1
|
Robinson CD, Hale MD, Cox CL, John-Alder HB, Cox RM. Effects of Testosterone on Gene Expression Are Concordant between Sexes but Divergent across Species of Sceloporus Lizards. Am Nat 2024; 204:517-532. [PMID: 39486031 DOI: 10.1086/732200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
AbstractHormones mediate sexual dimorphism by regulating sex-specific patterns of gene expression, but it is unclear how much of this regulation involves sex-specific hormone levels versus sex-specific transcriptomic responses to the same hormonal signal. Moreover, transcriptomic responses to hormones can evolve, but the extent to which hormonal pleiotropy in gene regulation is conserved across closely related species is not well understood. We addressed these issues by elevating testosterone levels in juvenile females and males of three Sceloporus lizard species before sexual divergence in circulating testosterone and then characterizing transcriptomic responses in the liver. In each species, more genes were responsive to testosterone in males than in females, suggesting that early developmental processes prime sex-specific transcriptomic responses to testosterone later in life. However, overall transcriptomic responses to testosterone were concordant between sexes, with no genes exhibiting sex-by-treatment interactions. By contrast, hundreds of genes exhibited species-by-treatment interactions, particularly when comparing distantly related species with different patterns of sexual dimorphism, suggesting evolutionary lability in gene regulation by testosterone. Collectively, our results indicate that early organizational effects may lead to sex-specific differences in the magnitude, but not the direction, of transcriptomic responses to testosterone and that the hormone-genome interface accrues regulatory changes over evolutionary time.
Collapse
|
2
|
Tian Y, Qiao H, Zhu LQ, Man HY. Sexually dimorphic phenotypes and the role of androgen receptors in UBE3A-dependent autism spectrum disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592248. [PMID: 38746146 PMCID: PMC11092617 DOI: 10.1101/2024.05.02.592248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Autism spectrum disorders (ASDs) are characterized by social, communication, and behavioral challenges. UBE3A is one of the most common ASD genes. ASDs display a remarkable sex difference with a 4:1 male to female prevalence ratio; however, the underlying mechanism remains largely unknown. Using the UBE3A-overexpressing mouse model for ASD, we studied sex differences at behavioral, genetic, and molecular levels. We found that male mice with extra copies of Ube3A exhibited greater impairments in social interaction, repetitive self-grooming behavior, memory, and pain sensitivity, whereas female mice with UBE3A overexpression displayed greater olfactory defects. Social communication was impaired in both sexes, with males making more calls and females preferring complex syllables. At the molecular level, androgen receptor (AR) levels were reduced in both sexes due to enhanced degradation mediated by UBE3A. However, AR reduction significantly dysregulated AR target genes only in male, not female, UBE3A-overexpressing mice. Importantly, restoring AR levels in the brain effectively normalized the expression of AR target genes, and rescued the deficits in social preference, grooming behavior, and memory in male UBE3A-overexpressing mice, without affecting females. These findings suggest that AR and its signaling cascade play an essential role in mediating the sexually dimorphic changes in UBE3A-dependent ASD.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Hui Qiao
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA 02215, USA
| |
Collapse
|
3
|
Barcelos MN, Gonçalves-Santos E, Souza MA, Santos EC, Gonçalves RV, Castro-Gamero AM, Novaes RD. Prolonged testosterone 17β-cyclopentylpropionate exposition induces behavioral, ovarian, oviductal, uterine and reproductive disturbances in female mice. Life Sci 2024; 338:122408. [PMID: 38181852 DOI: 10.1016/j.lfs.2023.122408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/16/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
Anabolic-androgenic steroids (AAS) abuse is often associated with metabolic disorders and infertility. However, the current evidence on AAS-induced reproductive toxicity is mainly based on male studies. Thus, AAS repercussions on female reproductive capacity remain poorly understood, despite scarce evidence that fertility determinants may be more severely impaired in females than males exposed to these drugs. Accordingly, this study used an integrated framework to investigate the impact of different testosterone 17β-cyclopentylpropionate (TC) doses on pain sensitivity, aggressiveness, anxiety, sexual behavior, ovarian, oviductal, uterine and reproductive morphofunctional and molecular outcomes. These parameters were used to explore the reproductive capacity in female mice exposed to this synthetic testosterone ester. The animals were untreated or intraperitoneally treated with 5, 10 and 20 mg/kg TC every 48 h for 12 weeks. Our findings indicated that testosterone was upregulated while the hormones luteinizing, follicle-stimulating, estrogen and progesterone were down-regulated by TC. This AAS also exerted deleterious effects on anxiety, aggressivity, nociception, exploratory and sexual behavior in female mice. Concurrently, TC attenuated ovarian follicle maturation, interrupted the estrous cycle, induced oviductal and uterine hypotrophy. Estrous cyclicity was reestablished 60 days after AAS treatment. However, TC-treated mice still exhibited impaired reproductive capacity, a disturbance potentially related to deficiency in folliculogenesis, sex hormones production, and endometrial receptivity mediate by ER-α, PR, HOXA-10 and LIF down-regulation. Taken together, our findings indicated that in addition to female behavior, reproductive organs microstructure and function are markedly impaired by TC in a dose-dependent manner, whose time-dependent reversibility remains to be clarified.
Collapse
Affiliation(s)
- Mônica N Barcelos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Matheus A Souza
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Eliziária C Santos
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina 39100-000, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Programa de Pós-Graduação em Biologia Animal, Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Angel Mauricio Castro-Gamero
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências da Natureza, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biologia Animal, Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Burke FF, Hinks M, Salia S, Sparkes KM, Swift-Gallant A. Using Animal Models to Study the Interplay Between the Biodevelopmental Pathways Underlying Human Sexual Orientation. ARCHIVES OF SEXUAL BEHAVIOR 2023; 52:2979-2984. [PMID: 36477673 DOI: 10.1007/s10508-022-02499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Affiliation(s)
- Francine F Burke
- Department of Psychology, Memorial University of Newfoundland, 230 Elizabeth Ave., St John's, NL, A1B 3X9, Canada
| | - Meagan Hinks
- Department of Psychology, Memorial University of Newfoundland, 230 Elizabeth Ave., St John's, NL, A1B 3X9, Canada
| | - Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, 230 Elizabeth Ave., St John's, NL, A1B 3X9, Canada
| | - Kerri M Sparkes
- Department of Psychology, Memorial University of Newfoundland, 230 Elizabeth Ave., St John's, NL, A1B 3X9, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, 230 Elizabeth Ave., St John's, NL, A1B 3X9, Canada.
| |
Collapse
|
5
|
Salia S, Martin Y, Burke FF, Myles LA, Jackman L, Halievski K, Bambico FR, Swift-Gallant A. Antibiotic-induced socio-sexual behavioral deficits are reversed via cecal microbiota transplantation but not androgen treatment. Brain Behav Immun Health 2023; 30:100637. [PMID: 37256194 PMCID: PMC10225889 DOI: 10.1016/j.bbih.2023.100637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/03/2023] [Accepted: 05/07/2023] [Indexed: 06/01/2023] Open
Abstract
Recent evidence has demonstrated a sex-specific role of the gut microbiome on social behavior such as anxiety, possibly driven by a reciprocal relationship between the gut microbiome and gonadal hormones. For instance, gonadal hormones drive sex differences in gut microbiota composition, and certain gut bacteria can produce androgens from glucocorticoids. We thus asked whether the gut microbiome can influence androgen-dependent socio-sexual behaviors. We first treated C57BL/6 mice with broad-spectrum antibiotics (ABX) in drinking water to deplete the gut microbiota either transiently during early development (embryonic day 16-postnatal day [PND] 21) or in adulthood (PND 60-85). We hypothesized that if ABX interferes with androgens, then early ABX would interfere with critical periods for sexual differentiation of brain and thus lead to long-term decreases in males' socio-sexual behavior, while adult ABX would interfere with androgens' activational effects on behavior. We found that in males but not females, early and adult ABX treatment decreased territorial aggression, and adult ABX also decreased sexual odor preference. We then assessed whether testosterone and/or cecal microbiota transplantation (CMT) via oral gavage could prevent ABX-induced socio-sexual behavioral deficits in adult ABX-treated males. Mice were treated with same- or other-sex control cecum contents or with testosterone for two weeks. While testosterone was not effective in rescuing any behavior, we found that male CMT restored both olfactory preference and aggression in adult ABX male mice, while female CMT restored olfactory preference but not aggression. These results suggest sex-specific effects of the gut microbiome on socio-sexual behaviors, independent of androgens.
Collapse
|
6
|
Aspesi D, Bass N, Kavaliers M, Choleris E. The role of androgens and estrogens in social interactions and social cognition. Neuroscience 2023:S0306-4522(23)00151-3. [PMID: 37080448 DOI: 10.1016/j.neuroscience.2023.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 03/02/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Gonadal hormones are becoming increasingly recognized for their effects on cognition. Estrogens, in particular, have received attention for their effects on learning and memory that rely upon the functioning of various brain regions. However, the impacts of androgens on cognition are relatively under investigated. Testosterone, as well as estrogens, have been shown to play a role in the modulation of different aspects of social cognition. This review explores the impact of testosterone and other androgens on various facets of social cognition including social recognition, social learning, social approach/avoidance, and aggression. We highlight the relevance of considering not only the actions of the most commonly studied steroids (i.e., testosterone, 17β-estradiol, and dihydrotestosterone), but also that of their metabolites and precursors, which interact with a plethora of different receptors and signalling molecules, ultimately modulating behaviour. We point out that it is also essential to investigate the effects of androgens, their precursors and metabolites in females, as prior studies have mostly focused on males. Overall, a comprehensive analysis of the impact of steroids such as androgens on behaviour is fundamental for a full understanding of the neural mechanisms underlying social cognition, including that of humans.
