 DOI: 10.3389/fnins.2020.00797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/07/2020] [Indexed: 01/08/2023] Open
Abstract
Testicular androgens during the perinatal period play an important role in the sexual differentiation of the brain of rodents. Testicular androgens transported into the brain act via androgen receptors or are the substrate of aromatase, which synthesizes neuroestrogens that act via estrogen receptors. The latter that occurs in the perinatal period significantly contributes to the sexual differentiation of the brain. The preoptic area (POA) and the bed nucleus of the stria terminalis (BNST) are sexually dimorphic brain regions that are involved in the regulation of sex-specific social behaviors and the reproductive neuroendocrine system. Here, we discuss how neuroestrogens of testicular origin act in the perinatal period to organize the sexually dimorphic structures of the POA and BNST. Accumulating data from rodent studies suggest that neuroestrogens induce the sex differences in glial and immune cells, which play an important role in the sexually dimorphic formation of the dendritic synapse patterning in the POA, and induce the sex differences in the cell number of specific neuronal cell groups in the POA and BNST, which may be established by controlling the number of cells dying by apoptosis or the phenotypic organization of living cells. Testicular androgens in the peripubertal period also contribute to the sexual differentiation of the POA and BNST, and thus their aromatization to estrogens may be unnecessary. Additionally, we discuss the notion that testicular androgens that do not aromatize to estrogens can also induce significant effects on the sexually dimorphic formation of the POA and BNST.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| |
Collapse
|
4
|
Kammel LG, Correa SM. Selective sexual differentiation of neurone populations may contribute to sex-specific outputs of the ventromedial nucleus of the hypothalamus. J Neuroendocrinol 2020; 32:e12801. [PMID: 31605642 PMCID: PMC6982598 DOI: 10.1111/jne.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/26/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
Sex differences among neurones in the ventrolateral region of the ventromedial hypothalamic nucleus (VMHvl) allow for the display of a diversity of sex-typical behaviours and physiological responses, ranging from mating behaviour to metabolism. Here, we review recent studies that interrogate the relationship between sex-typical responses and changes in cellular phenotypes. We discuss technologies that increase the resolution of molecular profiling or targeting of cell populations, including single-cell transcriptional profiling and conditional viral genetic approaches to manipulate neurone survival or activity. Overall, emerging studies indicate that sex-typical functions of the VMH may be mediated by phenotypically distinct and sexually differentiated neurone populations within the VMHvl. Future studies in this and other brain regions could exploit cell-type-specific tools to reveal the cell populations and molecular mediators that modulate sex-typical responses. Furthermore, cell-type-specific analyses of the effects of sexually differentiating factors, including sex hormones, can test the hypothesis that distinct cell types within a single brain region vary with respect to sexual differentiation.
Collapse
Affiliation(s)
- Laura G Kammel
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
- Molecular, Cellular, Integrative Physiology Graduate Program, University of California, Los Angeles, CA, USA
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
5
|
Gegenhuber B, Tollkuhn J. Signatures of sex: Sex differences in gene expression in the vertebrate brain. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e348. [PMID: 31106965 PMCID: PMC6864223 DOI: 10.1002/wdev.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | |
Collapse
|
6
|
John N, Rehman H, Razak S, David M, Ullah W, Afsar T, Almajwal A, Alam I, Jahan S. Comparative study of environmental pollutants bisphenol A and bisphenol S on sexual differentiation of anteroventral periventricular nucleus and spermatogenesis. Reprod Biol Endocrinol 2019; 17:53. [PMID: 31292004 PMCID: PMC6621953 DOI: 10.1186/s12958-019-0491-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bisphenol A is well known endocrine-disrupting chemical while Bisphenol S was considered a safe alternative. The present study aims to examine the comparative effects of xenobiotic bisphenol-A (BPA) and its substitute bisphenol-S (BPS) on spermatogenesis and development of sexually dimorphic nucleus population of dopaminergic neurons in the anteroventral periventricular nucleus (AVPV) of the hypothalamus in male pups. METHODS Sprague Dawley rat's pups were administered subcutaneously at the neonatal stage from postnatal day PND1 to PND 27. Thirty animals were divided into six experimental groups (6 animals/group). The first group served as control and was provided with normal olive oil. The four groups were treated with 2 μg/kg and 200 μg/kg of BPA and BPS, respectively. The sixth group was given with 50 μg/kg of estradiol dissolved in olive oil as a standard to find the development of dopaminergic tyrosine hydroxylase neurons in AVPV regions. Histological analysis for testicular tissues and immunohistochemistry for brain tissues was performed. RESULTS The results revealed adverse histopathological changes in testis after administration of different doses of BPA and BPS. These degenerative changes were marked by highly significant (p < 0.001) decrease in tubular and luminal diameters of seminiferous tubule and epithelial height among bisphenols treated groups as compared to control. Furthermore, significantly increased (p < 0.001) TH-ir cell bodies in the AVPV region of the brain with 200 μg/kg dose of BPA and BPS was evident. CONCLUSION It is concluded that exposure of BPA and BPS during a critical developmental period can structural impairments in testes and affects sexual differentiation of a dimorphic dopaminergic population of AVPV region of hypothalamus in the male brain.
Collapse
Affiliation(s)
- Naham John
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Humaira Rehman
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Suhail Razak
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mehwish David
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Waheed Ullah
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Tayyaba Afsar
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ali Almajwal
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Iftikhar Alam
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sarwat Jahan
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| |
Collapse
|
7
|
Forger NG, Ruszkowski E, Jacobs A, Wallen K. Effects of sex and prenatal androgen manipulations on Onuf's nucleus of rhesus macaques. Horm Behav 2018; 100:39-46. [PMID: 29510099 PMCID: PMC6084473 DOI: 10.1016/j.yhbeh.2018.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/15/2018] [Accepted: 03/02/2018] [Indexed: 10/17/2022]
Abstract
The role of gonadal steroids in sexual differentiation of the central nervous system (CNS) is well established in rodents, but no study to date has manipulated androgens prenatally and examined their effects on any CNS structure in a primate. Onuf's nucleus is a column of motoneurons in the sacral spinal cord that innervates the striated perineal muscles. This cell group is larger in males than in females of many species, due to androgens acting during a sensitive perinatal period. Here, we examined Onuf's nucleus in 21 adult rhesus monkeys, including control males and females, as well as males whose mothers had been treated with an anti-androgen or testosterone during gestation. We found a robust sex difference, with more motoneurons in control males than in females. The soma size of Onuf's nucleus motoneurons was also marginally larger in males. Treatment with the anti-androgen flutamide for 35-40 days during early gestation partially blocked masculinization of Onuf's nucleus: motoneuron number in flutamide-treated males was decreased relative to control and testosterone-treated males, but remained greater than in females, with no effect on cell size. A control motor nucleus that innervates foot muscles (Pes9) showed no difference in motoneuron number or size between control males and females. Prenatal testosterone treatment of males did not alter Onuf's nucleus motoneuron number, but did increase the size of both Onuf's and Pes9 motoneurons. Thus, prenatal androgen manipulations cause cellular-level changes in the primate CNS, which may underlie previously observed effects of these manipulations on behavior.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - Elara Ruszkowski
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Andrew Jacobs
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Kim Wallen
- Department of Psychology, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
8
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
9
|
Mohr MA, Garcia FL, DonCarlos LL, Sisk CL. Neurons and Glial Cells Are Added to the Female Rat Anteroventral Periventricular Nucleus During Puberty. Endocrinology 2016; 157:2393-402. [PMID: 27145006 PMCID: PMC4891785 DOI: 10.1210/en.2015-2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Francisca L Garcia
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Lydia L DonCarlos
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Cheryl L Sisk
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| |
Collapse
|
10
|
Forger NG, Strahan JA, Castillo-Ruiz A. Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 2016; 40:67-86. [PMID: 26790970 PMCID: PMC4897775 DOI: 10.1016/j.yfrne.2016.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/31/2015] [Accepted: 01/09/2016] [Indexed: 01/16/2023]
Abstract
Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | - J Alex Strahan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | | |
Collapse
|
11
|
McCarthy MM, Pickett LA, VanRyzin JW, Kight KE. Surprising origins of sex differences in the brain. Horm Behav 2015; 76:3-10. [PMID: 25917865 PMCID: PMC4620061 DOI: 10.1016/j.yhbeh.2015.04.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 11/22/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Discerning the biologic origins of neuroanatomical sex differences has been of interest since they were first reported in the late 60's and early 70's. The centrality of gonadal hormone exposure during a developmental critical window cannot be denied but hormones are indirect agents of change, acting to induce gene transcription or modulate membrane bound signaling cascades. Sex differences in the brain include regional volume differences due to differential cell death, neuronal and glial genesis, dendritic branching and synaptic patterning. Early emphasis on mechanism therefore focused on neurotransmitters and neural growth factors, but by and large these endpoints failed to explain the origins of neural sex differences. More recently evidence has accumulated in favor of inflammatory mediators and immune cells as principle regulators of brain sexual differentiation and reveal that the establishment of dimorphic circuits is not cell autonomous but instead requires extensive cell-to-cell communication including cells of non-neuronal origin. Despite the multiplicity of cells involved the nature of the sex differences in the neuroanatomical endpoints suggests canalization, a process that explains the robustness of individuals in the face of intrinsic and extrinsic variability. We propose that some neuroanatomical endpoints are canalized to enhance sex differences in the brain by reducing variability within one sex while also preventing the sexes from diverging too greatly. We further propose mechanisms by which such canalization could occur and discuss what relevance this may have to sex differences in behavior.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Lindsay A Pickett
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jonathan W VanRyzin
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine E Kight
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Reddy RC, Scheldrup M, Meaker M, Stormshak F, Estill CT, Roselli CE. Cell death in the central division of the medial preoptic nucleus of male and female lamb fetuses. Brain Res 2014; 1554:21-8. [PMID: 24491631 DOI: 10.1016/j.brainres.2014.01.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/22/2014] [Accepted: 01/25/2014] [Indexed: 10/25/2022]
Abstract
The medial preoptic area of the adult sheep contains an ovine sexually dimorphic nucleus (oSDN) that is larger and has more neurons in males than in females. In the lamb fetus, the nascent oSDN occupies the central division of the medial preoptic nucleus (MPNc) and consists of a cluster of cells that is organized by the action of testosterone during gestational days 60-90 of a 147 day term pregnancy. The current study sought to determine whether programmed cell death contributes to the emergence of the oSDN. Male and female lamb fetuses were euthanized at different ages spanning the period during which the oSDN is organized. The expression of the pro- and anti-apoptotic genes bcl-2 and bax, respectively, was measured by quantitative RT-PCR to assess possible sex differences in neuron vulnerability to programmed cell death. The appearance of DNA-fragmentation was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and used to estimate the occurrence of apoptotic cell death. We found that bcl-2 and bax mRNA expression in the medial preoptic area of the developing lamb fetus decreased during the last half of the 147-day gestation. The ratio of bcl-2/bax gene expression was highest at gestational day 85 but was equivalent between males and females. TUNEL staining in the MPNc was very low and although it decreased significantly with age, it was not significantly different between sexes. These results using two different methods to assess cell death indicate that a sex difference in the incidence of cell death is not a primary mechanism leading to sexual differentiation of the oSDN.
