1
|
Geleta U, Prajapati P, Bachstetter A, Nelson PT, Wang WX. Sex-Biased Expression and Response of microRNAs in Neurological Diseases and Neurotrauma. Int J Mol Sci 2024; 25:2648. [PMID: 38473893 PMCID: PMC10931569 DOI: 10.3390/ijms25052648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Neurological diseases and neurotrauma manifest significant sex differences in prevalence, progression, outcome, and therapeutic responses. Genetic predisposition, sex hormones, inflammation, and environmental exposures are among many physiological and pathological factors that impact the sex disparity in neurological diseases. MicroRNAs (miRNAs) are a powerful class of gene expression regulator that are extensively involved in mediating biological pathways. Emerging evidence demonstrates that miRNAs play a crucial role in the sex dimorphism observed in various human diseases, including neurological diseases. Understanding the sex differences in miRNA expression and response is believed to have important implications for assessing the risk of neurological disease, defining therapeutic intervention strategies, and advancing both basic research and clinical investigations. However, there is limited research exploring the extent to which miRNAs contribute to the sex disparities observed in various neurological diseases. Here, we review the current state of knowledge related to the sexual dimorphism in miRNAs in neurological diseases and neurotrauma research. We also discuss how sex chromosomes may contribute to the miRNA sexual dimorphism phenomenon. We attempt to emphasize the significance of sexual dimorphism in miRNA biology in human diseases and to advocate a gender/sex-balanced science.
Collapse
Affiliation(s)
- Urim Geleta
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Paresh Prajapati
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
| | - Adam Bachstetter
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Neuroscience, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Peter T. Nelson
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (U.G.); (P.P.); (A.B.); (P.T.N.)
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Pathology and Laboratory Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
2
|
Smiley KO, Munley KM, Aghi K, Lipshutz SE, Patton TM, Pradhan DS, Solomon-Lane TK, Sun SED. Sex diversity in the 21st century: Concepts, frameworks, and approaches for the future of neuroendocrinology. Horm Behav 2024; 157:105445. [PMID: 37979209 PMCID: PMC10842816 DOI: 10.1016/j.yhbeh.2023.105445] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 11/20/2023]
Abstract
Sex is ubiquitous and variable throughout the animal kingdom. Historically, scientists have used reductionist methodologies that rely on a priori sex categorizations, in which two discrete sexes are inextricably linked with gamete type. However, this binarized operationalization does not adequately reflect the diversity of sex observed in nature. This is due, in part, to the fact that sex exists across many levels of biological analysis, including genetic, molecular, cellular, morphological, behavioral, and population levels. Furthermore, the biological mechanisms governing sex are embedded in complex networks that dynamically interact with other systems. To produce the most accurate and scientifically rigorous work examining sex in neuroendocrinology and to capture the full range of sex variability and diversity present in animal systems, we must critically assess the frameworks, experimental designs, and analytical methods used in our research. In this perspective piece, we first propose a new conceptual framework to guide the integrative study of sex. Then, we provide practical guidance on research approaches for studying sex-associated variables, including factors to consider in study design, selection of model organisms, experimental methodologies, and statistical analyses. We invite fellow scientists to conscientiously apply these modernized approaches to advance our biological understanding of sex and to encourage academically and socially responsible outcomes of our work. By expanding our conceptual frameworks and methodological approaches to the study of sex, we will gain insight into the unique ways that sex exists across levels of biological organization to produce the vast array of variability and diversity observed in nature.