Collapse
Affiliation(s)
- Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph
| | - Noah Bass
- Department of Psychology and Neuroscience Program, University of Guelph
| | - Martin Kavaliers
- Department of Psychology and Neuroscience Program, University of Guelph; Department of Psychology, University of Western Ontario, London, Canada; Graduate Program in Neuroscience, University of Western Ontario, London, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph.
| |
Collapse
|
7
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
8
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
9
|
Neural and Hormonal Basis of Opposite-Sex Preference by Chemosensory Signals. Int J Mol Sci 2021; 22:ijms22158311. [PMID: 34361077 PMCID: PMC8347621 DOI: 10.3390/ijms22158311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
In mammalian reproduction, sexually active males seek female conspecifics, while estrous females try to approach males. This sex-specific response tendency is called sexual preference. In small rodents, sexual preference cues are mainly chemosensory signals, including pheromones. In this article, we review the physiological mechanisms involved in sexual preference for opposite-sex chemosensory signals in well-studied laboratory rodents, mice, rats, and hamsters of both sexes, especially an overview of peripheral sensory receptors, and hormonal and central regulation. In the hormonal regulation section, we discuss potential rodent brain bisexuality, as it includes neural substrates controlling both masculine and feminine sexual preferences, i.e., masculine preference for female odors and the opposite. In the central regulation section, we show the substantial circuit regulating sexual preference and also the influence of sexual experience that innate attractants activate in the brain reward system to establish the learned attractant. Finally, we review the regulation of sexual preference by neuropeptides, oxytocin, vasopressin, and kisspeptin. Through this review, we clarified the contradictions and deficiencies in our current knowledge on the neuroendocrine regulation of sexual preference and sought to present problems requiring further study.
Collapse
|
10
|
McKenna BG, Huang Y, Vervier K, Hofammann D, Cafferata M, Al-Momani S, Lowenthal F, Zhang A, Koh JY, Thenuwara S, Brueggeman L, Bahl E, Koomar T, Pottschmidt N, Kalmus T, Casten L, Thomas TR, Michaelson JJ. Genetic and morphological estimates of androgen exposure predict social deficits in multiple neurodevelopmental disorder cohorts. Mol Autism 2021; 12:43. [PMID: 34108004 PMCID: PMC8190870 DOI: 10.1186/s13229-021-00450-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 06/01/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) such as autism spectrum disorder (ASD) display a strong male bias. Androgen exposure is profoundly increased in typical male development, but it also varies within the sexes, and previous work has sought to connect morphological proxies of androgen exposure, including digit ratio and facial morphology, to neurodevelopmental outcomes. The results of these studies have been mixed, and the relationships between androgen exposure and behavior remain unclear. METHODS Here, we measured both digit ratio masculinity (DRM) and facial landmark masculinity (FLM) in the same neurodevelopmental cohort (N = 763) and compared these proxies of androgen exposure to clinical and parent-reported features as well as polygenic risk scores. RESULTS We found that FLM was significantly associated with NDD diagnosis (ASD, ADHD, ID; all [Formula: see text]), while DRM was not. When testing for association with parent-reported problems, we found that both FLM and DRM were positively associated with concerns about social behavior ([Formula: see text], [Formula: see text]; [Formula: see text], [Formula: see text], respectively). Furthermore, we found evidence via polygenic risk scores (PRS) that DRM indexes masculinity via testosterone levels ([Formula: see text], [Formula: see text]), while FLM indexes masculinity through a negative relationship with sex hormone binding globulin (SHBG) levels ([Formula: see text], [Formula: see text]). Finally, using the SPARK cohort (N = 9419) we replicated the observed relationship between polygenic estimates of testosterone, SHBG, and social functioning ([Formula: see text], [Formula: see text], and [Formula: see text], [Formula: see text] for testosterone and SHBG, respectively). Remarkably, when considered over the extremes of each variable, these quantitative sex effects on social functioning were comparable to the effect of binary sex itself (binary male: [Formula: see text]; testosterone: [Formula: see text] from 0.1%-ile to 99.9%-ile; SHBG: [Formula: see text] from 0.1%-ile to 99.9%-ile). LIMITATIONS In the devGenes and SPARK cohorts, our analyses rely on indirect, rather than direct measurement of androgens and related molecules. CONCLUSIONS These findings and their replication in the large SPARK cohort lend support to the hypothesis that increasing net androgen exposure diminishes capacity for social functioning in both males and females.
Collapse
Affiliation(s)
| | - Yongchao Huang
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Kévin Vervier
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | | | - Mary Cafferata
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Seima Al-Momani
- Department of Psychology, University of Nebraska, Omaha, USA
| | | | - Angela Zhang
- University of Washington School of Public Health, Seattle, USA
| | - Jin-Young Koh
- Molecular Otolaryngology and Renal Research Laboratories, University of Iowa, Iowa City, USA
| | | | - Leo Brueggeman
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Ethan Bahl
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Tanner Koomar
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | | | - Taylor Kalmus
- Department of Psychology, University of Washington, Seattle, USA
| | - Lucas Casten
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Taylor R Thomas
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | | |
Collapse
|
11
|
Cross SKJ, Martin YH, Salia S, Gamba I, Major CA, Hassan S, Parsons KA, Swift-Gallant A. Puberty is a Critical Period for Vomeronasal Organ Mediation of Socio-sexual Behavior in Mice. Front Behav Neurosci 2021; 14:606788. [PMID: 33551763 PMCID: PMC7862124 DOI: 10.3389/fnbeh.2020.606788] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
Genetic disruption of the vomeronasal organ (VNO), an organ responsible for pheromone processing, drastically alters socio-sexual behavior in mice. However, it is not known whether the VNO has a role during the pubertal organizational period when sex-typical socio-sexual behaviors emerge, or if disruption of the organ in adulthood is sufficient to alter socio-sexual behavior. To bypass the lifelong VNO disruption of genetic knockout models, we surgically ablated the VNO of male and female mice either during the peripubertal period [postnatal day (PND) 28–30] or adulthood (PND 58–60), with sham controls at both ages. We ruled out anosmia via the buried food test and assessed sexual odor preferences by simultaneously exposing mice to same- and opposite-sex soiled-bedding. We then measured territorial aggression with the resident-intruder paradigm and assessed sexual behavior in response to an encounter with an estrus-induced female. Neural activity approximated by FOS-immunoreactivity along the VNO-accessory olfactory pathway was measured in response to opposite-sex odors. We found that peripubertal VNO ablation decreased sexual odor preferences and neural activity in response to opposite-sex odors, and drastically reduced territorial aggression in male mice. Conversely, adult VNO ablation resulted in subtle differences in sexual odor preferences compared with sham controls. Regardless of the VNO condition, mice displayed sex-typical copulatory behaviors. Together, these results suggest that puberty is a critical period in development whereby the VNO contributes to the sexual differentiation of behavior and neural response to conspecific odors.
Collapse
Affiliation(s)
- Sarah K J Cross
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Yellow H Martin
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Iain Gamba
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Christina A Major
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Suhail Hassan
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Katelyn A Parsons
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
12
|
Swift-Gallant A, Coome LA, Ramzan F, Monks DA. Non-androgenic testicular mediation of androphilia in male mice with global overexpression of androgen receptors. Behav Brain Res 2020; 391:112694. [PMID: 32428632 DOI: 10.1016/j.bbr.2020.112694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Sexual attraction is robustly sexually differentiated among mammalian species. Gonadal androgens acting perinatally and in adulthood are required for male-typical preference for female sexual cues. Recent evidence suggests that at the high extent of AR signaling, male mice show an increased preference for same-sex odor cues. These findings were found only in mice that overexpress AR globally in all tissues (CMV-AR), whereas neural AR overexpression (Nestin-AR) did not affect sexual preference. The present studies investigated the endocrine basis of this phenotype and examined whether preference for male or female stimulus animals (partner preference) was also affected in these transgenic animals. We manipulated adult gonadal hormones in male mice that overexpress AR globally and males that overexpress AR only in neural tissue. We replicate the finding that androphilia is increased in gonadally intact CMV-AR males, and these males exhibited reduced neural activation in response to estrus female odors. Testosterone treatment of gonadectomized CMV-AR males was sufficient to induce a gynephilic olfactory preference, while a gynephilic partner preference was induced with gonadectomy alone. These findings suggest that altered sexual preference of CMV-AR male mice is mediated by inhibitory activational functions of the testes. Together, these results suggest that at the high extent of AR signaling, non-neural AR via the gonads, can promote androphilia.
Collapse
Affiliation(s)
- A Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - L A Coome
- Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - F Ramzan
- Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - D A Monks
- Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Cells and Systems Biology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| |
Collapse
|
13
|
Balthazart J. Sexual partner preference in animals and humans. Neurosci Biobehav Rev 2020; 115:34-47. [PMID: 32450091 PMCID: PMC7484171 DOI: 10.1016/j.neubiorev.2020.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/11/2020] [Accepted: 03/24/2020] [Indexed: 12/25/2022]
Abstract
Sex differences in brain and behavior of animals including humans result from an interaction between biological and environmental influences. This is also true for the differences between men and women concerning sexual orientation. Sexual differentiation is mediated by three groups of biological mechanisms: early actions of sex steroids, more direct actions of sex-specific genes not mediated by gonadal sex steroids and epigenetic mechanisms. Differential interactions with parents and conspecifics have additionally long-term influences on behavior. This presentation reviews available evidence indicating that these different mechanisms play a significant role in the control of sexual partner preference in animals and humans, in other words the homosexual versus heterosexual orientation. Clinical and epidemiological studies of phenotypically selected populations indicate that early actions of hormones and genetic factors clearly contribute to the determination of sexual orientation. The maternal embryonic environment also modifies the incidence of male homosexuality via immunological mechanisms. The relative contribution of each of these mechanisms remains however to be determined.