Collapse
Affiliation(s)
- Radhika C Reddy
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Melissa Scheldrup
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Mary Meaker
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Fred Stormshak
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Charles T Estill
- College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Charles E Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| |
Collapse
|
13
|
de Vries GJ, Fields CT, Peters NV, Whylings J, Paul MJ. Sensitive periods for hormonal programming of the brain. Curr Top Behav Neurosci 2014; 16:79-108. [PMID: 24549723 DOI: 10.1007/7854_2014_286] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During sensitive periods, information from the external and internal environment that occurs during particular phases of development is relayed to the brain to program neural development. Hormones play a central role in this process. In this review, we first discuss sexual differentiation of the brain as an example of hormonal programming. Using sexual differentiation, we define sensitive periods, review cellular and molecular processes that can explain their restricted temporal window, and discuss challenges in determining the precise timing of the temporal window. We then briefly review programming effects of other hormonal systems and discuss how programming of these systems interact with sexual differentiation.
Collapse
Affiliation(s)
- Geert J de Vries
- Neuroscience Institute, Georgia State University, PO Box 5030, Atlanta, GA, 30302-5030, USA,
| | | | | | | | | |
Collapse
|
14
|
Ahern TH, Krug S, Carr AV, Murray EK, Fitzpatrick E, Bengston L, McCutcheon J, De Vries GJ, Forger NG. Cell death atlas of the postnatal mouse ventral forebrain and hypothalamus: effects of age and sex. J Comp Neurol 2013; 521:2551-69. [PMID: 23296992 PMCID: PMC4968939 DOI: 10.1002/cne.23298] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/28/2012] [Accepted: 12/26/2012] [Indexed: 01/21/2023]
Abstract
Naturally occurring cell death is essential to the development of the mammalian nervous system. Although the importance of developmental cell death has been appreciated for decades, there is no comprehensive account of cell death across brain areas in the mouse. Moreover, several regional sex differences in cell death have been described for the ventral forebrain and hypothalamus, but it is not known how widespread the phenomenon is. We used immunohistochemical detection of activated caspase-3 to identify dying cells in the brains of male and female mice from postnatal day (P) 1 to P11. Cell death density, total number of dying cells, and regional volume were determined in 16 regions of the hypothalamus and ventral forebrain (the anterior hypothalamus, arcuate nucleus, anteroventral periventricular nucleus, medial preoptic nucleus, paraventricular nucleus, suprachiasmatic nucleus, and ventromedial nucleus of the hypothalamus; the basolateral, central, and medial amygdala; the lateral and principal nuclei of the bed nuclei of the stria terminalis; the caudate-putamen; the globus pallidus; the lateral septum; and the islands of Calleja). All regions showed a significant effect of age on cell death. The timing of peak cell death varied between P1 to P7, and the average rate of cell death varied tenfold among regions. Several significant sex differences in cell death and/or regional volume were detected. These data address large gaps in the developmental literature and suggest interesting region-specific differences in the prevalence and timing of cell death in the hypothalamus and ventral forebrain.
Collapse
Affiliation(s)
- Todd H. Ahern
- Center for Behavioral Neuroscience, Department of Psychology, Quinnipiac University, Hamden, Connecticut 06518
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Stefanie Krug
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Audrey V. Carr
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Elaine K. Murray
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Emmett Fitzpatrick
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lynn Bengston
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Jill McCutcheon
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Geert J. De Vries
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Nancy G. Forger
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| |
Collapse
|
15
|
Iwakura T, Sakoh M, Tsutiya A, Yamashita N, Ohtani A, Tsuda MC, Ogawa S, Tsukahara S, Nishihara M, Shiga T, Goshima Y, Kato T, Ohtani-Kaneko R. Collapsin response mediator protein 4 affects the number of tyrosine hydroxylase-immunoreactive neurons in the sexually dimorphic nucleus in female mice. Dev Neurobiol 2013; 73:502-17. [PMID: 23420586 DOI: 10.1002/dneu.22076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/25/2012] [Accepted: 02/12/2013] [Indexed: 01/04/2023]
Abstract
In the sexually dimorphic anteroventral periventricular nucleus (AVPV) of the hypothalamus, females have a greater number of tyrosine hydroxylase-immunoreactive (TH-ir) and kisspeptin-immunoreactive (kisspeptin-ir) neurons than males. In this study, we used proteomics analysis and gene-deficient mice to identify proteins that regulate the number of TH-ir and kisspeptin-ir neurons in the AVPV. Analysis of protein expressions in the rat AVPV on postnatal day 1 (PD1; the early phase of sex differentiation) using two-dimensional fluorescence difference gel electrophoresis followed by MALDI-TOF-MS identified collapsin response mediator protein 4 (CRMP4) as a protein exhibiting sexually dimorphic expression. Interestingly, this sexually differential expressions of CRMP4 protein and mRNA in the AVPV was not detected on PD6. Prenatal testosterone exposure canceled the sexual difference in the expression of Crmp4 mRNA in the rat AVPV. Next, we used CRMP4-knockout (CRMP4-KO) mice to determine the in vivo function of CRMP4 in the AVPV. Crmp4 knockout did not change the number of kisspeptin-ir neurons in the adult AVPV in either sex. However, the number of TH-ir neurons was increased in the AVPV of adult female CRMP4-KO mice as compared with the adult female wild-type mice. During development, no significant difference in the number of TH-ir neurons was detected between sexes or genotypes on embryonic day 15, but a female-specific increase in TH-ir neurons was observed in CRMP4-KO mice on PD1, when the sex difference was not yet apparent in wild-type mice. These results indicate that CRMP4 regulates the number of TH-ir cell number in the female AVPV.