Collapse
Affiliation(s)
- Kristina O Smiley
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, 639 North Pleasant Street, Morrill IVN Neuroscience, Amherst, MA 01003, USA.
| | - Kathleen M Munley
- Department of Psychology, University of Houston, 3695 Cullen Boulevard, Houston, TX 77204, USA.
| | - Krisha Aghi
- Department of Integrative Biology and Physiology, University of California Los Angeles, 405 Hilgard Ave, Los Angeles, CA 90095, USA.
| | - Sara E Lipshutz
- Department of Biology, Duke University, 130 Science Drive, Durham, NC 27708, USA.
| | - Tessa M Patton
- Bioinformatics Program, Loyola University Chicago, 1032 West Sheridan Road, LSB 317, Chicago, IL 60660, USA.
| | - Devaleena S Pradhan
- Department of Biological Sciences, Idaho State University, 921 South 8th Avenue, Mail Stop 8007, Pocatello, ID 83209, USA.
| | - Tessa K Solomon-Lane
- Scripps, Pitzer, Claremont McKenna Colleges, 925 North Mills Avenue, Claremont, CA 91711, USA.
| | - Simón E D Sun
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
3
|
Holm SM, Balmes JR, Gunier RB, Kogut K, Harley KG, Eskenazi B. Cognitive Development and Prenatal Air Pollution Exposure in the CHAMACOS Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:37007. [PMID: 36913239 PMCID: PMC10010399 DOI: 10.1289/ehp10812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Because fine particulate matter [PM, with aerodynamic diameter ≤ 2.5 μ m (PM 2.5 )] is a ubiquitous environmental exposure, small changes in cognition associated with PM 2.5 exposure could have great societal costs. Prior studies have demonstrated a relationship between in utero PM 2.5 exposure and cognitive development in urban populations, but it is not known whether these effects are similar in rural populations and whether they persist into late childhood. OBJECTIVES In this study, we tested for associations between prenatal PM 2.5 exposure and both full-scale and subscale measures of IQ among a longitudinal cohort at age 10.5 y. METHODS This analysis used data from 568 children enrolled in the Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS), a birth cohort study in California's agricultural Salinas Valley. Exposures were estimated at residential addresses during pregnancy using state of the art, modeled PM 2.5 surfaces. IQ testing was performed by bilingual psychometricians in the dominant language of the child. RESULTS A 3 - μ g / m 3 higher average PM 2.5 over pregnancy was associated with - 1.79 full-scale IQ points [95% confidence interval (CI): - 2.98 , - 0.58 ], with decrements specifically in Working Memory IQ (WMIQ) and Processing Speed IQ (PSIQ) subscales [WMIQ - 1.72 (95% CI: - 2.98 , - 0.45 ) and PSIQ - 1.19 (95% CI: - 2.54 , 0.16)]. Flexible modeling over the course of pregnancy illustrated mid-to-late pregnancy (months 5-7) as particularly susceptible times, with sex differences in the timing of susceptible windows and in which subscales were most affected [Verbal Comprehension IQ (VCIQ) and WMIQ in males; and PSIQ in females]. DISCUSSION We found that small increases in outdoor PM 2.5 exposure in utero were associated with slightly lower IQ in late childhood, robust to many sensitivity analyses. In this cohort there was a larger effect of PM 2.5 on childhood IQ than has previously been observed, perhaps due to differences in PM composition or because developmental disruption could alter the cognitive trajectory and thus appear more pronounced as children get older. https://doi.org/10.1289/EHP10812.