Collapse
|
14
|
Lee SLJ, Horsfield JA, Black MA, Rutherford K, Gemmell NJ. Identification of sex differences in zebrafish (Danio rerio) brains during early sexual differentiation and masculinization using 17α-methyltestoterone. Biol Reprod 2019; 99:446-460. [PMID: 29272338 DOI: 10.1093/biolre/iox175] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/18/2017] [Indexed: 12/26/2022] Open
Abstract
Sexual behavior in teleost fish is highly plastic. It can be attributed to the relatively few sex differences found in adult brain transcriptomes. Environmental and hormonal factors can influence sex-specific behavior. Androgen treatment stimulates behavioral masculinization. Sex dimorphic gene expression in developing teleost brains and the molecular basis for androgen-induced behavioral masculinization are poorly understood. In this study, juvenile zebrafish (Danio rerio) were treated with 100 ng/L of 17 alpha-methyltestosterone (MT) during sexual development from 20 days post fertilization to 40 days and 60 days post fertilization. We compared brain gene expression patterns in MT-treated zebrafish with control males and females using RNA-Seq to shed light on the dynamic changes in brain gene expression during sexual development and how androgens affect brain gene expression leading to behavior masculinization. We found modest differences in gene expression between juvenile male and female zebrafish brains. Brain aromatase (cyp19a1b), prostaglandin 3a synthase (ptges3a), and prostaglandin reductase 1 (ptgr1) were among the genes with sexually dimorphic expression patterns. MT treatment significantly altered gene expression relative to both male and female brains. Fewer differences were found among MT-treated brains and male brains compared to female brains, particularly at 60 dpf. MT treatment upregulated the expression of hydroxysteroid 11-beta dehydrogenase 2 (hsd11b2), deiodinase, iodothyronine, type II (dio2), and gonadotrophin releasing hormones (GnRH) 2 and 3 (gnrh2 and gnrh3) suggesting local synthesis of 11-ketotestosterone, triiodothyronine, and GnRHs in zebrafish brains which are influenced by androgens. Androgen, estrogen, prostaglandin, thyroid hormone, and GnRH signaling pathways likely interact to modulate teleost sexual behavior.
Collapse
Affiliation(s)
- Stephanie L J Lee
- Department of Anatomy, University of Otago, Dunedin, Otago, New Zealand
| | - Julia A Horsfield
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, Otago, New Zealand
| | - Michael A Black
- Department of Biochemistry, University of Otago, Dunedin, Otago, New Zealand
| | - Kim Rutherford
- Department of Anatomy, University of Otago, Dunedin, Otago, New Zealand
| | - Neil J Gemmell
- Department of Anatomy, University of Otago, Dunedin, Otago, New Zealand
| |
Collapse
|
15
|
Ramzan F, Phung T, Swift-Gallant A, Coome LA, Holmes MM, Monks DA. Both neural and global androgen receptor overexpression affect sexual dimorphism in the mouse brain. J Neuroendocrinol 2019; 31:e12715. [PMID: 30920021 DOI: 10.1111/jne.12715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/14/2019] [Accepted: 03/22/2019] [Indexed: 01/28/2023]
Abstract
Testosterone is the main endocrine mechanism mediating sexual differentiation of the mammalian brain, although testosterone signalling is complex and important mechanistic questions remain. Notably, the extent to which testosterone acts via androgen receptors (AR) in this process remains unknown and it is also not clear where testosterone acts in the body to produce sexual dimorphisms in neuroanatomy. To address these questions, we used a transgenic mouse model of Cre/loxP-driven AR overexpression in which AR was induced selectively in neural tissue (Nestin-cre) or in all tissues (CMV-cre). We then studied sexually dimorphic features of several well-characterised sexual dimorphisms: calbindin-immunoreactive neurones in the medial preoptic area (CALB-SDN), tyrosine hydroxylase neurones in the anteroventral periventricular nucleus, and vasopressin-immunoreactive neurones originating in the bed nucleus of the stria terminalis and their projections in the lateral septum. We additionally evaluated oestrogen receptor α immunoreactivity in these nuclei. Briefly, we found that global but not neural overexpression of AR resulted in masculinisation of CALB-SDN nucleus volume, cell number and cell size in transgenic females. Furthermore, neural AR overexpression resulted in increased oestrogen receptor α staining in females compared to males in the medial preoptic area. AR overexpression did not affect other measures. Overall, the results of the present study provide support for the hypothesis that androgenic mechanisms external to the nervous system can affect sexual differentiation of the brain.
Collapse
Affiliation(s)
- Firyal Ramzan
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Thanh Phung
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Lindsay A Coome
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - D Ashley Monks
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Arias-Reyes C, Losantos-Ramos K, Gonzales M, Furrer D, Soliz J. NADH-linked mitochondrial respiration in the developing mouse brain is sex-, age- and tissue-dependent. Respir Physiol Neurobiol 2019; 266:156-162. [PMID: 31128272 DOI: 10.1016/j.resp.2019.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/09/2019] [Accepted: 05/20/2019] [Indexed: 11/17/2022]
Abstract
Mitochondria play a major role in the brain. Apart from energy production, mitochondria regulate key factors in the activation of cell signaling pathways such as survival, proliferation, and differentiation. While all these processes occur during the physiological development of the brain, it is surprising that the mitochondrial functions and functioning in the brain during the postnatal development remain poorly explored. In this work, we collected samples of brainstem and cortex of mice at postnatal ages 3 (P3), 21 (P21), and at adulthood (3 months old) and evaluated the mitochondrial oxygen consumption after complex I activation. To do so, we used our oxygraph-2 K system (OROBOROS) that measures the mitochondrial bioenergetics in saponin-permeabilized tissue punches of 2 mg weight. Furthermore, as sex dimorphism in the brain occurs since very early stages of development, we performed experiments in brain samples of male and female mice. Accordingly, the mitochondrial oxygen consumption rate (OCR) was evaluated under activation of complex I (NADH-linked respiration - mitochondrial state 3), and during the inhibition of the complex V (ATP synthase) with oligomycin (mitochondrial state 4). In following, the respiratory control ratio (RCR - state 3/state4) was calculated as an index of mitochondrial oxidative-phosphorylation coupling. Our results show that the activity of the mitochondrial complex I in the brain increases along with the postnatal development in a sex- and tissue-dependent manner, with males showing higher activity than females, and with brainstem tissue showing higher activity than cortex. Our data may contribute to a better understanding of the sex-dependent maturation of the cortex and the cardiorespiratory network located in the brainstem.
Collapse
Affiliation(s)
- C Arias-Reyes
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada; Instituto de Biología Molecular y Biotecnología, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - K Losantos-Ramos
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - M Gonzales
- Instituto Boliviano de Biología de la Altura, Facultad de Medicina, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - D Furrer
- Oncology Axis, CHU of Quebec Research Center, Laval University, Quebec City, Canada
| | - J Soliz
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada; Instituto de Biología Molecular y Biotecnología, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andrés, La Paz, Bolivia.
| |
Collapse
|
17
|
Zhang YT, Hong WS, Qiu HT, Wang Q, Chen SX. Androgen induces olfactory expression of prostaglandin E 2 receptor Ep1 in the burrow-living fish Bostrychus sinensis. J Steroid Biochem Mol Biol 2019; 188:156-165. [PMID: 30659898 DOI: 10.1016/j.jsbmb.2019.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/03/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
Abstract
It is well documented that androgens modify olfactory processing in vertebrates. In fish, several lines of evidence indicate that androgens increase olfactory sensitivity to prostaglandin pheromone, but the molecular mechanism is still unclear. Our previous studies showed that prostaglandin E2 (PGE2) is a sex pheromone in the burrowing-living fish Chinese black sleeper (Bostrychus sinensis) and that the PGE2 receptor 1 (Ep1) in the olfactory rosette is a candidate receptor for sensing sex pheromone PGE2. In the present study, we found that testosterone (T) and 11-ketotestosterone (11-KT) exhibited stimulatory effects on the expression of ep1 in the olfactory rosette in vivo and ex vivo. Moreover, the androgen receptor (Ar) agonist R1881 had similar effects to 11-KT on the expression of ep1 ex vivo, suggesting the up-regulatory effect is mediated by Ar. The amount of arα transcripts (˜1500 copies/100 ng total RNA) was greater than that of arβ (˜300 copies/100 ng total RNA) in the olfactory rosette, and the expression levels of arα increased with spermatogenesis and peaked at late meiosis stage. Moreover, activated Arα but not Arβ transactivated a 2k bp ep1 promoter in HEK293T cell, and some OSNs exhibited co-localization of arα mRNA and Ep1 protein signals. Taken together, our results suggest that Arα, but not Arβ, plays a crucial role in mediating the androgen-induced up-regulation of ep1 expression in B. sinensis. The present study is the first to shed light on the molecular mechanisms whereby androgens enhance responsiveness to prostaglandin sex pheromones in teleosts.
Collapse
Affiliation(s)
- Yu Ting Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, 361102, PR China
| | - Wan Shu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, 361102, PR China
| | - Heng Tong Qiu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, 361102, PR China
| | - Qiong Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, 361102, PR China
| | - Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, 361102, PR China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Fujian, 361102, PR China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Fujian, 361102, PR China.
| |
Collapse
|
18
|
Tsuneoka Y. Molecular neuroanatomy of the mouse medial preoptic area with reference to parental behavior. Anat Sci Int 2018; 94:39-52. [DOI: 10.1007/s12565-018-0468-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 10/28/2018] [Indexed: 11/28/2022]
|
19
|
Cisternas CD, Garcia-Segura LM, Cambiasso MJ. Hormonal and genetic factors interact to control aromatase expression in the developing brain. J Neuroendocrinol 2018; 30. [PMID: 28891264 DOI: 10.1111/jne.12535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 02/03/2023]
Abstract
Brain expression of the enzyme P450-aromatase has been studied extensively. Subsequent to the aromatisation hypothesis having established brain aromatase as a key factor to convert gonadal testosterone to oestradiol, several studies have investigated the regulation of aromatase during the critical period of brain sexual differentiation. We review previous and recent findings concerning regulation of aromatase. The role of gonadal hormones, sex chromosome genes and neurosteroids is analysed in terms of their contribution to aromatase expression, as well as implications for the organisational effect of steroids during development.