Collapse
Affiliation(s)
- Takashi Iwakura
- Doctoral Program in Kansei, Behavioral and Brain Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang Y, Xu L, Pan Y, Wang Z, Zhang Z. Species differences in the immunoreactive expression of oxytocin, vasopressin, tyrosine hydroxylase and estrogen receptor alpha in the brain of Mongolian gerbils (Meriones unguiculatus) and Chinese striped hamsters (Cricetulus barabensis). PLoS One 2013; 8:e65807. [PMID: 23762431 PMCID: PMC3676338 DOI: 10.1371/journal.pone.0065807] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/28/2013] [Indexed: 01/21/2023] Open
Abstract
Species differences in neurochemical expression and activity in the brain may play an important role in species-specific patterns of social behavior. In the present study, we used immunoreactive (ir) labeling to compare the regional density of cells containing oxytocin (OT), vasopressin (AVP), tyrosine hydroxylase (TH), or estrogen receptor alpha (ERα) staining in the brains of social Mongolian gerbils (Meriones unguiculatus) and solitary Chinese striped hamsters (Cricetulus barabensis). Multiple region- and neurochemical-specific species differences were found. In the anterior hypothalamus (AH), Mongolian gerbils had higher densities of AVP-ir and ERα-ir cells than Chinese striped hamsters. In the lateral hypothalamus (LH), Mongolian gerbils also had higher densities of AVP-ir and TH-ir cells, but a lower density of OT-ir cells, than Chinese striped hamsters. Furthermore, in the anterior nucleus of the medial preoptic area (MPOAa), Mongolian gerbils had higher densities of OT-ir and AVP-ir cells than Chinese striped hamsters, and an opposite pattern was found in the posterior nucleus of the MPOA (MPOAp). Some sex differences were also observed. Females of both species had higher densities of TH-ir cells in the MPOAa and of OT-ir cells in the intermediate nucleus of the MPOA (MPOAi) than males. Given the role of these neurochemicals in social behaviors, our data provide additional evidence to support the notion that species-specific patterns of neurochemical expression in the brain may be involved in species differences in social behaviors associated with different life strategies.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Linxi Xu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Yongliang Pan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, United States of America
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
- * E-mail:
| |
Collapse
|
17
|
Abstract
Sex differences in many behaviors such as cognition, mood, and motor skills are well-documented in animals and humans and are regulated by many neural circuits. Sexual dimorphisms within cell populations in these circuits play critical roles in the production of these behavioral dichotomies. Here we focus on three proteins that have well described sexual dimorphisms; calbindin-D28k, a calcium binding protein, tyrosine hydroxylase, the rate limiting enzyme involved in dopamine synthesis and vasopressin, a neuropeptide with central and peripheral sites of action. We describe the sex differences in subpopulations of these proteins, with particular emphasis on laboratory mice. Our thrust is to examine genetic bases of sex differences and how the use of genetically modified models has advanced our understanding of this topic. Regional sex differences in the expression of these three proteins are driven by sex chromosome complement, steroid receptors or in some instances both. While studies of sex differences attributable to sex chromosome genes are still few in number it is exciting to note that this variable factors into expression differences for all three of these proteins. Different genetic mechanisms, which elaborate sex differences, may be employed stochastically in different cell populations. Alternately, general patterns involving the timing of differentiation of the sex differences, relative to the "critical period" in hormonal differences between males and female neonates may emerge. In conclusion, future directions in this area should include examination of the importance of location, timing, steroidal receptor/sex chromosome gene synergy and epigenetics in molding neural sex differences.
Collapse
Affiliation(s)
- Jean LeBeau Abel
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, PO Box 800733, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
18
|
Gilmore RF, Varnum MM, Forger NG. Effects of blocking developmental cell death on sexually dimorphic calbindin cell groups in the preoptic area and bed nucleus of the stria terminalis. Biol Sex Differ 2012; 3:5. [PMID: 22336348 PMCID: PMC3305593 DOI: 10.1186/2042-6410-3-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 02/15/2012] [Indexed: 01/01/2023] Open
Abstract
Background Calbindin-D28 has been used as a marker for the sexually dimorphic nucleus of the preoptic area (SDN-POA). Males have a distinct cluster of calbindin-immunoreactive (ir) cells in the medial preoptic area (CALB-SDN) that is reduced or absent in females. However, it is not clear whether the sex difference is due to the absolute number of calbindin-ir cells or to cell position (that is, spread), and the cellular mechanisms underlying the sex difference are not known. We examined the number of cells in the CALB-SDN and surrounding regions of C57Bl/6 mice and used mice lacking the pro-death gene, Bax, to test the hypothesis that observed sex differences are due to cell death. Methods Experiment 1 compared the number of cells in the CALB-SDN and surrounding regions in adult males, females, and females injected with estradiol benzoate on the day of birth. In experiment 2, cell number in the CALB-SDN and adjacent regions were compared in wild-type and Bax knockout mice of both sexes. In addition, calbindin-ir cells were quantified within the principal nucleus of the bed nucleus of the stria terminalis (BNSTp), a nearby region that is larger in males due to Bax-dependent cell death. Results Males had more cells in the CALB-SDN as well as in surrounding regions than did females, and estradiol treatment of females at birth masculinized both measures. Bax deletion had no effect on cell number in the CALB-SDN or surrounding regions but increased calbindin-ir cell number in the BNSTp. Conclusions The sex difference in the CALB-SDN of mice results from an estrogen-dependent difference in cell number with no evidence found for greater spread of cells in females. Blocking Bax-dependent cell death does not prevent sex differences in calbindin-ir cell number in the BNST or CALB-SDN but increases calbindin-ir cell number in the BNSTp of both sexes.
Collapse
Affiliation(s)
- Richard F Gilmore
- Department of Psychology and Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | |
Collapse
|
19
|
Waddell J, McCarthy MM. Sexual differentiation of the brain and ADHD: what is a sex difference in prevalence telling us? Curr Top Behav Neurosci 2012; 9:341-60. [PMID: 21120649 PMCID: PMC4841632 DOI: 10.1007/7854_2010_114] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Sexual differentiation of the brain is a function of various processes that prepare the organism for successful reproduction in adulthood. Release of gonadal steroids during both the perinatal and the pubertal stages of development organizes many sex differences, producing changes in brain excitability and morphology that endure across the lifespan. To achieve these sexual dimorphisms, gonadal steroids capitalize on a number of distinct mechanisms across brain regions. Comparison of the developing male and female brain provides insight into the mechanisms through which synaptic connections are made, and circuits are organized that mediate sexually dimorphic behaviors. The prevalence of most psychiatric and neurological disorders differ in males versus females, including disorders of attention, activity and impulse control. While there is a strong male bias in incidence of attention deficit and hyperactivity disorders, the source of that bias remains controversial. By elucidating the biological underpinnings of male versus female brain development, we gain a greater understanding of how hormones and genes do and do not contribute to the differential vulnerability in one sex versus the other.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA,
| | | |
Collapse
|
20
|
Petersen SL, Krishnan S, Aggison LK, Intlekofer KA, Moura PJ. Sexual differentiation of the gonadotropin surge release mechanism: a new role for the canonical NfκB signaling pathway. Front Neuroendocrinol 2012; 33:36-44. [PMID: 21741397 DOI: 10.1016/j.yfrne.2011.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 06/09/2011] [Accepted: 06/11/2011] [Indexed: 12/30/2022]
Abstract
Sex differences in luteinizing hormone (LH) release patterns are controlled by the hypothalamus, established during the perinatal period and required for fertility. Female mammals exhibit a cyclic surge pattern of LH release, while males show a tonic release pattern. In rodents, the LH surge pattern is dictated by the anteroventral periventricular nucleus (AVPV), an estrogen receptor-rich structure that is larger and more cell-dense in females. Sex differences result from mitochondrial cell death triggered in perinatal males by estradiol derived from aromatization of testosterone. Herein we provide an historical perspective and an update describing evidence that molecules important for cell survival and cell death in the immune system also control these processes in the developing AVPV. We conclude with a new model proposing that development of the female AVPV requires constitutive activation of the Tnfα, Tnf receptor 2, NfκB and Bcl2 pathway that is blocked by induction of Tnf receptor-associated factor 2-inhibiting protein (Traip) in the male.
Collapse
Affiliation(s)
- Sandra L Petersen
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, United States.
| | | | | | | | | |
Collapse
|
21
|
Semaan SJ, Murray EK, Poling MC, Dhamija S, Forger NG, Kauffman AS. BAX-dependent and BAX-independent regulation of Kiss1 neuron development in mice. Endocrinology 2010; 151:5807-17. [PMID: 20926580 PMCID: PMC2999490 DOI: 10.1210/en.2010-0783] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [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 and its product kisspeptin are important regulators of reproduction. In rodents, Kiss1 is expressed in the hypothalamic arcuate (ARC) and anteroventral periventricular (AVPV)/rostral periventricular (PeN) nuclei. In the AVPV/PeN, females have more Kiss1 and tyrosine hydroxylase (TH) neurons than males. We explored the ontogeny of the Kiss1 sex difference, and the role of cell death in establishing Kiss1 and TH cell number. We also determined whether Kiss1 cells in AVPV/PeN coexpress TH. AVPV/PeN Kiss1 neurons were first detected in both sexes on postnatal d 10, but the Kiss1 sex difference did not emerge until postnatal d 12. The role of BAX-mediated apoptosis in generating this sex difference was tested in adult Bax knockout (KO) and wild-type mice. Deletion of Bax did not diminish the sex difference in Kiss1 expression in the AVPV/PeN. TH expression was sexually dimorphic in the AVPV of both wild-type and Bax KO mice but, unlike Kiss1, was not sexually dimorphic in the PeN of either genotype. Double-label analysis determined that most Kiss1 neurons coexpress TH mRNA, but many TH neurons do not coexpress Kiss1, especially in the PeN. These findings suggest that several subpopulations of TH cells reside within the AVPV/PeN, only one of which coexpresses Kiss1. In the ARC, Kiss1 cell number was markedly increased in Bax KO mice of both sexes, indicating that although BAX-dependent apoptosis does not generate the sex difference in either Kiss1 or TH expression in AVPV/PeN, BAX does importantly regulate Kiss1 cell number in the ARC.