Collapse
Affiliation(s)
- Stephanie M. Holm
- Division of Epidemiology, School of Public Health, University of California Berkeley, Berkeley, California, USA
- Western States Pediatric Environmental Health Specialty Unit, University of California San Francisco, San Francisco, California, USA
- Division of Occupational and Environmental Medicine, University of California San Francisco San Francisco, California, USA
| | - John R. Balmes
- Western States Pediatric Environmental Health Specialty Unit, University of California San Francisco, San Francisco, California, USA
- Division of Occupational and Environmental Medicine, University of California San Francisco San Francisco, California, USA
- Division of Environmental Health Sciences, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Robert B. Gunier
- Center for Environmental Research and Children’s Health, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Katherine Kogut
- Center for Environmental Research and Children’s Health, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Kim G. Harley
- Center for Environmental Research and Children’s Health, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Brenda Eskenazi
- Center for Environmental Research and Children’s Health, School of Public Health, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
4
|
Swaab DF, Wolff SEC, Bao AM. Sexual differentiation of the human hypothalamus: Relationship to gender identity and sexual orientation. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:427-443. [PMID: 34238476 DOI: 10.1016/b978-0-12-820683-6.00031-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gender identity (an individual's perception of being male or female) and sexual orientation (heterosexuality, homosexuality, or bisexuality) are programmed into our brain during early development. During the intrauterine period in the second half of pregnancy, a testosterone surge masculinizes the fetal male brain. If such a testosterone surge does not occur, this will result in a feminine brain. As sexual differentiation of the brain takes place at a much later stage in development than sexual differentiation of the genitals, these two processes can be influenced independently of each other and can result in gender dysphoria. Nature produces a great variability for all aspects of sexual differentiation of the brain. Mechanisms involved in sexual differentiation of the brain include hormones, genetics, epigenetics, endocrine disruptors, immune response, and self-organization. Furthermore, structural and functional differences in the hypothalamus relating to gender dysphoria and sexual orientation are described in this review. All the genetic, postmortem, and in vivo scanning observations support the neurobiological theory about the origin of gender dysphoria, i.e., it is the sizes of brain structures, the neuron numbers, the molecular composition, functions, and connectivity of brain structures that determine our gender identity or sexual orientation. There is no evidence that one's postnatal social environment plays a crucial role in the development of gender identity or sexual orientation.
Collapse
Affiliation(s)
- Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Samantha E C Wolff
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Premachandran H, Zhao M, Arruda-Carvalho M. Sex Differences in the Development of the Rodent Corticolimbic System. Front Neurosci 2020; 14:583477. [PMID: 33100964 PMCID: PMC7554619 DOI: 10.3389/fnins.2020.583477] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, a growing body of research has shown sex differences in the prevalence and symptomatology of psychopathologies, such as depression, anxiety, and fear-related disorders, all of which show high incidence rates in early life. This has highlighted the importance of including female subjects in animal studies, as well as delineating sex differences in neural processing across development. Of particular interest is the corticolimbic system, comprising the hippocampus, amygdala, and medial prefrontal cortex. In rodents, these corticolimbic regions undergo dynamic changes in early life, and disruption to their normative development is believed to underlie the age and sex-dependent effects of stress on affective processing. In this review, we consolidate research on sex differences in the hippocampus, amygdala, and medial prefrontal cortex across early development. First, we briefly introduce current principles on sexual differentiation of the rodent brain. We then showcase corticolimbic regional sex differences in volume, morphology, synaptic organization, cell proliferation, microglia, and GABAergic signaling, and explain how these differences are influenced by perinatal and pubertal gonadal hormones. In compiling this research, we outline evidence of what and when sex differences emerge in the developing corticolimbic system, and illustrate how temporal dynamics of its maturational trajectory may differ in male and female rodents. This will help provide insight into potential neural mechanisms underlying sex-specific critical windows for stress susceptibility and behavioral emergence.
Collapse
Affiliation(s)
| | - Mudi Zhao
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
6
|
Caballero-Huertas M, Moraleda-Prados J, Joly S, Ribas L. Immune genes, IL1β and Casp9, show sexual dimorphic methylation patterns in zebrafish gonads. FISH & SHELLFISH IMMUNOLOGY 2020; 97:648-655. [PMID: 31830572 DOI: 10.1016/j.fsi.2019.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 06/10/2023]
Abstract
There is crosstalk between the immune and reproductive systems in which sexual dimorphism is a common pattern in vertebrates. In recent years, epigenetics has emerged as a way to study the molecular mechanisms involved in gonadal development, those responsible for integrating environmental information that contribute to assigning a specific sexual phenotype (either an ovary or a testis). The knowledge of epigenetic mechanisms in certain molecular processes allows the development of epigenetic markers. In fish gonads, the existence of reproduction-immune system interactions is known, although the epigenetic mechanisms involved are far from clear. Here, we used the zebrafish (Danio rerio) as a model to study the DNA methylation patterns in gonads of two well-known innate immune genes: IL1β and Casp9. DNA methylation levels were studied by a candidate gene approach at single nucleotide resolution and gene expression analyses were also carried out. Results showed that there was clear sexual dimorphism in the DNA methylation levels of the two immune genes studied, being significantly higher in the testes when compared to the ovaries. In summary, and although further research is needed, this paper presents sexual dimorphic methylation patterns of two immune-related genes, thus sex-biased differences in methylation profiles should considered when analyzing immune responses in fish. Data showed here can help to develop epimarkers with forthcoming applications in livestock and fish farming production, for example, in immune fish diseases or sexual control programs as epigenetic molecular tools to predict environmental pressure in the gonads.