Collapse
Affiliation(s)
- C D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - L M Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
20
|
Monks DA, Swift-Gallant A. Non-neural androgen receptors affect sexual differentiation of brain and behaviour. J Neuroendocrinol 2018; 30. [PMID: 28590577 DOI: 10.1111/jne.12493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/02/2017] [Accepted: 06/03/2017] [Indexed: 01/08/2023]
Abstract
Although gonadal testosterone is the principal endocrine factor that promotes masculine traits in mammals, the development of a male phenotype requires local production of both androgenic and oestrogenic signals within target tissues. Much of our knowledge concerning androgenic components of testosterone signalling in sexual differentiation comes from studies of androgen receptor (Ar) loss of function mutants. Here, we review these studies of loss of Ar function and of AR overexpression either globally or selectively in the nervous system of mice. Global and neural mutations affect socio-sexual behaviour and the neuroanatomy of these mice in a sexually differentiated manner. Some masculine traits are affected by both global and neural mutation, indicative of neural mediation, whereas other masculine traits are affected only by global mutation, indicative of an obligatory non-neural androgen target. These results support a model in which multiple sites of androgen action coordinate to produce masculine phenotypes. Furthermore, AR overexpression does not always have a phenotype opposite to that of loss of Ar function mutants, indicative of a nonlinear relationship between androgen dose and masculine phenotype in some cases. Potential mechanisms of Ar gene function in non-neural targets in producing masculine phenotypes are discussed.
Collapse
Affiliation(s)
- D A Monks
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cells and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - A Swift-Gallant
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| |
Collapse
|
21
|
Ujjainwala AL, Courtney CD, Rhoads SG, Rhodes JS, Christian CA. Genetic loss of diazepam binding inhibitor in mice impairs social interest. GENES BRAIN AND BEHAVIOR 2017; 17:e12442. [PMID: 29193847 DOI: 10.1111/gbb.12442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 01/21/2023]
Abstract
Neuropsychiatric disorders in which reduced social interest is a common symptom, such as autism, depression, and anxiety, are frequently associated with genetic mutations affecting γ-aminobutyric acid (GABA)ergic transmission. Benzodiazepine treatment, acting via GABA type-A receptors, improves social interaction in male mouse models with autism-like features. The protein diazepam binding inhibitor (DBI) can act as an endogenous benzodiazepine, but a role for DBI in social behavior has not been described. Here, we investigated the role of DBI in the social interest and recognition behavior of mice. The responses of DBI wild-type and knockout male and female mice to ovariectomized female wild-type mice (a neutral social stimulus) were evaluated in a habituation/dishabituation task. Both male and female knockout mice exhibited reduced social interest, and DBI knockout mice lacked the sex difference in social interest levels observed in wild-type mice, in which males showed higher social interest levels than females. The ability to discriminate between familiar and novel stimulus mice (social recognition) was not impaired in DBI-deficient mice of either sex. DBI knockouts could learn a rotarod motor task, and could discriminate between social and nonsocial odors. Both sexes of DBI knockout mice showed increased repetitive grooming behavior, but not in a manner that would account for the decrease in social investigation time. Genetic loss of DBI did not alter seminal vesicle weight, indicating that the social interest phenotype of males lacking DBI is not due to reduced circulating testosterone. Together, these studies show a novel role of DBI in driving social interest and motivation.
Collapse
Affiliation(s)
- A L Ujjainwala
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - C D Courtney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - S G Rhoads
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - J S Rhodes
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - C A Christian
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
22
|
Balthazart J, Court L. Human Sexual Orientation: The Importance of Evidentiary Convergence. ARCHIVES OF SEXUAL BEHAVIOR 2017; 46:1595-1600. [PMID: 28500563 PMCID: PMC5532062 DOI: 10.1007/s10508-017-0997-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/02/2017] [Indexed: 05/26/2023]
Affiliation(s)
- Jacques Balthazart
- GIGA Neurosciences, University of Liège, 15 Avenue Hippocrate, 4000, Liège, Belgium.
| | - Lucas Court
- GIGA Neurosciences, University of Liège, 15 Avenue Hippocrate, 4000, Liège, Belgium
| |
Collapse
|
23
|
Tsuneoka Y, Tsukahara S, Yoshida S, Takase K, Oda S, Kuroda M, Funato H. Moxd1 Is a Marker for Sexual Dimorphism in the Medial Preoptic Area, Bed Nucleus of the Stria Terminalis and Medial Amygdala. Front Neuroanat 2017; 11:26. [PMID: 28396628 PMCID: PMC5366752 DOI: 10.3389/fnana.2017.00026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 03/14/2017] [Indexed: 12/26/2022] Open
Abstract
The brain shows various sex differences in its structures. Various mammalian species exhibit sex differences in the sexually dimorphic nucleus of the preoptic area (SDN-POA) and parts of the extended amygdala such as the principal nucleus of the bed nucleus of the stria terminalis (BNSTpr) and posterodorsal part of the medial amygdala (MePD). The SDN-POA and BNSTpr are male-biased sexually dimorphic nuclei, and characterized by the expression of calbindin D-28K (calbindin 1). However, calbindin-immunoreactive cells are not restricted to the SDN-POA, but widely distributed outside of the SDN-POA. To find genes that are more specific to sexually dimorphic nuclei, we selected candidate genes by searching the Allen brain atlas and examined the detailed expressions of the candidate genes using in situ hybridization. We found that the strong expression of monooxygenase DBH-like 1 (Moxd1) was restricted to the SDN-POA, BNSTpr and MePD. The numbers of Moxd1-positive cells in the SDN-POA, BNSTpr and MePD in male mice were larger than those in female mice. Most of the Moxd1-positive cells in the SDN-POA and BNSTpr expressed calbindin. Neonatal castration of male mice reduced the number of Moxd1-positive cells in the SDN-POA, whereas gonadectomy in adulthood did not change the expression of the Moxd1 gene in the SDN-POA in both sexes. These results suggest that the Moxd1 gene is a suitable marker for sexual dimorphic nuclei in the POA, BNST and amygdala, which enables us to manipulate sexually dimorphic neurons to examine their roles in sex-biased physiology and behaviors.
Collapse
Affiliation(s)
- Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University Tokyo, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Sachine Yoshida
- Department of Anatomy, Faculty of Medicine, Toho UniversityTokyo, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology AgencySaitama, Japan
| | - Kenkichi Takase
- Department of Anatomy, Faculty of Medicine, Toho UniversityTokyo, Japan; Laboratory of Psychology, Jichi Medical UniversityTochigi, Japan
| | - Satoko Oda
- Department of Anatomy, Faculty of Medicine, Toho University Tokyo, Japan
| | - Masaru Kuroda
- Department of Anatomy, Faculty of Medicine, Toho University Tokyo, Japan
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho UniversityTokyo, Japan; International Institutes for Integrative Sleep Medicine (WPI-IIIS), University of TsukubaIbaraki, Japan
| |
Collapse
|
24
|
Opposite-sex attraction in male mice requires testosterone-dependent regulation of adult olfactory bulb neurogenesis. Sci Rep 2016; 6:36063. [PMID: 27782186 PMCID: PMC5080553 DOI: 10.1038/srep36063] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 10/11/2016] [Indexed: 11/23/2022] Open
Abstract
Opposite-sex attraction in most mammals depends on the fine-tuned integration of pheromonal stimuli with gonadal hormones in the brain circuits underlying sexual behaviour. Neural activity in these circuits is regulated by sensory processing in the accessory olfactory bulb (AOB), the first central station of the vomeronasal system. Recent evidence indicates adult neurogenesis in the AOB is involved in sex behaviour; however, the mechanisms underlying this function are unknown. By using Semaphorin 7A knockout (Sema7A ko) mice, which show a reduced number of gonadotropin-releasing-hormone neurons, small testicles and subfertility, and wild-type males castrated during adulthood, we demonstrate that the level of circulating testosterone regulates the sex-specific control of AOB neurogenesis and the vomeronasal system activation, which influences opposite-sex cue preference/attraction in mice. Overall, these data highlight adult neurogenesis as a hub for the integration of pheromonal and hormonal cues that control sex-specific responses in brain circuits.