Collapse
Affiliation(s)
- Sheila J Semaan
- Department of Reproductive Medicine, University of California San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | |
Collapse
|
22
|
Cell death and sexual differentiation of behavior: worms, flies, and mammals. Curr Opin Neurobiol 2010; 20:776-83. [PMID: 20934320 DOI: 10.1016/j.conb.2010.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 09/08/2010] [Accepted: 09/09/2010] [Indexed: 11/24/2022]
Abstract
Sex differences in the nervous system are found throughout the animal kingdom. Here, we discuss three prominent genetic models: nematodes, fruit flies, and mice. In all three, differential cell death is central to sexual differentiation and shared molecular mechanisms have been identified. Our knowledge of the precise function of neural sex differences lags behind. One fruitful approach to the 'function' question is to contrast sexual differentiation in standard laboratory animals with differentiation in species exhibiting unique social and reproductive organizations. Advanced genetic strategies are also addressing this question in worms and flies, and may soon be applicable to vertebrates.
Collapse
|
23
|
Central role of TRAF-interacting protein in a new model of brain sexual differentiation. Proc Natl Acad Sci U S A 2009; 106:16692-7. [PMID: 19805359 DOI: 10.1073/pnas.0906293106] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sexually dimorphic brain nuclei underlie gender-specific neural functions and susceptibility to disease, but the developmental basis of dimorphisms is poorly understood. In these studies, we focused on the anteroventral periventricular nucleus (AVPV), a nucleus that is larger in females and critical for the female-typical cyclic surge pattern of luteinizing hormone (LH) release. Sex differences in the size and function of the AVPV result from apoptosis that occurs preferentially in the developing male. To identify upstream pathways responsible for sexual differentiation of the AVPV, we used targeted apoptosis microarrays and in vivo and in vitro follow-up studies. We found that the tumor necrosis factor alpha (TNFalpha)-TNF receptor 2 (TNFR2)-NFkappaB cell survival pathway is active in postnatal day 2 (PND2) female AVPV and repressed in male counterparts. Genes encoding key members of this pathway were expressed exclusively in GABAergic neurons. One gene in particular, TNF receptor-associated factor 2 (TRAF2)-inhibiting protein (trip), was higher in males and it inhibited both TNFalpha-dependent NFkappaB activation and bcl-2 gene expression. The male AVPV also had higher levels of bax and bad mRNA, but neither of these genes was regulated by either TNFalpha or TRIP. Finally, the trip gene was not expressed in the sexually dimorphic nucleus of the preoptic area (SDN-POA), a nucleus in which apoptosis is higher in females than males. These findings form the basis of a new model of sexual differentiation of the AVPV that may also apply to the development of other sexually dimorphic nuclei.
Collapse
|
24
|
Wilson CA, Dakin CL, Rico JA, Golmohamad A, Ahmad-Jauhari Y, Davies DC. The anti-dopaminergic agent, haloperidol, antagonises the feminising effect of neonatal serotonin on sexually dimorphic hypothalamic nuclei and tyrosine hydroxylase immunoreactive neurones. J Neuroendocrinol 2009; 21:648-56. [PMID: 19453825 DOI: 10.1111/j.1365-2826.2009.01883.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is a transient fall in hypothalamic serotonin (5-hydroxytryptamine; 5-HT) activity in the second week post partum in male but not female rats. When this fall is masked by administration of the 5-HT(2) agonist (-) 2,5-dimethoxy-4-iodophenyl]-2-aminopropane hydrochloride [(-)DOI], over days 8-16 post partum, males are feminised in adulthood. To investigate whether the effect of 5-HT is mediated by dopamine and whether testosterone exerts its masculinising effect by reducing 5-HT and dopamine activity, male pups were treated with (-)DOI alone or together with the dopamine antagonist, haloperidol, over days 8-16 post partum, whereas females were treated with testosterone propionate on day 2 post partum. In adulthood, the volumes of the anteroventral periventricular nucleus (AVPV), sexually dimorphic nucleus of the preoptic area (SDN-POA) and arcuate nucleus (ARC) were determined, together with the number of tyrosine hydroxylase-immunoreactive (TH-ir) cells and fibres within them. The concentrations of 5-HT, dopamine and their metabolites were also measured. (-)DOI treatment increased the volume of the AVPV, decreased that of the SDN-POA and increased the number of TH-ir cells in the AVPV. These feminising effects were antagonised by concurrent haloperidol treatment. Neonatal testosterone propionate masculinised the volumes of the female AVPV and SDN-POA and reduced the number of TH-ir cells in the AVPV. Dopamine and 5-HT turnover in the AVPV was greater in female compared to male rats and neonatal testosterone propionate reduced dopamine concentration in the female AVPV. Neonatal (-)DOI had no effect on dopamine and 5-HT activity in the AVPV but increased both in the ARC. The findings that TH-ir neurone number and dopamine activity are greater in the female AVPV; the feminising effect of 5-HT is prevented by a haloperidol; and the masculinising effect of testosterone propionate is accompanied by a decrease in TH-ir neurone number and dopamine concentration in the female AVPV, suggest that dopamine is involved in hypothalamic sexual differentiation and may mediate the effect of 5-HT.
Collapse
Affiliation(s)
- C A Wilson
- Division of Basic Medical Sciences St George's, University of London, London, UK
| | | | | | | | | | | |
Collapse
|
25
|
Broad KD, Curley JP, Keverne EB. Increased apoptosis during neonatal brain development underlies the adult behavioral deficits seen in mice lacking a functional paternally expressed gene 3 (Peg3). Dev Neurobiol 2009; 69:314-25. [PMID: 19224563 DOI: 10.1002/dneu.20702] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Inactivation of the maternally imprinted, paternally expressed gene 3 (Peg3) induces deficits in olfactory function, sexual and maternal behaviors, oxytocin neuron number, metabolic homeostasis and growth. Peg3 is expressed in a number of developing hypothalamic and basal forebrain structures and is a component of the P53 apoptosis pathway. Peg3 inactivation in neuronal cell culture lines inhibits P53 mediated apoptosis, which is important in the early postnatal development and sexual differentiation of the brain. In this study, we investigated the effect of inactivating the Peg3 gene on the incidence of caspase 3 positive cells (a marker of apoptosis) in 4- and 6-day postpartum mouse brain. Inactivating the Peg3 gene resulted in an increase in the incidence of total forebrain caspase 3 positive cells at 4 and 6 days postpartum. Increases in specific neuroanatomical regions including the bed nucleus of the stria terminalis, nucleus accumbens, caudate putamen, medial pre-optic area, arcuate nucleus, medial amygdala, anterior cortical and posteriodorsal amygdaloid nuclei, were also observed. In wild-type mice, sex differences in the incidence of caspase 3 positive cells in the medial amygdala, bed nucleus of the stria terminalis, nucleus accumbens, arcuate nucleus and the M2 motor cortex, were also observed. This neural sex difference was ameliorated in the Peg-3 mutant. These findings suggest that the neuronal and behavioral deficits seen in mice lacking a functional Peg3 gene are mediated by increases in the incidence of early neonatal apoptosis in neuroanatomical regions important for reproductive behavior, olfactory and pheromonal processing, thermoregulation and reward.
Collapse
Affiliation(s)
- Kevin D Broad
- Sub-Department of Animal Behaviour, University of Cambridge, Madingley, Cambridge, CB3 8AA, United Kingdom.
| | | | | |
Collapse
|
26
|
Tsukahara S. Sex differences and the roles of sex steroids in apoptosis of sexually dimorphic nuclei of the preoptic area in postnatal rats. J Neuroendocrinol 2009; 21:370-6. [PMID: 19226350 DOI: 10.1111/j.1365-2826.2009.01855.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The brain contains several sexually dimorphic nuclei that exhibit sex differences with respect to cell number. It is likely that the control of cell number by apoptotic cell death in the developing brain contributes to creating sex differences in cell number in sexually dimorphic nuclei, although the mechanisms responsible for this have not been determined completely. The milieu of sex steroids in the developing brain affects sexual differentiation in the brain. The preoptic region of rats has two sexually dimorphic nuclei. The sexually dimorphic nucleus of the preoptic area (SDN-POA) has more neurones in males, whereas the anteroventral periventricular nucleus (AVPV) has a higher cell density in females. Sex differences in apoptotic cell number arise in the SDN-POA and AVPV of rats in the early postnatal period, and an inverse correlation exists between sex differences in apoptotic cell number and the number of living cells in the mature period. The SDN-POA of postnatal male rats exhibits a higher expression of anti-apoptotic Bcl-2 and lower expression of pro-apoptotic Bax compared to that in females and, as a potential result, apoptotic cell death via caspase-3 activation more frequently occurs in the SDN-POA of females. The patterns of expression of Bcl-2 and Bax in the SDN-POA of postnatal female rats are changed to male-typical ones by treatment with oestrogen, which is normally synthesised from testicular androgen and affects the developing brain in males. In the AVPV of postnatal rats, apoptotic regulation also differs between the sexes, although Bcl-2 expression is increased and Bax expression and caspase-3 activity are decreased in females. The mechanisms of apoptosis possibly contributing to the creation of sex differences in cell number and the roles of sex steroids in apoptosis are discussed.