Collapse
Affiliation(s)
- M Caballero-Huertas
- Institute of Marine Sciences, Spanish National Research Council (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain
| | - J Moraleda-Prados
- Institute of Marine Sciences, Spanish National Research Council (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain
| | - S Joly
- Institute of Marine Sciences, Spanish National Research Council (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain
| | - L Ribas
- Institute of Marine Sciences, Spanish National Research Council (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain.
| |
Collapse
|
7
|
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
|
8
|
Breton E, Gagné-Ouellet V, Thibeault K, Guérin R, Van Lieshout R, Perron P, Hivert M, Bouchard L. Placental NEGR1 DNA methylation is associated with BMI and neurodevelopment in preschool-age children. Epigenetics 2019; 15:323-335. [PMID: 31510847 DOI: 10.1080/15592294.2019.1666653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Studies have linked maternal pre-pregnancy obesity and hyperglycaemia with metabolic and neurodevelopmental complications in childhood. DNA methylation (DNAm) might enable foetal adaptations to environmental adversities through important gene loci. NEGR1 is involved in both energy balance and behaviour regulation. The aim of this study was to investigate associations between placental DNAm at the NEGR1 gene locus and childhood anthropometric and neurodevelopmental profiles in preschoolers. We analysed 276 mother-child dyads from Gen3G, a prospective birth cohort from Sherbrooke. At 3yo (40.4 ± 3.0 months), we measured body mass index (BMI) and the mothers reported on offspring neurobehavior using the Strengths and Difficulties Questionnaire (SDQ). We quantified DNAm levels at 30 CpGs at the NEGR1 locus using the MethylationEPIC Array in placental biopsies. DNAm at four CpGs located before NEGR1 second exon predicted child's BMI z-score (cg26153364: β=-0.16 ± 0.04; p=0.008, cg23166710: β=0.14 ± 0.08; p=0.03) and SDQ total score (cg04932878: β=0.22 ± 1.0; p= 3.0x10-4, cg16525738: β=-0.14 ± 0.18; p=0.01, cg23166710: β=-0.13 ± 0.36; p= 0.04), explaining 4.2% (p=0.003) and 7.3% (p= 1.3 x 10-4) of BMI-z and SDQ variances. cg23166710 was associated with both childhood phenotypes and correlated with NEGR1 placental expression (r=-0.22, p=0.04), suggesting its possible functional role. Together, maternal metabolic characteristics during pregnancy with NEGR1 DNAm levels explained 7.4% (p=4.2 x 10-4) of BMI-z and 14.2% (p=2.8 x 10-7) of SDQ variance at 3yo. This longitudinal study suggests that placental NEGR1 DNAm is associated with adiposity and neurodevelopment in preschool children and highlights its potential role in their comorbidity.