Collapse
|
25
|
Swift-Gallant A, Coome L, Srinivasan S, Monks DA. Non-neural androgen receptor promotes androphilic odor preference in mice. Horm Behav 2016; 83:14-22. [PMID: 27191855 DOI: 10.1016/j.yhbeh.2016.05.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/30/2016] [Accepted: 05/13/2016] [Indexed: 11/21/2022]
Abstract
In mice, male-typical preference for female olfactory cues results largely from sexually differentiated testosterone production. It is currently unclear on which cells and tissues testosterone acts to produce male-typical preference for female olfactory cues. To further address the site of androgen action on olfactory preference, we have developed a loxP-based transgenic mouse that overexpresses androgen receptors (AR) only when activated by Cre. We used this transgene to overexpress AR globally in all tissues using a CMV-Cre driver and a Nestin-Cre driver to overexpress AR selectively in neural tissue. We then examined olfactory preference in transgenic and wildtype (Wt) littermates by simultaneously exposing animals to female-soiled, male-soiled and clean bedding. Ubiquitous overexpression of AR in CMV-AR mice increased preference for male bedding, whereas neural-specific AR overexpression in Nestin-AR transgenic mice did not differ from wildtype siblings in olfactory preference. Neural activation of olfactory brain areas in response to female-soiled bedding was also evaluated in these mice by measuring FOS immunoreactivity. This revealed a decrease in neural activity along the accessory olfactory pathway that accompanied the decrease in preference for female odors in CMV-AR males, compared to both Nestin-AR and Wt male siblings. Together, results indicate that androgens act via non-neural AR to mediate olfactory preference and neural responses to olfactory stimuli, and further suggest that AR in non-neural tissues can promote androphilic odor preferences in male mice.In mice, male-typical preference for female olfactory cues results largely from sexually differentiated testosterone production. It is currently unclear on which cells and tissues testosterone acts to produce male-typical preference for female olfactory cues. To further address the site of androgen action on olfactory preference, we have developed a loxP-based transgenic mouse that overexpresses androgen receptors (AR) only when activated by Cre. We used this transgene to overexpress AR globally in all tissues using a CMV-Cre driver and a Nestin-Cre driver to overexpress AR selectively in neural tissue. We then examined olfactory preference in transgenic and wildtype (Wt) littermates by simultaneously exposing animals to female-soiled, male-soiled and clean bedding. Ubiquitous overexpression of AR in CMV-AR mice increased preference for male bedding, whereas neural-specific AR overexpression in Nestin-AR transgenic mice did not differ from wildtype siblings in olfactory preference. Neural activation of olfactory brain areas in response to female-soiled bedding was also evaluated in these mice by measuring FOS immunoreactivity. This revealed a decrease in neural activity along the accessory olfactory pathway that accompanied the decrease in preference for female odors in CMV-AR males, compared to both Nestin-AR and Wt male siblings. Together, results indicate that androgens act via non-neural AR to mediate olfactory preference and neural responses to olfactory stimuli, and further suggest that AR in non-neural tissues can promote androphilic odor preferences in male mice.
Collapse
Affiliation(s)
- A Swift-Gallant
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada
| | - L Coome
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada; Department of Neuroscience, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada
| | - S Srinivasan
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada
| | - D A Monks
- Department of Psychology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada; Department of Neuroscience, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada; Department of Cells and Systems Biology, University of Toronto, 100 St. George Street, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
26
|
Harris EP, Abel JM, Tejada LD, Rissman EF. Calbindin Knockout Alters Sex-Specific Regulation of Behavior and Gene Expression in Amygdala and Prefrontal Cortex. Endocrinology 2016; 157:1967-79. [PMID: 27010449 PMCID: PMC4870870 DOI: 10.1210/en.2016-1055] [Citation(s) in RCA: 15] [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: 02/06/2023]
Abstract
Calbindin-D(28K) (Calb1), a high-affinity calcium buffer/sensor, shows abundant expression in neurons and has been associated with a number of neurobehavioral diseases, many of which are sexually dimorphic in incidence. Behavioral and physiological end points are affected by experimental manipulations of calbindin levels, including disruption of spatial learning, hippocampal long-term potentiation, and circadian rhythms. In this study, we investigated novel aspects of calbindin function on social behavior, anxiety-like behavior, and fear conditioning in adult mice of both sexes by comparing wild-type to littermate Calb1 KO mice. Because Calb1 mRNA and protein are sexually dimorphic in some areas of the brain, we hypothesized that sex differences in behavioral responses of these behaviors would be eliminated or revealed in Calb1 KO mice. We also examined gene expression in the amygdala and prefrontal cortex, two areas of the brain intimately connected with limbic system control of the behaviors tested, in response to sex and genotype. Our results demonstrate that fear memory and social behavior are altered in male knockout mice, and Calb1 KO mice of both sexes show less anxiety. Moreover, gene expression studies of the amygdala and prefrontal cortex revealed several significant genotype and sex effects in genes related to brain-derived neurotrophic factor signaling, hormone receptors, histone deacetylases, and γ-aminobutyric acid signaling. Our findings are the first to directly link calbindin with affective and social behaviors in rodents; moreover, the results suggest that sex differences in calbindin protein influence behavior.
Collapse
Affiliation(s)
- Erin P Harris
- Neuroscience Graduate Program (E.P.H., L.D.T.) and Department of Biochemistry and Molecular Genetics (J.M.A., E.F.R.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Jean M Abel
- Neuroscience Graduate Program (E.P.H., L.D.T.) and Department of Biochemistry and Molecular Genetics (J.M.A., E.F.R.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Lucia D Tejada
- Neuroscience Graduate Program (E.P.H., L.D.T.) and Department of Biochemistry and Molecular Genetics (J.M.A., E.F.R.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Emilie F Rissman
- Neuroscience Graduate Program (E.P.H., L.D.T.) and Department of Biochemistry and Molecular Genetics (J.M.A., E.F.R.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
27
|
Roselli CE, Meaker M, Stormshak F, Estill CT. Effects of Long-Term Flutamide Treatment During Development on Sexual Behaviour and Hormone Responsiveness in Rams. J Neuroendocrinol 2016; 28:10.1111/jne.12389. [PMID: 27005749 PMCID: PMC4882258 DOI: 10.1111/jne.12389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/14/2016] [Accepted: 03/19/2016] [Indexed: 01/06/2023]
Abstract
Testosterone exposure during midgestation differentiates neural circuits controlling sex-specific behaviours and patterns of gonadotrophin secretion in male sheep. Testosterone acts through androgen receptors (AR) and/or after aromatisation to oestradiol and binding to oestrogen receptors. The present study assessed the role of AR activation in male sexual differentiation. We compared rams that were exposed to the AR antagonist flutamide (Flu) throughout the critical period (i.e. days 30-90 of gestation) to control rams and ewes that received no prenatal treatments. The external genitalia of all Flu rams were phenotypically female. Testes were positioned s.c. in the inguinal region of the abdomen, exhibited seasonally impaired androgen secretion and were azospermic. Flu rams displayed male-typical precopulatory and mounting behaviours but could not intromit or ejaculate because they lacked a penis. Flu rams exhibited greater mounting behaviour than control rams and, similar to controls, showed sexual partner preferences for oestrous ewes. Neither control, nor Flu rams responded to oestradiol treatments with displays of female-typical receptive behaviour or LH surge responses, whereas all control ewes responded as expected. The ovine sexually dimorphic nucleus in Flu rams was intermediate in volume between control rams and ewes and significantly different from both. These results indicate that prenatal anti-androgen exposure is not able to block male sexual differentiation in sheep and suggest that compensatory mechanisms intervene to maintain sufficient androgen stimulation during development.
Collapse
Affiliation(s)
- Charles E. Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239-3098
| | - Mary Meaker
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
| | - Fred Stormshak
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331-4501
- College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331-4501
| |
Collapse
|
28
|
Balthazart J. Sex differences in partner preferences in humans and animals. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150118. [PMID: 26833838 PMCID: PMC4785903 DOI: 10.1098/rstb.2015.0118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2015] [Indexed: 11/12/2022] Open
Abstract
A large number of morphological, physiological and behavioural traits are differentially expressed by males and females in all vertebrates including humans. These sex differences, sometimes, reflect the different hormonal environment of the adults, but they often remain present after subjects of both sexes are placed in the same endocrine conditions following gonadectomy associated or not with hormonal replacement therapy. They are then the result of combined influences of organizational actions of sex steroids acting early during development, or genetic differences between the sexes, or epigenetic mechanisms differentially affecting males and females. Sexual partner preference is a sexually differentiated behavioural trait that is clearly controlled in animals by the same type of mechanisms. This is also probably true in humans, even if critical experiments that would be needed to obtain scientific proof of this assertion are often impossible for pragmatic or ethical reasons. Clinical, epidemiological and correlative studies provide, however, converging evidence strongly suggesting, if not demonstrating, that endocrine, genetic and epigenetic mechanisms acting during the pre- or perinatal life control human sexual orientation, i.e. homosexuality versus heterosexuality. Whether they interact with postnatal psychosexual influences remains, however, unclear at present.
Collapse
Affiliation(s)
- Jacques Balthazart
- GIGA Neurosciences, University of Liège, 15 avenue Hippocrate, 4000 Liège, Belgium
| |
Collapse
|
29
|
Derouiche L, Keller M, Duittoz AH, Pillon D. Developmental exposure to Ethinylestradiol affects transgenerationally sexual behavior and neuroendocrine networks in male mice. Sci Rep 2015; 5:17457. [PMID: 26640081 PMCID: PMC4671018 DOI: 10.1038/srep17457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
Reproductive behavior and physiology in adulthood are controlled by hypothalamic sexually dimorphic neuronal networks which are organized under hormonal control during development. These organizing effects may be disturbed by endocrine disrupting chemicals (EDCs). To determine whether developmental exposure to Ethinylestradiol (EE2) may alter reproductive parameters in adult male mice and their progeny, Swiss mice (F1 generation) were exposed from prenatal to peripubertal periods to EE2 (0.1–1 μg/kg/d). Sexual behavior and reproductive physiology were evaluated on F1 males and their F2, F3 and F4 progeny. EE2-exposed F1 males and their F2 to F4 progeny exhibited EE2 dose-dependent increased sexual behavior, with reduced latencies of first mount and intromission, and higher frequencies of intromissions with a receptive female. The EE2 1 μg/kg/d exposed animals and their progeny had more calbindin immunoreactive cells in the medial preoptic area, known to be involved in the control of male sexual behavior in rodents. Despite neuroanatomical modifications in the Gonadotropin-Releasing Hormone neuron population of F1 males exposed to both doses of EE2, no major deleterious effects on reproductive physiology were detected. Therefore EE2 exposure during development may induce a hypermasculinization of the brain, illustrating how widespread exposure of animals and humans to EDCs can impact health and behaviors.