Collapse
Affiliation(s)
- S Tsukahara
- Research Centre for Environmental Risk, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
27
|
Abstract
The hormonal control of cell death is currently the best-established mechanism for creating sex differences in cell number in the brain and spinal cord. For example, males have more cells than do females in the principal nucleus of the bed nucleus of the stria terminalis (BNSTp) and spinal nucleus of the bulbocavernosus (SNB), whereas females have a cell number advantage in the anteroventral periventricular nucleus (AVPV). In each case, the difference in cell number in adulthood correlates with a sex difference in the number of dying cells at some point in development. Mice with over- or under-expression of cell death genes have been used to test more directly the contribution of cell death to neural sex differences, to identify molecular mechanisms involved, and to determine the behavioural consequences of suppressing developmental cell death. Bax is a pro-death gene of the Bcl-2 family that is singularly important for apoptosis in neural development. In mice lacking bax, the number of cells in the BNSTp, SNB and AVPV are significantly increased, and sex differences in total cell number in each of these regions are eliminated. Cells rescued by bax gene deletion in the BNSTp express markers of differentiated neurones and the androgen receptor. On the other hand, sex differences in other phenotypically identified populations, such as vasopressin-expressing neurones in the BNSTp or dopaminergic neurones in AVPV, are not affected by either bax deletion or bcl-2 over-expression. Possible mechanisms by which testosterone may regulate cell death in the nervous system are discussed, as are the behavioural effects of eliminating sex differences in neuronal cell number.
Collapse
Affiliation(s)
- N G Forger
- Department of Psychology and Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
28
|
Miller SM, Lonstein JS. Dopaminergic projections to the medial preoptic area of postpartum rats. Neuroscience 2009; 159:1384-96. [PMID: 19409227 DOI: 10.1016/j.neuroscience.2009.01.060] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 01/19/2009] [Accepted: 01/27/2009] [Indexed: 11/19/2022]
Abstract
Dopamine receptor activity in the rodent medial preoptic area (mPOA) is crucial for the display of maternal behaviors, as well as numerous other physiological and behavioral functions. However, the origin of dopaminergic input to the mPOA has not been identified through neuroanatomical tracing. To accomplish this, the retrograde tracer Fluorogold was iontophoretically applied to the mPOA of postpartum laboratory rats, and dual-label immunocytochemistry for Fluorogold and tyrosine hydroxylase later performed to identify dopaminergic cells of the forebrain and midbrain projecting to the mPOA. Results indicate that the number of dopaminergic cells projecting to the mPOA is moderate ( approximately 90 cells to one hemisphere), and that these cells have an unexpectedly wide distribution. Even so, more than half of the dual-labeled cells were found in either what has been considered extensions of the A10 dopamine group (particularly the ventrocaudal posterior hypothalamus and adjacent medial supramammillary nucleus), or in the A10 group of the ventral tegmental area. The rostral hypothalamus and surrounding region also contained numerous dual-labeled cells, with the greatest number found within the mPOA itself (including in the anteroventral preoptic area and preoptic periventricular nucleus). Notably, dual-labeled cells were rare in the zona incerta (A13), a site previously suggested to provide dopaminergic input to the mPOA. This study is the first to use anatomical tracing to detail the dopaminergic projections to the mPOA in the laboratory rat, and indicates that much of this projection originates more caudally than previously suggested.
Collapse
Affiliation(s)
- S M Miller
- Department of Psychology, Michigan State University, East Lansing, MI 48824, USA
| | | |
Collapse
|
29
|
de Vries GJ, Jardon M, Reza M, Rosen GJ, Immerman E, Forger NG. Sexual differentiation of vasopressin innervation of the brain: cell death versus phenotypic differentiation. Endocrinology 2008; 149:4632-7. [PMID: 18499746 PMCID: PMC2553370 DOI: 10.1210/en.2008-0448] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In most vertebrates studied, males have more vasopressin (VP) cells in the bed nucleus of the stria terminalis, or homologous vasotocin cells in nonmammalian species, than females. Previous research excluded differential cell birth and migration as likely mechanisms underlying this difference, leaving just differential cell death and phenotypic differentiation of existing cells. To differentiate between these remaining possibilities, we compared VP cell number in wild-type mice vs. mice overexpressing the anti-cell death factor, Bcl-2. All animals were gonadectomized in adulthood and given testosterone capsules. Three weeks later, brains were processed for in situ hybridization to identify VP cells. Bcl-2 overexpression increased VP cell number in both sexes but did not reduce the sex difference. We repeated this experiment in mice with a null mutation of the pro-cell death gene, Bax, and obtained similar results; cell number was increased in Bax(-/-) mice of both sexes, but males had about 40% more VP cells, regardless of Bax gene status. Taken together, cell death is unlikely to account for the sex difference in VP cell number, leaving differentiation of cell phenotype as the most likely underlying mechanism. We also used immunocytochemistry to examine VP projections in Bcl-2-overexpressing mice. As expected, males showed denser VP-immunoreactive fibers than females in the lateral septum, a projection area of the bed nucleus of the stria terminalis. However, even though Bcl-2 overexpression increased VP cell number, it did not affect fiber density. Thus, a compensatory mechanism may control total septal innervation regardless of the number of contributing cells.
Collapse
Affiliation(s)
- Geert J de Vries
- Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Schwarz JM, McCarthy MM. Steroid-induced sexual differentiation of the developing brain: multiple pathways, one goal. J Neurochem 2008; 105:1561-72. [PMID: 18384643 PMCID: PMC2565863 DOI: 10.1111/j.1471-4159.2008.05384.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hormone exposure, including testosterone and its metabolite estradiol, induces a myriad of effects during a critical period of brain development that are necessary for brain sexual differentiation. Nuclear volume, neuronal morphology, and astrocyte complexity are examples of the wide range of effects by which testosterone and estradiol can induce permanent changes in the function of neurons for the purpose of reproduction in adulthood. This review will examine the multitude of mechanisms by which steroid hormones induce these permanent changes in brain structure and function. Elucidating how steroids alter brain development sheds light on how individual variation in neuronal phenotype is established during a critical period.
Collapse
Affiliation(s)
- Jaclyn M Schwarz
- Program in Neuroscience, University of Maryland, Baltimore, Baltimore, Maryland 21201, USA.
| | | |
Collapse
|
31
|
Gore AC. Developmental programming and endocrine disruptor effects on reproductive neuroendocrine systems. Front Neuroendocrinol 2008; 29:358-74. [PMID: 18394690 PMCID: PMC2702520 DOI: 10.1016/j.yfrne.2008.02.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2007] [Revised: 02/21/2008] [Accepted: 02/22/2008] [Indexed: 01/01/2023]
Abstract
The ability of a species to reproduce successfully requires the careful orchestration of developmental processes during critical time points, particularly the late embryonic and early postnatal periods. This article begins with a brief presentation of the evidence for how gonadal steroid hormones exert these imprinting effects upon the morphology of sexually differentiated hypothalamic brain regions, the mechanisms underlying these effects, and their implications in adulthood. Then, I review the evidence that aberrant exposure to hormonally-active substances such as exogenous endocrine-disrupting chemicals (EDCs), may result in improper hypothalamic programming, thereby decreasing reproductive success in adulthood. The field of endocrine disruption has shed new light on the discipline of basic reproductive neuroendocrinology through studies on how early life exposures to EDCs may alter gene expression via non-genomic, epigenetic mechanisms, including DNA methylation and histone acetylation. Importantly, these effects may be transmitted to future generations if the germline is affected via transgenerational, epigenetic actions. By understanding the mechanisms by which natural hormones and xenobiotics affect reproductive neuroendocrine systems, we will gain a better understanding of normal developmental processes, as well as develop the potential ability to intervene when development is disrupted.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology and Toxicology, Institute for Neuroscience and Institute for Cellular and Molecular Biology, The University of Texas at Austin, 1 University Station A1915, Austin, TX 78712, USA.
| |
Collapse
|
32
|
Foecking EM, McDevitt MA, Acosta-Martínez M, Horton TH, Levine JE. Neuroendocrine consequences of androgen excess in female rodents. Horm Behav 2008; 53:673-92. [PMID: 18374922 PMCID: PMC2413177 DOI: 10.1016/j.yhbeh.2007.12.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Revised: 12/17/2007] [Accepted: 12/18/2007] [Indexed: 12/28/2022]
Abstract
Androgens exert significant organizational and activational effects on the nervous system and behavior. Despite the fact that female mammals generally produce low levels of androgens, relative to the male of the same species, increasing evidence suggests that androgens can exert profound effects on the normal physiology and behavior of females during fetal, neonatal, and adult stages of life. This review examines the effects of exposure to androgens at three stages of development--as an adult, during early postnatal life and as a fetus, on reproductive hormone secretions in female rats. We examine the effects of androgen exposure both as a model of neuroendocrine sexual differentiation and with respect to the role androgens play in the normal female. We then discuss the hypothesis that androgens may cause epigenetic modification of estrogen target genes in the brain. Finally we consider the clinical consequences of excess androgen exposure in women.