Collapse
Affiliation(s)
- E Breton
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - V Gagné-Ouellet
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - K Thibeault
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - R Guérin
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean - Hôpital de Chicoutimi, Saguenay, QC, Canada
| | - Rj Van Lieshout
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - P Perron
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mf Hivert
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA.,Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - L Bouchard
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medical Biology, CIUSSS Saguenay-Lac-Saint-Jean - Hôpital de Chicoutimi, Saguenay, QC, Canada
| |
Collapse
|
9
|
Xu W, Huang J, Li L, Zhang X, Wang Y, Tong G, Sun Y. Alterations of estradiol-induced histone H3 acetylation in the preoptic area and anteroventral periventricular nucleus of middle-aged female rats. Biochem Biophys Res Commun 2019; 516:894-899. [PMID: 31272713 DOI: 10.1016/j.bbrc.2019.06.145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
In this study we investigated the characteristics of histone H3 acetylation in the anterior hypothalamus under E2 positive feedback to gain a better understanding of the mechanism underlying reduced GnRH neuron activation and altered gene expression in female reproductive aging. Young and middle-aged female rats were ovariectomized (OVX) and treated with estradiol (E2) or oil. C-Fos expression, the number of GnRH neurons co-localized with c-Fos in the preoptic area (POA), and the number of acetylated histone H3 cells in the POA and anteroventral periventricular nucleus (AVPV) were quantified at the time of the expected GnRH neuron activation. We used real-time PCR to evaluate the expression of Esr1 target genes including Kiss1 and VGluT2 and genes known as Esr1 coregulators in the anterior hypothalamus. Our results show that in the young females, E2 markedly increased histone H3 acetylation in the POA and AVPV, coincident with increased c-Fos and GnRH neuron activation in the POA. In middle-aged females, E2-induced histone H3 acetylation was reduced in the POA but was not significantly altered in the AVPV. This occurred in association with a reduction of c-Fos expression and the number of GnRH cells expressing c-Fos in the POA as well as a down-regulation of Kiss1 and VGluT2 mRNA expression in the anterior hypothalamus of the animals. E2 caused significant decreases in Ncoa2 and Crebbp mRNA expression in the anterior hypothalamus of young, but not middle-aged females. Taken together, these data suggest that alterations of histone H3 acetylation in the POA and AVPV and the inability of Ncoa2 and Crebbp to respond to E2 in the middle-aged anterior hypothalamus partially contribute to the decline of GnRH neuron activation and E2 target gene expression changes that occur in female along with reproductive aging.
Collapse
Affiliation(s)
- Wen Xu
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Jianqin Huang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China; The Academy of Integrative Medicine of Fudan University, Shanghai, 200011, China
| | - Lisha Li
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Xinyan Zhang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yan Wang
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Guoqing Tong
- Reproduction Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yan Sun
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China; The Academy of Integrative Medicine of Fudan University, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease, Shanghai, 200011, China.
| |
Collapse
|
10
|
Xu W, An X, Zhang N, Li L, Zhang X, Wang Y, Wang L, Sun Y. Middle-aged female rats lack changes in histone H3 acetylation in the anterior hypothalamus observed in young females on the day of a luteinizing hormone surge. Biosci Trends 2019; 13:334-341. [PMID: 31434815 DOI: 10.5582/bst.2019.01162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Histone acetylation has recently been implicated in gene transcription and estradiol (E2) actions in the hypothalamus. This study aims to determine the involvement of histone acetylation in mediating E2-induced luteinizing hormone (LH) surge to understand the mechanism underlying LH surge dysfunction in female reproductive aging. Young and middle-aged female rats were ovariectomized (OVX) and treated with hormone or oil once per day for two days. At the time of the expected LH surge, blood samples were taken for LH assay. The anterior and posterior hypothalami were dissected, histone H3/H4 acetylation and histone deacetylases (HDACs) 4, -5, -10 and -11 protein expressions were measured using Western blotting. Our results show that in the young females, E2 markedly increased histone H3 acetylation while significantly reducing HDAC10 protein expression in the anterior hypothalamus. Notably, E2-induced alterations of histone H3 acetylation and HDAC10 in the anterior hypothalamus were absent in middle-aged females, associated with a reduced LH release. However, age alters histone H4 acetylation in both the anterior and posterior hypothalamus and significantly increased HDAC 4 and -5 protein expression in the anterior hypothalamus. Taken together, these data suggest that histone H3 acetylation in the anterior hypothalamus may mediate E2 regulation of LH surge and the process possibly through decreasing HDAC10. The missed responsiveness of histone H3 acetylation and HDAC10 expression to E2 in the anterior hypothalamus may contribute to LH surge failure that occurs in female reproductive aging.