Collapse
Affiliation(s)
- Lyes Derouiche
- PRC, UMR 7247 INRA/CNRS/Université François-Rabelais de Tours/IFCE, Nouzilly, France
| | - Matthieu Keller
- PRC, UMR 7247 INRA/CNRS/Université François-Rabelais de Tours/IFCE, Nouzilly, France
| | - Anne Hélène Duittoz
- PRC, UMR 7247 INRA/CNRS/Université François-Rabelais de Tours/IFCE, Nouzilly, France
| | - Delphine Pillon
- PRC, UMR 7247 INRA/CNRS/Université François-Rabelais de Tours/IFCE, Nouzilly, France
| |
Collapse
|
30
|
Cisternas CD, Tome K, Caeiro XE, Dadam FM, Garcia-Segura LM, Cambiasso MJ. Sex chromosome complement determines sex differences in aromatase expression and regulation in the stria terminalis and anterior amygdala of the developing mouse brain. Mol Cell Endocrinol 2015; 414:99-110. [PMID: 26231585 DOI: 10.1016/j.mce.2015.07.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 10/23/2022]
Abstract
Aromatase, which converts testosterone in estradiol, is involved in the generation of brain sex dimorphisms. Here we used the "four core genotypes" mouse model, in which the effect of gonadal sex and sex chromosome complement is dissociated, to determine if sex chromosomes influence the expression of brain aromatase. The brain of 16 days old XY mouse embryos showed higher aromatase expression in the stria terminalis and the anterior amygdaloid area than the brain of XX embryos, independent of gonadal sex. Furthermore, estradiol or dihydrotestosterone increased aromatase expression in cultures of anterior amygdala neurons derived from XX embryos, but not in those derived from XY embryos. This effect was also independent of gonadal sex. The expression of other steroidogenic molecules, estrogen receptor-α and androgen receptor was not influenced by sex chromosomes. In conclusion, sex chromosomes determine sex dimorphisms in aromatase expression and regulation in the developing mouse brain.
Collapse
Affiliation(s)
- Carla D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Karina Tome
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ximena E Caeiro
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia M Dadam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - María J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
31
|
Jahan MR, Kokubu K, Islam MN, Matsuo C, Yanai A, Wroblewski G, Fujinaga R, Shinoda K. Species differences in androgen receptor expression in the medial preoptic and anterior hypothalamic areas of adult male and female rodents. Neuroscience 2014; 284:943-961. [PMID: 25446364 DOI: 10.1016/j.neuroscience.2014.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 11/25/2022]
Abstract
The medial preoptic and anterior hypothalamic areas (MPO/AH) are important androgen targets regulating homeostasis, neuroendocrinology and circadian rhythm as well as instinctive and sociosexual behaviors. Although species differences between rats and mice have been pointed out in terms of morphology and physiology, detailed distributions of androgen receptor (AR) have never been compared between the two rodents. In the present study, AR distribution was examined immunohistochemically in serial sections of the MPO/AH and compared for adult rats and mice. Western blotting and immunohistochemistry clearly demonstrated that AR expression in the brain was stronger in mice than in rats and was stronger in males than in females. In addition, we found (1) an "obliquely elongated calbindin-ir cell island" in mice medial preoptic nucleus (MPN) expressed AR intensely, as well as the sexually dimorphic nucleus in the MPN (SDN-MPN) in rats, strongly supporting a "putative SDN-MPN" previously proposed in mice; (2) AR expression in the suprachiasmatic nucleus (SCN) was much more prominent in mice than in rats and differed in localization between the two species; (3) a mouse-specific AR-ir cell cluster was newly identified as the "tear drop nucleus (TDN)", with male-dominant sexual dimorphism; and (4) two rat-specific AR-ir cell clusters were also newly identified as the "rostral and caudal nebular islands", with male-dominant sexual dimorphism. The present results may provide basic morphological evidence underlying species differences in androgen-modified psychological, physiological and endocrinergic responses. Above all, the findings of the mouse-specific TDN and differing AR expression in the SCN might explain not only species difference in gonadal modification of circadian rhythm, but also distinct structural bases in the context of transduction of SCN oscillation. The current study could also serve as a caution that data on androgen-sensitive functions obtained from one species should not always be directly applied to others among rodents.
Collapse
Affiliation(s)
- M R Jahan
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - K Kokubu
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Md N Islam
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - C Matsuo
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - A Yanai
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - G Wroblewski
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - R Fujinaga
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - K Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| |
Collapse
|
32
|
The 2D:4D digit ratio and social behaviour in wild female chacma baboons (Papio ursinus) in relation to dominance, aggression, interest in infants, affiliation and heritability. Behav Ecol Sociobiol 2014. [DOI: 10.1007/s00265-014-1817-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
33
|
He Z, Ferguson SA, Cui L, Greenfield LJ, Paule MG. Development of the sexually dimorphic nucleus of the preoptic area and the influence of estrogen-like compounds. Neural Regen Res 2014; 8:2763-74. [PMID: 25206587 PMCID: PMC4145994 DOI: 10.3969/j.issn.1673-5374.2013.29.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 08/29/2013] [Indexed: 11/18/2022] Open
Abstract
One of the well-defined sexually dimorphic structures in the brain is the sexually dimorphic nucleus, a cluster of cells located in the preoptic area of the hypothalamus. The rodent sexually dimorphic nucleus of the preoptic area can be delineated histologically using conventional Nissl staining or immunohistochemically using calbindin D28K immunoreactivity. There is increasing use of the bindin D28K-delineated neural cluster to define the sexually dimorphic nucleus of the preoptic area in rodents. Several mechanisms are proposed to underlie the processes that contribute to the sexual dimorphism (size difference) of the sexually dimorphic nucleus of the preoptic area. Recent evidence indicates that stem cell activity, including proliferation and migration presumably from the 3rd ventricle stem cell niche, may play a critical role in the postnatal development of the sexually dimorphic nucleus of the preoptic area and its distinguishing sexually dimorphic feature: a signifi-cantly larger volume in males. Sex hormones and estrogen-like compounds can affect the size of the sexually dimorphic nucleus of the preoptic area. Despite considerable research, it remains un-clear whether estrogen-like compounds and/or sex hormones increase size of the sexually dimor-phic nucleus of the preoptic area via an increase in stem cell activity originating from the 3rd ventricle stem cell niche.
Collapse
Affiliation(s)
- Zhen He
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA ; Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72005, USA
| | - Sherry Ann Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA
| | - Li Cui
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72005, USA
| | - Lazar John Greenfield
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72005, USA
| | - Merle Gale Paule
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
34
|
Goodman WA, Garg RR, Reuter BK, Mattioli B, Rissman EF, Pizarro TT. Loss of estrogen-mediated immunoprotection underlies female gender bias in experimental Crohn's-like ileitis. Mucosal Immunol 2014; 7:1255-65. [PMID: 24621993 PMCID: PMC4139459 DOI: 10.1038/mi.2014.15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 02/01/2014] [Accepted: 02/10/2014] [Indexed: 02/04/2023]
Abstract
The incidence and severity of Crohn's disease (CD) are increased in female patients. Using SAMP1/YitFc (SAMP) mice, a spontaneous model of chronic intestinal inflammation that displays histologic and pathogenic similarities to human CD, we investigated the potential mechanism(s) contributing to sex differences observed in CD. Similar to gender differences observed in CD patients, SAMP female (SAMP-F) mice displayed an earlier onset and more severe ileitis compared with SAMP male (SAMP-M) mice. Furthermore, T-regulatory cells (Tregs) from gut-associated lymphoid tissue (GALT) of SAMP-F mice were reduced in frequency and impaired in their in vitro and in vivo suppressive functions compared with that of SAMP-M mice. Given the interaction between sex hormones and Treg function, we investigated the possible role of estrogen (E2) in SAMP ileitis. SAMP-M mice responded to exogenous E2 administration by expanding Treg frequency and reducing ileal inflammation, whereas SAMP-F mice were resistant. Conventional T cells and Tregs responded differentially to estrogen signaling, leading to distinct immunoprotective effects mediated by distinct estrogen receptor (ER) isoforms. These mechanisms were impaired in T cells from SAMP-F mice. Thus, hormone signaling influences the expansion and function of GALT Tregs in an ER-dependent manner and contributes to gender-based differences in experimental CD.
Collapse
Affiliation(s)
- Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rekha R. Garg
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian K. Reuter
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, Alberta, Canada T6G 2X8
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Emilie F. Rissman
- Department of Biochemistry & Molecular Genetics, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
35
|
Mueller SC, Grissom EM, Dohanich GP. Assessing gonadal hormone contributions to affective psychopathologies across humans and animal models. Psychoneuroendocrinology 2014; 46:114-28. [PMID: 24882164 DOI: 10.1016/j.psyneuen.2014.04.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/15/2014] [Accepted: 04/21/2014] [Indexed: 10/25/2022]
Abstract
Despite increasing acknowledgement of hormonal contributions to mood and anxiety disorders, the underlying mechanisms by which gonadal hormones influence psychopathology-related behaviours remain unknown. This review focuses on recent research that examines the influence of gonadal steroid hormones, including androgens, oestrogens, and progesterone, on mood and anxiety-related behaviours in human health and disease. To this aim, the literature was surveyed for studies that assess conditions with suspected underlying hormonal imbalances in otherwise healthy participants (e.g., premenstrual dysphoric disorder, postmenopausal depression) as well as conditions linked to congenital endocrine abnormalities (e.g., Turner Syndrome, Klinefelter Syndrome, polycystic ovary syndrome, congenital adrenal hyperplasia, familial male precocious puberty, androgen insensitivity syndrome). Furthermore, to better inform clinical work and to create a translational bridge, a second goal was to set human psychopathologies and animal models of these conditions side-by-side. In the second part of the review, based on consistencies revealed in the existing literature across conditions, a new model for the impact of gonadal hormones on anxious and depressed behavioural states is proposed. Finally, we conclude by proposing directions for future research, including the development of specific tasks suitable for cross-species comparisons to increase our knowledge of the role of gonadal hormones in mood and anxiety.