Collapse
Affiliation(s)
- Eileen M Foecking
- Department of Neurobiology and Physiology, Northwestern University, Evanston, IL 60208, USA
| | | | | | | | | |
Collapse
|
33
|
Juntti SA, Coats JK, Shah NM. A genetic approach to dissect sexually dimorphic behaviors. Horm Behav 2008; 53:627-37. [PMID: 18313055 PMCID: PMC2464277 DOI: 10.1016/j.yhbeh.2007.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Revised: 12/20/2007] [Accepted: 12/24/2007] [Indexed: 12/29/2022]
Abstract
It has been known since antiquity that gender-specific behaviors are regulated by the gonads. We now know that testosterone is required for the appropriate display of male patterns of behavior. Estrogen and progesterone, on the other hand, are essential for female typical responses. Research from several groups also indicates that estrogen signaling is required for male typical behaviors. This finding raises the issue of the relative contribution of these two hormonal systems in the control of male typical behavioral displays. In this review we discuss the findings that led to these conclusions and suggest various genetic strategies that may be required to understand the relative roles of testosterone and estrogen signaling in the control of gender-specific behavior.
Collapse
Affiliation(s)
| | | | - Nirao M. Shah
- 1550 4th Street, MC2722, Neuroscience Graduate Program, Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
34
|
Dakin CL, Wilson CA, Kalló I, Coen CW, Davies DC. Neonatal stimulation of 5-HT2receptors reduces androgen receptor expression in the rat anteroventral periventricular nucleus and sexually dimorphic preoptic area. Eur J Neurosci 2008; 27:2473-80. [DOI: 10.1111/j.1460-9568.2008.06216.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Abstract
There is accumulating evidence for nongenetic transgenerational inheritance with conspicuous marked sexual dimorphism for both the modes of transmission and the effects. Given the critical spatiotemporal windows, the role of the sex chromosomes, the regulatory pathways underlying sexual differentiation during gonad and brain development, and other developmental processes, as well as the lifelong impact of sex hormones, it is not surprising that most of the common diseases, which often take root in early development, display some degree of sex bias. The flexibility of epigenetic marks may make it possible for environmental and nutritional factors, or endocrine disruptors to alter-during a particular spatiotemporal window in a sex-specific manner-the sex-specific methylation or demethylation of specific CpGs and histone/chromatin modifications underlying sex-specific expression of a substantial proportion of genes. Thus, finely tuned developmental program aspects, specific to one sex, may be more sensitive to specific environmental challenges, particularly during developmental programming and gametogenesis, but also throughout the individual's life under the influence of sex steroid hormones. This review highlights the importance of studying both sexes in epidemiologic protocols or dietary interventions both in humans and in experimental models in animals.
Collapse
Affiliation(s)
- Alexandre Vigé
- Inserm U781 Genetics and Epigenetics of Metabolic Diseases, Neurosensorial Diseases and Development, Hôpital Necker-Enfants Malades, Université Paris Deseartes, Paris, France
| | | | | |
Collapse
|
36
|
Tsukahara S, Hojo R, Kuroda Y, Fujimaki H. Estrogen modulates Bcl-2 family protein expression in the sexually dimorphic nucleus of the preoptic area of postnatal rats. Neurosci Lett 2007; 432:58-63. [PMID: 18164816 DOI: 10.1016/j.neulet.2007.12.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 12/02/2007] [Accepted: 12/04/2007] [Indexed: 11/15/2022]
Abstract
In the sexually dimorphic nucleus of the preoptic area (SDN-POA) of postnatal rats, apoptotic cells are detected more frequently in females than males. This sex difference is under the influence of aromatized androgen. We have reported that there are sex differences in the levels of Bcl-2 (female<male) and Bax (female>male) in the central division of the medial preoptic nucleus (MPNc), a significant component of the SDN-POA, followed by a sex difference in induction of apoptosis via caspase-3 activation (female>male). In the present study, we examined effects of estradiol benzoate (EB) on expression of Bcl-2 and Bax in the MPNc. Female rats were subcutaneously injected with EB (25 or 50 microg per head) on postnatal day 5. MPNc and caudate putamen (CP) tissues were obtained from EB-treated female and male rats on postnatal day 6. Protein levels of Bcl-2 and Bax were determined by Western blotting. In the MPNc of female rats, EB at a dose of 50 microg/head but not 25 microg/head significantly increased Bcl-2 protein level and decreased Bax protein level. The levels of Bcl-2 and Bax of female rats treated with 50 microg of EB were comparable to those of male rats. However, the protein levels of Bcl-2 and Bax in the CP did not change with EB treatment. These results suggest that estrogen up-regulates Bcl-2 expression and down-regulates Bax expression in the MPNc of postnatal rats. Effects of estrogen on the Bcl-2 family are presumably responsible for sex difference in postnatal apoptosis of the SDN-POA.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Research Center for Environmental Risk, National Institute for Environmental Studies, Onogawa 16-2, Tsukuba, Ibaraki 305-8506, Japan.
| | | | | | | |
Collapse
|
37
|
Jyotika J, McCutcheon J, Laroche J, Blaustein JD, Forger NG. Deletion of the Bax gene disrupts sexual behavior and modestly impairs motor function in mice. Dev Neurobiol 2007; 67:1511-9. [PMID: 17525992 DOI: 10.1002/dneu.20525] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cell death is a nearly ubiquitous feature of the developing nervous system, and differential death in males and females contributes to several well studied sex differences in neuron number. Nonetheless, the functional importance of neuronal cell death has been subjected to few direct tests. Bax, a pro-apoptotic protein, is required for cell death in many neural regions. Deletion of the Bax gene in mice increases neuron number in several areas and eliminates sex differences in cell number in the brain and spinal cord. Here, sexual and motor behaviors were examined in Bax-/- mice and their wild-type siblings to test the functional consequences of preventing Bax-dependent cell death. Animals were gonadectomized in adulthood and provided with ovarian hormones or with testosterone for tests of feminine and masculine sexual behaviors, respectively. Wild-type mice exhibited a sex difference in feminine sexual behavior, with high lordosis scores in females and low scores in males. This sex difference was eliminated by Bax deletion, with very low receptivity exhibited by both male and female Bax-/- mice. Masculine sexual behavior was not sexually dimorphic among wild-type mice, but mounts and pelvic thrusts were nearly eliminated in Bax-/- mice of both sexes. Motor strength and performance at low speeds on a RotaRod apparatus did not differ by sex or Bax gene status. However, Bax-/- animals exhibited impairments on the RotaRod at higher speeds. Thus, developmental cell death may be required for masculine and feminine sexual behaviors and the fine tuning of motor coordination.
Collapse
Affiliation(s)
- Jigyasa Jyotika
- Department of Psychology and Neuroscience and Behavior Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | | | | | | |
Collapse
|
38
|
Northcutt KV, Wang Z, Lonstein JS. Sex and species differences in tyrosine hydroxylase-synthesizing cells of the rodent olfactory extended amygdala. J Comp Neurol 2007; 500:103-15. [PMID: 17099901 DOI: 10.1002/cne.21148] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bed nucleus of the stria terminalis (BST) and the medial amygdala (MeA) are anatomically connected sites necessary for chemosensory regulation of social behaviors in rodents. Prairie voles (Microtus ochrogaster) are a valuable model for studying the neural regulation of social behaviors because, unlike many other rodents, they are gregarious, pair bond after copulating, and are biparental. We herein describe sex and species differences in immunoreactivity for tyrosine hydroxylase (TH), the rate-limiting enzyme for catecholamine synthesis, in the BST and MeA. Virgin male prairie voles had a large number of TH-immunoreactive cells in areas analogous to the rat principal nucleus of the BST (pBST) and the posterodorsal medial amygdala (MeAPd). Virgin female prairie voles had far fewer TH-immunoreactive cells in these sites ( approximately 17% of the number of cells as males in the pBST, approximately 35% of the number of cells in the MeAPd). A few TH-immunoreactive cells were found in the BST of male and female hamsters and meadow voles, but not in rats. The MeApd also contained a few TH-immunoreactive cells in male and female hamsters and male meadow voles, but not rats. Castration greatly reduced the number of TH-immunoreactive cells in the male prairie vole pBST and MeAPd, an effect that could be reversed with testosterone. Furthermore, treating ovariectomized females with testosterone substantially increased TH-immunoreactive cells in both sites. Therefore, a species-specific sex difference in TH expression is found in a chemosensory pathway in prairie voles. Expression of TH in these sites is influenced by circulating gonadal hormones in adults, which may be related to changes in their display of social behaviors across the reproductive cycle.