Collapse
Affiliation(s)
- Wen Xu
- Hospital of Obstetrics and Gynecology, Fudan University
| | - Xiaofei An
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Na Zhang
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease
| | - Lisha Li
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease
| | - Xinyan Zhang
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease
| | - Yan Wang
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease
| | - Ling Wang
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease.,The Academy of Integrative Medicine of Fudan University
| | - Yan Sun
- Hospital of Obstetrics and Gynecology, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease.,The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
11
|
Post-translational histone modifications and their interaction with sex influence normal brain development and elaboration of neuropsychiatric disorders. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1968-1981. [DOI: 10.1016/j.bbadis.2018.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023]
|
12
|
Gegenhuber B, Tollkuhn J. Sex Differences in the Epigenome: A Cause or Consequence of Sexual Differentiation of the Brain? Genes (Basel) 2019; 10:genes10060432. [PMID: 31181654 PMCID: PMC6627918 DOI: 10.3390/genes10060432] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
Females and males display differences in neural activity patterns, behavioral responses, and incidence of psychiatric and neurological diseases. Sex differences in the brain appear throughout the animal kingdom and are largely a consequence of the physiological requirements necessary for the distinct roles of the two sexes in reproduction. As with the rest of the body, gonadal steroid hormones act to specify and regulate many of these differences. It is thought that transient hormonal signaling during brain development gives rise to persistent sex differences in gene expression via an epigenetic mechanism, leading to divergent neurodevelopmental trajectories that may underlie sex differences in disease susceptibility. However, few genes with a persistent sex difference in expression have been identified, and only a handful of studies have employed genome-wide approaches to assess sex differences in epigenomic modifications. To date, there are no confirmed examples of gene regulatory elements that direct sex differences in gene expression in the brain. Here, we review foundational studies in this field, describe transcriptional mechanisms that could act downstream of hormone receptors in the brain, and suggest future approaches for identification and validation of sex-typical gene programs. We propose that sexual differentiation of the brain involves self-perpetuating transcriptional states that canalize sex-specific development.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
13
|
Sexual dimorphism in brain transcriptomes of Amami spiny rats (Tokudaia osimensis): a rodent species where males lack the Y chromosome. BMC Genomics 2019; 20:87. [PMID: 30683046 PMCID: PMC6347839 DOI: 10.1186/s12864-019-5426-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Brain sexual differentiation is sculpted by precise coordination of steroid hormones during development. Programming of several brain regions in males depends upon aromatase conversion of testosterone to estrogen. However, it is not clear the direct contribution that Y chromosome associated genes, especially sex-determining region Y (Sry), might exert on brain sexual differentiation in therian mammals. Two species of spiny rats: Amami spiny rat (Tokudaia osimensis) and Tokunoshima spiny rat (T. tokunoshimensis) lack a Y chromosome/Sry, and these individuals possess an XO chromosome system in both sexes. Both Tokudaia species are highly endangered. To assess the neural transcriptome profile in male and female Amami spiny rats, RNA was isolated from brain samples of adult male and female spiny rats that had died accidentally and used for RNAseq analyses. Results RNAseq analyses confirmed that several genes and individual transcripts were differentially expressed between males and females. In males, seminal vesicle secretory protein 5 (Svs5) and cytochrome P450 1B1 (Cyp1b1) genes were significantly elevated compared to females, whereas serine (or cysteine) peptidase inhibitor, clade A, member 3 N (Serpina3n) was upregulated in females. Many individual transcripts elevated in males included those encoding for zinc finger proteins, e.g. zinc finger protein X-linked (Zfx). Conclusions This method successfully identified several genes and transcripts that showed expression differences in the brain of adult male and female Amami spiny rat. The