Collapse
Affiliation(s)
- S C Mueller
- Department of Experimental Clinical and Health Psychology, Ghent University, Ghent, Belgium.
| | - E M Grissom
- Department of Psychology, Tulane University, New Orleans, LA, USA
| | - G P Dohanich
- Department of Psychology, Tulane University, New Orleans, LA, USA; Program in Neuroscience, Tulane University, New Orleans, LA, USA
| |
Collapse
|
36
|
Holmes MM, Van Mil S, Bulkowski C, Goldman SL, Goldman BD, Forger NG. Androgen receptor distribution in the social decision-making network of eusocial naked mole-rats. Behav Brain Res 2013; 256:214-8. [DOI: 10.1016/j.bbr.2013.08.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 08/13/2013] [Accepted: 08/15/2013] [Indexed: 10/26/2022]
|
37
|
Petrulis A. Chemosignals and hormones in the neural control of mammalian sexual behavior. Front Neuroendocrinol 2013; 34:255-67. [PMID: 23911848 DOI: 10.1016/j.yfrne.2013.07.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 01/05/2023]
Abstract
Males and females of most mammalian species depend on chemosignals to find, attract and evaluate mates and, in most cases, these appetitive sexual behaviors are strongly modulated by activational and organizational effects of sex steroids. The neural circuit underlying chemosensory-mediated pre- and peri-copulatory behavior involves the medial amygdala (MA), the bed nucleus of the stria terminalis (BNST), medial preoptic area (MPOA) and ventromedial hypothalamus (VMH), each area being subdivided into interconnected chemoreceptive and hormone-sensitive zones. For males, MA-BNST connections mediate chemoinvestigation whereas the MA-MPOA pathway regulates copulatory initiation. For females, MA-MPOA/BNST connections also control aspects of precopulatory behavior whereas MA-VMH projections control both precopulatory and copulatory behavior. Significant gaps in understanding remain, including the role of VMH in male behavior and MPOA in female appetitive behavior, the function of cortical amygdala, the underlying chemical architecture of this circuit and sex differences in hormonal and neurochemical regulation of precopulatory behavior.
Collapse
Affiliation(s)
- Aras Petrulis
- Georgia State University, Neuroscience Institute, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
38
|
Petrulis A. Chemosignals, hormones and mammalian reproduction. Horm Behav 2013; 63:723-41. [PMID: 23545474 PMCID: PMC3667964 DOI: 10.1016/j.yhbeh.2013.03.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 11/21/2022]
Abstract
Many mammalian species use chemosignals to coordinate reproduction by altering the physiology and behavior of both sexes. Chemosignals prime reproductive physiology so that individuals become sexually mature and active at times when mating is most probable and suppress it when it is not. Once in reproductive condition, odors produced and deposited by both males and females are used to find and select individuals for mating. The production, dissemination and appropriate responses to these cues are modulated heavily by organizational and activational effects of gonadal sex steroids and thereby intrinsically link chemical communication to the broader reproductive context. Many compounds have been identified as "pheromones" but very few have met the expectations of that term: a unitary, species-typical substance that is both necessary and sufficient for an experience-independent behavioral or physiological response. In contrast, most responses to chemosignals are dependent or heavily modulated by experience, either in adulthood or during development. Mechanistically, chemosignals are perceived by both main and accessory (vomeronasal) olfactory systems with the importance of each system tied strongly to the nature of the stimulus rather than to the response. In the central nervous system, the vast majority of responses to chemosignals are mediated by cortical and medial amygdala connections with hypothalamic and other forebrain structures. Despite the importance of chemosignals in mammals, many details of chemical communication differ even among closely related species and defy clear categorization. Although generating much research and public interest, strong evidence for the existence of a robust chemical communication among humans is lacking.
Collapse
Affiliation(s)
- Aras Petrulis
- Georgia State University, Neuroscience Institute, Atlanta, GA 30303, USA.
| |
Collapse
|
39
|
He Z, Ferguson SA, Cui L, Greenfield LJ, Paule MG. Role of neural stem cell activity in postweaning development of the sexually dimorphic nucleus of the preoptic area in rats. PLoS One 2013; 8:e54927. [PMID: 23383001 PMCID: PMC3559780 DOI: 10.1371/journal.pone.0054927] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 12/19/2012] [Indexed: 02/07/2023] Open
Abstract
The sexually dimorphic nucleus of the preoptic area (SDN-POA) has received increased attention due to its apparent sensitivity to estrogen-like compounds found in food and food containers. The mechanisms that regulate SDN-POA volume remain unclear as is the extent of postweaning development of the SDN-POA. Here we demonstrate that the female Sprague-Dawley SDN-POA volume increased from weaning to adulthood, although this increase was not statistically significant as it was in males. The number of cells positive for Ki67, a marker of cell proliferation, in both the SDN-POA and the hypothalamus was significantly higher at weaning than at adulthood in male rats. In contrast, the number of Ki67-positive cells was significantly higher in the hypothalamus but not in the SDN-POA (p>0.05) at weaning than at adulthood in female rats. A subset of the Ki67-positive cells in the SDN-POA displayed the morphology of dividing cells. Nestin-immunoreactivity delineated a potential macroscopic neural stem cell niche in the rostral end of the 3rd ventricle. In conclusion, stem cells may partially account for the sexually dimorphic postweaning development of the SDN-POA.
Collapse
Affiliation(s)
- Zhen He
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, USA.
| | | | | | | | | |
Collapse
|
40
|
Ottem EN, Bailey DJ, Jordan CL, Breedlove SM. With a little help from my friends: androgens tap BDNF signaling pathways to alter neural circuits. Neuroscience 2012; 239:124-38. [PMID: 23262234 DOI: 10.1016/j.neuroscience.2012.12.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 12/03/2012] [Accepted: 12/05/2012] [Indexed: 12/20/2022]
Abstract
Gonadal androgens are critical for the development and maintenance of sexually dimorphic regions of the male nervous system, which is critical for male-specific behavior and physiological functioning. In rodents, the motoneurons of the spinal nucleus of the bulbocavernosus (SNB) provide a useful example of a neural system dependent on androgen. Unless rescued by perinatal androgens, the SNB motoneurons will undergo apoptotic cell death. In adulthood, SNB motoneurons remain dependent on androgen, as castration leads to somal atrophy and dendritic retraction. In a second vertebrate model, the zebra finch, androgens are critical for the development of several brain nuclei involved in song production in males. Androgen deprivation during a critical period during postnatal development disrupts song acquisition and dimorphic size-associated nuclei. Mechanisms by which androgens exert masculinizing effects in each model system remain elusive. Recent studies suggest that brain-derived neurotrophic factor (BDNF) may play a role in androgen-dependent masculinization and maintenance of both SNB motoneurons and song nuclei of birds. This review aims to summarize studies demonstrating that BDNF signaling via its tyrosine receptor kinase (TrkB) receptor may work cooperatively with androgens to maintain somal and dendritic morphology of SNB motoneurons. We further describe studies that suggest the cellular origin of BDNF is of particular importance in androgen-dependent regulation of SNB motoneurons. We review evidence that androgens and BDNF may synergistically influence song development and plasticity in bird species. Finally, we provide hypothetical models of mechanisms that may underlie androgen- and BDNF-dependent signaling pathways.
Collapse
Affiliation(s)
- E N Ottem
- Department of Biology, Northern Michigan University, Marquette, MI 49855, USA.
| | | | | | | |
Collapse
|
41
|
Soga T, Wong D, Putteeraj M, Song K, Parhar I. Early-life citalopram-induced impairments in sexual behavior and the role of androgen receptor. Neuroscience 2012; 225:172-84. [DOI: 10.1016/j.neuroscience.2012.08.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 08/28/2012] [Accepted: 08/28/2012] [Indexed: 01/27/2023]
|
42
|
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
|
43
|
Abstract
We examined the role of the androgen receptor (AR) in the investigatory behaviour of conspecifics using mice carrying the testicular feminisation mutation (X(Tfm) Y). Responses to members of the same and opposite sex were evaluated in a habituation/dishabituation task. Adult mice were gonadectomised and treated with oestradiol (E(2) ) or testosterone. After E(2) treatment, regardless of the sex of the stimulus mouse, wild-type (WT) males engaged in significantly more investigation than WT females. X(Tfm) Y males treated with E(2) showed 'male-like' behaviour in response to a male but behaved 'female-like' when the stimulus was a female. Because WT and X(Tfm) Y males behaved the same in response to another male, we used two additional mouse models to ask whether sex chromosomes were responsible for this phenomenon. Regardless of sex chromosome complement, gonadal males displayed high levels of investigation. When mice were treated with testosterone, investigation by WT females was enhanced, which eliminated the sex differences. Most strikingly, X(Tfm) Y males receiving testosterone-treatment increased the investigation of females to levels equal to those shown by WT mice. Given that testosterone, but not its metabolite E(2) , caused X(Tfm) Y males to investigate female conspecifics at high levels, it is plausible that nonclassical actions of AR, and/or activation of a novel AR, may be involved in this behaviour. Taken together, our data show that AR activation during adulthood is not required for males to investigate mice of either sex. However, 'male-like' levels of investigation of a female stimulus may depend on neonatal activation of the classic nuclear AR.