Collapse
|
39
|
Wilson CA, Davies DC. The control of sexual differentiation of the reproductive system and brain. Reproduction 2007; 133:331-59. [PMID: 17307903 DOI: 10.1530/rep-06-0078] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This review summarizes current knowledge of the genetic and hormonal control of sexual differentiation of the reproductive system, brain and brain function. While the chromosomal regulation of sexual differentiation has been understood for over 60 years, the genes involved and their actions on the reproductive system and brain are still under investigation. In 1990, the predicted testicular determining factor was shown to be theSRYgene. However, this discovery has not been followed up by elucidation of the actions of SRY, which may either stimulate a cascade of downstream genes, or inhibit a suppressor gene. The number of other genes known to be involved in sexual differentiation is increasing and the way in which they may interact is discussed. The hormonal control of sexual differentiation is well-established in rodents, in which prenatal androgens masculinize the reproductive tract and perinatal oestradiol (derived from testosterone) masculinizes the brain. In humans, genetic mutations have revealed that it is probably prenatal testosterone that masculinizes both the reproductive system and the brain. Sexual differentiation of brain structures and the way in which steroids induce this differentiation, is an active research area. The multiplicity of steroid actions, which may be specific to individual cell types, demonstrates how a single hormonal regulator, e.g. oestradiol, can exert different and even opposite actions at different sites. This complexity is enhanced by the involvement of neurotransmitters as mediators of steroid hormone actions. In view of current environmental concerns, a brief summary of the effects of endocrine disruptors on sexual differentiation is presented.
Collapse
Affiliation(s)
- C A Wilson
- Basic Medical Sciences, Clinical Developmental Sciences, St George's, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK.
| | | |
Collapse
|
40
|
Bu L, Lephart ED. AVPV neurons containing estrogen receptor-beta in adult male rats are influenced by soy isoflavones. BMC Neurosci 2007; 8:13. [PMID: 17266774 PMCID: PMC1797051 DOI: 10.1186/1471-2202-8-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 02/01/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Isoflavones, the most abundant phytoestrogens in soy foods, are structurally similar to 17beta-estradiol. It is known that 17beta-estradiol induces apoptosis in anteroventral periventricular nucleus (AVPV) in rat brain. Also, there is evidence that consumption of soy isoflavones reduces the volume of AVPV in male rats. Therefore, in this study, we examined the influence of dietary soy isoflavones on apoptosis in AVPV of 150 day-old male rats fed either a soy isoflavone-free diet (Phyto-free) or a soy isoflavone-rich diet (Phyto-600). RESULTS The occurrence of apoptosis in AVPV was examined by TUNEL staining. The incidence of apoptosis was about 10 times higher in the Phyto-600 group (33.1 +/- 1.7%) than in the Phyto-free group (3.6 +/- 1.0%). Furthermore, these apoptotic cells were identified as neurons by dual immunofluorescent staining of GFAP and NeuN as markers of astrocytes and neurons, respectively. Then the dopaminergic neurons in AVPV were detected by immunohistochemistry staining of tyrosine hydroxylase (TH). No significant difference in the number of TH neurons was observed between the diet treatment groups. When estrogen receptor (ER) alpha and beta were examined by immunohistochemistry, we observed a 22% reduction of ERbeta-positive cell numbers in AVPV with consumption of soy isoflavones, whereas no significant change in ERalpha-positive cell numbers was detected. Furthermore, almost all the apoptotic cells were ERbeta-immunoreactive (ir), but not ERalpha-ir. Last, subcutaneous injections of equol (a major isoflavone metabolite) that accounts for approximately 70-90% of the total circulating plasma isoflavone levels did not alter the volume of AVPV in adult male rats. CONCLUSION In summary, these findings provide direct evidence that consumption of soy isoflavones, but not the exposure to equol, influences the loss of ERbeta-containing neurons in male AVPV.
Collapse
Affiliation(s)
- Lihong Bu
- Physiology and Developmental Biology Department and Neuroscience Center Brigham Young University, Provo, UT, USA
- Division of Newborn Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA, 02115, USA
| | - Edwin D Lephart
- Physiology and Developmental Biology Department and Neuroscience Center Brigham Young University, Provo, UT, USA
| |
Collapse
|
41
|
Tsukahara S, Kakeyama M, Toyofuku Y. Sex differences in the level of Bcl-2 family proteins and caspase-3 activation in the sexually dimorphic nuclei of the preoptic area in postnatal rats. ACTA ACUST UNITED AC 2007; 66:1411-9. [PMID: 17013925 DOI: 10.1002/neu.20276] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In developing rats, sex differences in the number of apoptotic cells are found in the central division of the medial preoptic nucleus (MPNc), which is a significant component of the sexually dimorphic nucleus of the preoptic area, and in the anteroventral periventricular nucleus (AVPV). Specifically, male rats have more apoptotic cells in the developing AVPV, whereas females have more apoptotic cells in the developing MPNc. To determine the mechanisms for the sex differences in apoptosis in these nuclei, we compared the expression of the Bcl-2 family members and active caspase-3 in postnatal female and male rats. Western blot analyses for the Bcl-2 family proteins were performed using preoptic tissues isolated from the brain on postnatal day (PD) 1 (day of birth) or on PD8. In the AVPV-containing tissues of PD1 rats, there were significant sex differences in the level of Bcl-2 (female > male) and Bax (female < male) proteins, but not of Bcl-xL or Bad proteins. In the MPNc-containing tissues of PD8 rats, there were significant sex differences in the protein levels for Bcl-2 (female < male), Bax (female > male), and Bad (female < male), but not for Bcl-xL. Immunohistochemical analyses showed significant sex differences in the number of active caspase-3-immunoreactive cells in the AVPV on PD1 (female < male) and in the MPNc on PD8 (female > male). We further found that active caspase-3-immunoreactive cells of the AVPV and MPNc were immunoreactive for NeuN, a neuronal marker. These results suggest that there are sex differences in the induction of apoptosis via the mitochondrial pathway during development of the AVPV and MPNc.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Environmental Health Science Division, National Institute for Environmental Studies, Onogawa16-2, Tsukuba 305-8506, Japan.
| | | | | |
Collapse
|
42
|
Rubin BS, Lenkowski JR, Schaeberle CM, Vandenberg LN, Ronsheim PM, Soto AM. Evidence of altered brain sexual differentiation in mice exposed perinatally to low, environmentally relevant levels of bisphenol A. Endocrinology 2006; 147:3681-91. [PMID: 16675520 DOI: 10.1210/en.2006-0189] [Citation(s) in RCA: 255] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Humans are routinely exposed to bisphenol A (BPA), an estrogenic chemical present in food and beverage containers, dental composites, and many products in the home and workplace. BPA binds both classical nuclear estrogen receptors and facilitates membrane-initiated estrogenic effects. Here we explore the ability of environmentally relevant exposure to BPA to affect anatomical and functional measures of brain development and sexual differentiation. Anatomical evidence of alterations in brain sexual differentiation were examined in male and female offspring born to mouse dams exposed to 0, 25, or 250 ng BPA/kg body weight per day from the evening of d 8 of gestation through d 16 of lactation. These studies examined the sexually dimorphic population of tyrosine hydroxylase (TH) neurons in the rostral periventricular preoptic area, an important brain region for estrous cyclicity and estrogen-positive feedback. The significant sex differences in TH neuron number observed in control offspring were diminished or obliterated in offspring exposed to BPA primarily because of a decline in TH neuron number in BPA-exposed females. As a functional endpoint of BPA action on brain sexual differentiation, we examined the effects of perinatal BPA exposure on sexually dimorphic behaviors in the open field. Data from these studies revealed significant sex differences in the vehicle-exposed offspring that were not observed in the BPA-exposed offspring. These data indicate that BPA may be capable of altering important events during critical periods of brain development.
Collapse
Affiliation(s)
- Beverly S Rubin
- Tufts University, School of Medicine, Boston, Massachusetts 02111, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The programmed cell death (PCD) of developing cells is considered an essential adaptive process that evolved to serve diverse roles. We review the putative adaptive functions of PCD in the animal kingdom with a major focus on PCD in the developing nervous system. Considerable evidence is consistent with the role of PCD in events ranging from neurulation and synaptogenesis to the elimination of adult-generated CNS cells. The remarkable recent progress in our understanding of the genetic regulation of PCD has made it possible to perturb (inhibit) PCD and determine the possible repercussions for nervous system development and function. Although still in their infancy, these studies have so far revealed few striking behavioral or functional phenotypes.
Collapse
Affiliation(s)
- Robert R Buss
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | |
Collapse
|
44
|
Nikolakopoulou AM, Parpas A, Panagis L, Zikopoulos B, Dermon CR. Early post-hatching sex differences in cell proliferation and survival in the quail telencephalic ventricular zone and intermediate medial mesopallium. Brain Res Bull 2006; 70:107-16. [PMID: 16782501 DOI: 10.1016/j.brainresbull.2006.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 04/19/2006] [Accepted: 04/21/2006] [Indexed: 02/05/2023]
Abstract
Previous studies indicated that avian telencephalic areas related to learned behavior, such as song perception and production, are sexually dimorphic. Our study focused on the eventual occurrence of dimorphism in the intermediate medial mesopallium, an area associated with learning in non-singing birds. During early post-hatching life (days 1 and 5) cell proliferation and survival of newborn cells were studied by means of 5-bromo-2-deoxy-uridine immunocytochemistry. Programmed cell death (apoptosis) was investigated at post-hatching day 10. The ventricular zone, intermediate medial part of mesopallium and lateral septal area was analyzed using stereological methods for cell counts. Short-term experiments revealed significantly higher numbers of newborn cells in male ventricular zone of mesopallium compared to female at post-hatching day 1. Long-term survival until post-hatching day 20 showed significantly higher numbers of labeled cells in the male compared to female intermediate medial part of mesopallium, which is the final destination of migrating cells born in the overlying ventricular zone. The vast majority of these early post-hatching newborn cells residing in the intermediate medial part of mesopallium expressed a neuronal phenotype. In addition to neurogenesis, higher numbers of apoptotic figures were found in the male intermediate medial part of mesopallium at post-hatching day 10, suggesting that cell death plays a role in the control of telencephalic regional cell density in males. Our findings indicate that sex-specific mechanisms possibly stimulate increased cell genesis and survival, as well as the counteracting event of increased apoptotic cell death that characterized the male intermediate medial part of mesopallium.