Collapse
Affiliation(s)
- L D Tejada
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
44
|
Female mice deficient in alpha-fetoprotein show female-typical neural responses to conspecific-derived pheromones. PLoS One 2012; 7:e39204. [PMID: 22720075 PMCID: PMC3376129 DOI: 10.1371/journal.pone.0039204] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 05/21/2012] [Indexed: 02/03/2023] Open
Abstract
The neural mechanisms controlling sexual behavior are sexually differentiated by the perinatal actions of sex steroid hormones. We recently observed using female mice deficient in alpha-fetoprotein (AFP-KO) and which lack the protective actions of AFP against maternal estradiol, that exposure to prenatal estradiol completely defeminized the potential to show lordosis behavior in adulthood. Furthermore, AFP-KO females failed to show any male-directed mate preferences following treatment with estradiol and progesterone, indicating a reduced sexual motivation to seek out the male. In the present study, we asked whether neural responses to male- and female-derived odors are also affected in AFP-KO female mice. Therefore, we compared patterns of Fos, the protein product of the immediate early gene, c-fos, commonly used as a marker of neuronal activation, between wild-type (WT) and AFP-KO female mice following exposure to male or estrous female urine. We also tested WT males to confirm the previously observed sex differences in neural responses to male urinary odors. Interestingly, AFP-KO females showed normal, female-like Fos responses, i.e. exposure to urinary odors from male but not estrous female mice induced equivalent levels of Fos protein in the accessory olfactory pathways (e.g. the medial part of the preoptic nucleus, the bed nucleus of the stria terminalis, the amygdala, and the lateral part of the ventromedial hypothalamic nucleus) as well as in the main olfactory pathways (e.g. the piriform cortex and the anterior cortical amygdaloid nucleus), as WT females. By contrast, WT males did not show any significant induction of Fos protein in these brain areas upon exposure to either male or estrous female urinary odors. These results thus suggest that prenatal estradiol is not involved in the sexual differentiation of neural Fos responses to male-derived odors.
Collapse
|
45
|
Seney ML, Walsh C, Stolakis R, Sibille E. Neonatal testosterone partially organizes sex differences in stress-induced emotionality in mice. Neurobiol Dis 2012; 46:486-96. [PMID: 22394611 DOI: 10.1016/j.nbd.2012.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 01/29/2012] [Accepted: 02/20/2012] [Indexed: 10/28/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating disorder of altered mood regulation. Despite well established sex differences in MDD prevalence, the mechanism underlying the increased female vulnerability remains unknown. Although evidence suggests an influence of adult circulating hormone levels on mood (i.e. activational effects of hormones), MDD prevalence is consistently higher in women across life stages (and therefore hormonal states), suggesting that additional underlying structural or biological differences place women at higher risk. Studies in human subjects and in rodent models suggest a developmental origin for mood disorders, and interestingly, a developmental process also establishes sex differences in the brain. Hence, based on these parallel developmental trajectories, we hypothesized that a proportion of the female higher vulnerability to MDD may originate from the differential organization of mood regulatory neural networks early in life (i.e. organizational effects of hormones). To test this hypothesis in a rodent system, we took advantage of a well-established technique used in the field of sexual differentiation (neonatal injection with testosterone) to masculinize sexually dimorphic brain regions in female mice. We then investigated adult behavioral consequences relating to emotionality by comparing neonatal testosterone-treated females to normal males and females. Under baseline/trait conditions, neonatal testosterone treatment of female mice did not influence adult emotionality, but masculinized adult locomotor activity, as revealed by the activational actions of hormones. Conversely, the increased vulnerability of female mice to develop high emotionality following unpredictable chronic mild stress (UCMS) was partially masculinized by neonatal testosterone exposure, with no effect on post-UCMS locomotion. The elevated female UCMS-induced vulnerability did not differ between adult hormone treated groups. These results demonstrate that sex differences in adult emotionality in mice are partially caused by the organizational effects of sex hormones during development, hence supporting a developmental hypothesis of the human adult female prevalence of MDD.
Collapse
Affiliation(s)
- Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA 15219, USA
| | | | | | | |
Collapse
|
46
|
Holmes MM, Niel L, Anyan JJ, Griffith AT, Monks DA, Forger NG. Effects of Bax gene deletion on social behaviors and neural response to olfactory cues in mice. Eur J Neurosci 2012; 34:1492-9. [PMID: 22034980 DOI: 10.1111/j.1460-9568.2011.07881.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bax is a pro-death protein that plays a crucial role in developmental neuronal cell death. Bax(-/-) mice exhibit increased neuron number and lack several neural sex differences. Here we examined the effects of Bax gene deletion on social behaviors (olfactory preference, social recognition, social approach and aggression) and the neural processing of olfactory cues. Bax deletion eliminated the normal sex difference in olfactory preference behavior. In the social recognition test, both genotypes discriminated a novel conspecific, but wild-type males and Bax(-/-) animals of both sexes spent much more time than wild-type females investigating stimulus animals. Similarly, Bax(-/-) mice were more sociable than wild-type mice in a social approach test. Bax deletion had no effect on aggression in a resident/intruder paradigm where males, regardless of genotype, exhibited a shorter latency to attack. Thus, the prevention of neuronal cell death by Bax gene deletion results in greater sociability as well as the elimination of sex differences in some social behaviors. To examine olfactory processing of socially relevant cues, we counted c-Fos-immunoreactive (Fos-ir) cells in several nodes of the accessory olfactory pathway after exposure to male-soiled or control bedding. In both genotypes, exposure to male-soiled bedding increased Fos-ir cells in the posterodorsal medial amygdala, principal nucleus of the bed nucleus of the stria terminalis and medial preoptic nucleus (MPN), and the response in the MPN was greater in females than in males. However, a reduction in Fos-ir cells was seen in the anteroventral periventricular nucleus of Bax(-/-) mice.
Collapse
Affiliation(s)
- Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Murray EK, Varnum MM, Fernandez JL, de Vries GJ, Forger NG. Effects of neonatal treatment with valproic acid on vasopressin immunoreactivity and olfactory behaviour in mice. J Neuroendocrinol 2011; 23:906-14. [PMID: 21793947 PMCID: PMC3183375 DOI: 10.1111/j.1365-2826.2011.02196.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent findings demonstrate that epigenetic modifications are required for the sexual differentiation of the brain. For example, neonatal administration of the histone deacetylase inhibitor, valproic acid, blocks masculinisation of cell number in the principal nucleus of the bed nucleus of the stria terminalis (BNST). In the present study, we examined the effects of valproic acid on neurochemistry and behaviour, focusing on traits that are sexually dimorphic and linked to the BNST. Newborn mice were treated with saline or valproic acid and the effect on vasopressin immunoreactivity and olfactory preference behaviour was examined in adulthood. As expected, males had more vasopressin immunoreactive fibres than females in the lateral septum and medial dorsal thalamus, which are two projection sites of BNST vasopressin neurones. Neonatal valproic acid increased vasopressin fibre density specifically in females in the lateral septum, thereby reducing the sex difference, and increased vasopressin fibres in both sexes in the medial dorsal thalamus. The effects were not specific to BNST vasopressin projections, however, because valproic acid also significantly increased vasopressin immunoreactivity in the anterior hypothalamic area in both sexes. Subtle sex-specific effects of neonatal valproic acid treatment were observed on olfactory behaviour. As predicted, males showed a preference for investigating female-soiled bedding, whereas females showed a preference for male-soiled bedding. Valproic acid did not significantly alter olfactory preference, per se, although it increased the number of visits females made to female-soiled bedding and the overall time females spent investigating soiled versus clean bedding. Taken together, these results suggest that a transient disruption of histone deacetylation at birth does not have generalised effects on sexual differentiation, although it does produce lasting effects on brain neurochemistry and behaviour.
Collapse
Affiliation(s)
- E K Murray
- Neuroscience and Behavior Program, Department of Psychology and Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Maternal stress experience is associated with neurodevelopmental disorders including schizophrenia and autism. Recent studies have examined mechanisms by which changes in the maternal milieu may be transmitted to the developing embryo and potentially translated into programming of the epigenome. Animal models of prenatal stress have identified important sex- and temporal-specific effects on offspring stress responsivity. As dysregulation of stress pathways is a common feature in most neuropsychiatric diseases, molecular and epigenetic analyses at the maternal-embryo interface, especially in the placenta, may provide unique insight into identifying much-needed predictive biomarkers. In addition, as most neurodevelopmental disorders present with a sex bias, examination of sex differences in the inheritance of phenotypic outcomes may pinpoint gene targets and specific windows of vulnerability in neurodevelopment, which have been disrupted. This review discusses the association and possible contributing mechanisms of prenatal stress in programming offspring stress pathway dysregulation and the importance of sex.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Henley CL, Nunez AA, Clemens LG. Hormones of choice: the neuroendocrinology of partner preference in animals. Front Neuroendocrinol 2011; 32:146-54. [PMID: 21377487 DOI: 10.1016/j.yfrne.2011.02.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 02/17/2011] [Accepted: 02/28/2011] [Indexed: 11/29/2022]
Abstract
Partner preference behavior can be viewed as the outcome of a set of hierarchical choices made by an individual in anticipation of mating. The first choice involves approaching a conspecific verses an individual of another species. As a rule, a conspecific is picked as a mating partner, but early life experiences can alter that outcome. Within a species, an animal then has the choice between a member of the same sex or the opposite sex. The final choice is for a specific individual. This review will focus on the middle choice, the decision to mate with either a male or a female. Available data from rats, mice, and ferrets point to the importance of perinatal exposure to steroid hormones in the development of partner preferences, as well as the importance of activational effects in adulthood. However, the particular effects of this hormone exposure show species differences in both the specific steroid hormone responsible for the organization of behavior and the developmental period when it has its effect. Where these hormones have an effect in the brain is mostly unknown, but regions involved in olfaction and sexual behavior, as well as sexually dimorphic regions, seem to play a role. One limitation of the literature base is that many mate or 'partner preference studies' rely on preference for a specific stimulus (usually olfaction) but do not include an analysis of the relation, if any, that stimulus has to the choice of a particular sexual partner. A second limitation has been the almost total lack of attention to the type of behavior that is shown by the choosing animal once a 'partner' has been chosen, specifically, if the individual plays a mating role typical of its own sex or the opposite sex. Additional paradigms that address these questions are needed for better understanding of partner preferences in rodents.
Collapse
Affiliation(s)
- C L Henley
- Departments of Zoology and Psychology, and the Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | | | | |
Collapse
|