Collapse
|
45
|
Lansing SW, Lonstein JS. Tyrosine hydroxylase-synthesizing cells in the hypothalamus of prairie voles (Microtus ochrogaster): sex differences in the anteroventral periventricular preoptic area and effects of adult gonadectomy or neonatal gonadal hormones. ACTA ACUST UNITED AC 2006; 66:197-204. [PMID: 16329116 DOI: 10.1002/neu.20212] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The vertebrate hypothalamus and surrounding region contain a large population of cells expressing tyrosine hydroxylase (TH), the rate limiting enzyme for synthesis of dopamine and other catecholamines. Some of these populations are sexually dimorphic in rats. We here examined sex differences in TH-immunoreactive populations in the forebrain of gonadally intact and gonadectomized prairie voles (Microtus ochrogaster), a species that sometimes shows unusual sexual differentiation of brain and behavior. A sex difference was found in the anteroventral periventricular preoptic area (AVPV; likely analogous to the rat rostral A14) only in gonadectomized subjects, which was due to a 50% reduction in the number of TH-immunoreactive cells after castration in males. There was no significant sex difference or effects of gonadectomy on the number of TH-immunoreactive cells in the anteroventral preoptic area (AVP), periventricular anterior hypothalamus (caudal A14), arcuate nucleus (A12), zona incerta (A13), or posterodorsal hypothalamus (A11). In a second experiment, testosterone propionate (TP; 500 microg), diethylstilbestrol (DES; 1 microg), or estradiol benzoate (EB; 30 microg) injected daily during the first week after birth each significantly reduced later TH expression in the AVPV of females by approximately 40-65% compared to oil-treated controls. Unlike rats, therefore, a sex difference in TH expression in the prairie vole AVPV is found only after removal of circulating gonadal hormones in males. Furthermore, unlike our previous findings on the generation of sex differences in extra-hypothalamic arginine-vasopressin expression in prairie voles, TH expression in the AVPV of female prairie voles can be highly masculinized by neonatal exposure to either aromatizable androgens or estrogens.
Collapse
Affiliation(s)
- Sarah W Lansing
- Neuroscience Program & Department of Psychology, Michigan State University, Giltner Hall, East Lansing, Michigan 48823, USA
| | | |
Collapse
|
46
|
Forger NG. Cell death and sexual differentiation of the nervous system. Neuroscience 2005; 138:929-38. [PMID: 16310316 DOI: 10.1016/j.neuroscience.2005.07.006] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 06/17/2005] [Accepted: 07/10/2005] [Indexed: 10/25/2022]
Abstract
Sex differences in nuclear volume or neuron number often are attributed to the hormonal control of cell death. In the spinal nucleus of the bulbocavernosus, the central portion of the medial preoptic nucleus, and the principal nucleus of the bed nucleus of the stria terminalis testicular hormones decrease cell death during perinatal life, resulting in a male advantage in neuron number in adulthood. Conversely, males have more dying cells during development and fewer neurons in adulthood than do females in the anteroventral periventricular nucleus of the hypothalamus. This review discusses several limitations and unresolved issues in the literature on sexually dimorphic cell death, and identifies molecular mechanisms by which gonadal steroids may control cell survival. In particular, evidence is presented for the hormonal regulation of neurotrophic factors and involvement of Bcl-2 family proteins in the determination of sex differences in neuron number.
Collapse
Affiliation(s)
- N G Forger
- Department of Psychology, Tobin Hall, University of Massachusetts, Amherst, 01003, USA.
| |
Collapse
|
47
|
Abstract
The hormones that regulate the hypothalamic circuits that control essential functions, such as reproduction and energy homeostasis, also specify brain architecture by regulating key developmental events. The cellular mechanisms underlying the developmental actions of testosterone and estrogen to determine patterns of neuronal cell death, synaptogenesis and axon guidance are being identified. Recent neuroanatomical evidence indicates that the adipocyte-derived hormone leptin may direct the development of hypothalamic pathways involved in energy homeostasis by promoting axonal projections from the arcuate nucleus of the hypothalamus to other hypothalamic sites that mediate the effects of leptin on food intake and body weight. Understanding how sex steroids and leptin regulate hypothalamic development will enable us to identify hormonally directed signaling events essential to the specification of neural circuitry that is optimized for sustained homeostasis.
Collapse
Affiliation(s)
- Richard B Simerly
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA.
| |
Collapse
|
48
|
Abstract
The consequences of eliminating the process of programmed cell death during the development of the nervous system is examined by reviewing studies in the genetic model organisms Caenorhabditis elegans, Drosophila melanogaster, Danio rerio and Mus musculus, where mutations of cell death genes have eliminated or reduced programmed cell death in the nervous system. In many cases, genetic elimination of cell death leads to embryonic mortality or gross anatomical malformations; however, there are cases where animals develop normally but with excess neurons and glia in the nervous system. Undead cells either differentiate and function as working neurons, in some instances being of smaller size, or fail to differentiate and lack normal connections with their targets. Changes in motor control and sensory processing are generally not observed, except for during the most complex of behaviors. Examination of organisms where death genes have been genetically eliminated reveals that programmed cell death may play an important role in sculpting gross brain structure during early development of the neural tube. In contrast, the consequences of preventing neuronal cell death at later developmental stages (e.g. during vertebrate synapse formation) are just beginning to be understood.
Collapse
Affiliation(s)
- Robert R Buss
- Department of Neurobiology and Anatomy and the Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157-1010, USA
| | | |
Collapse
|
49
|
Morris JA, Jordan CL, Breedlove SM. Sexual differentiation of the vertebrate nervous system. Nat Neurosci 2004; 7:1034-9. [PMID: 15452574 DOI: 10.1038/nn1325] [Citation(s) in RCA: 448] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Accepted: 08/13/2004] [Indexed: 11/09/2022]
Abstract
Understanding the mechanisms that give rise to sex differences in the behavior of nonhuman animals may contribute to the understanding of sex differences in humans. In vertebrate model systems, a single factor-the steroid hormone testosterone-accounts for most, and perhaps all, of the known sex differences in neural structure and behavior. Here we review some of the events triggered by testosterone that masculinize the developing and adult nervous system, promote male behaviors and suppress female behaviors. Testosterone often sculpts the developing nervous system by inhibiting or exacerbating cell death and/or by modulating the formation and elimination of synapses. Experience, too, can interact with testosterone to enhance or diminish its effects on the central nervous system. However, more work is needed to uncover the particular cells and specific genes on which testosterone acts to initiate these events.
Collapse
Affiliation(s)
- John A Morris
- Neuroscience Program, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | |
Collapse
|
50
|
Forger NG, Rosen GJ, Waters EM, Jacob D, Simerly RB, de Vries GJ. Deletion of Bax eliminates sex differences in the mouse forebrain. Proc Natl Acad Sci U S A 2004; 101:13666-71. [PMID: 15342910 PMCID: PMC518810 DOI: 10.1073/pnas.0404644101] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Several of the best-studied sex differences in the mammalian brain are ascribed to the hormonal control of cell death. This conclusion is based primarily on correlations between pyknotic cell counts in development and counts of mature neurons in adulthood; the molecular mechanisms of hormone-regulated, sexually dimorphic cell death are unknown. We asked whether Bax, a member of the Bcl-2 family of proteins that is required for cell death in many developing neurons, might be essential for sex differences in neuron number. We compared Bax knockout mice and their WT siblings, focusing on two regions of the mouse forebrain that show opposite patterns of sexual differentiation: the principal nucleus of the bed nucleus of the stria terminalis, in which males have more neurons than do females, and the anteroventral periventricular nucleus (AVPV), where females have more neurons overall and many more dopaminergic neurons than do males. Testosterone, or its metabolites, is responsible for the sex differences in both nuclei. A null mutation of the Bax gene completely eliminated sex differences in overall cell number in both the principal nucleus of the bed nucleus of the stria terminalis and AVPV. Thus, Bax-dependent cell death is required for sexual differentiation of cell number, regardless of whether testosterone decreases or increases cell death. In contrast, the sex difference in AVPV dopaminergic cell number, as measured by tyrosine hydroxylase immunohistochemistry, was not affected by Bax gene deletion, demonstrating heterogeneity of mechanisms controlling cell number within a single nucleus.
Collapse
Affiliation(s)
- Nancy G Forger
- Department of Psychology and Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | | | | | | | |
Collapse
|