functional significance of these findings, especially differential expression of transcripts encoding zinc finger proteins, in this unusual rodent species remains to be determined. Electronic supplementary material The online version of this article (10.1186/s12864-019-5426-6) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Huang Y, Jian W, Zhao J, Wang G. Overexpression of HDAC9 is associated with poor prognosis and tumor progression of breast cancer in Chinese females. Onco Targets Ther 2018; 11:2177-2184. [PMID: 29713186 PMCID: PMC5909784 DOI: 10.2147/ott.s164583] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Breast cancer represents a serious health issue among females. HDAC9 has been identified as an oncogene in human cancers. This study sought to assess the prognostic value and the biologic function of HDAC9 in breast cancer patients. METHODS Expression of HDAC9 in breast cancer tissues and cells was evaluated by quantitative real-time polymerase chain reaction. Kaplan-Meier survival analysis and Cox regression assay were conducted to explore the prognostic significance of HDAC9. Cell experiments were performed to investigate the effects of HDAC9 on the biologic behaviors of breast cancer cells. RESULTS Expression of HDAC9 was significantly upregulated in both cancerous tissues and cells compared with the normal controls (all P<0.05). Overexpression of HDAC9 was correlated with lymph node metastasis (P=0.021) and TNM stage (P=0.004). Patients with high HDAC9 had poor overall survival compared to those with low levels of HDAC9 (log-rank P<0.05). Elevated HDAC9 was found to be an independent prognostic factor for the patients (hazard ratio=2.996, 95% CI=1.611-5.572, P=0.001). According to the cell experiments, tumor cell proliferation, migration and invasion were suppressed by knockdown of HDAC9. CONCLUSION All data demonstrated that overexpression of HDAC9 serves as a prognostic biomarker and may be involved in the tumor progression of breast cancer.
Collapse
Affiliation(s)
- Yixiang Huang
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Wei Jian
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Junyong Zhao
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Gang Wang
- Department of General Surgery, Tenth People’s Hospital of Tongji University, Shanghai, China
| |
Collapse
|
15
|
Rosenfeld CS. Brain Sexual Differentiation and Requirement of SRY: Why or Why Not? Front Neurosci 2017; 11:632. [PMID: 29200993 PMCID: PMC5696354 DOI: 10.3389/fnins.2017.00632] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022] Open
Abstract
Brain sexual differentiation is orchestrated by precise coordination of sex steroid hormones. In some species, programming of select male brain regions is dependent upon aromatization of testosterone to estrogen. In mammals, these hormones surge during the organizational and activational periods that occur during perinatal development and adulthood, respectively. In various fish and reptiles, incubation temperature during a critical embryonic period results in male or female sexual differentiation, but this can be overridden in males by early exposure to estrogenic chemicals. Testes development in mammals requires a Y chromosome and testis determining gene SRY (in humans)/Sry (all other therian mammals), although there are notable exceptions. Two species of spiny rats: Amami spiny rat (Tokudaia osimensis) and Tokunoshima spiny rat (Tokudaia tokunoshimensis) and two species of mole voles (Ellobius lutescens and Ellobius tancrei), lack a Y chromosome/Sry and possess an XO chromosome system in both sexes. Such rodent species, prototherians (monotremes, who also lack Sry), and fish and reptile species that demonstrate temperature sex determination (TSD) seemingly call into question the requirement of Sry for brain sexual differentiation. This review will consider brain regions expressing SRY/Sry in humans and rodents, respectively, and potential roles of SRY/Sry in the brain will be discussed. The evidence from various taxa disputing the requirement of Sry for brain sexual differentiation in mammals (therians and prototherians) and certain fish and reptilian species will be examined. A comparative approach to address this question may elucidate other genes, pathways, and epigenetic modifications stimulating brain sexual differentiation in vertebrate species, including humans.
Collapse
Affiliation(s)
- Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.,Biomedical Sciences, University of Missouri, Columbia, MO, United States.,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, United States.,Genetics Area Program, University of Missouri, Columbia, MO, United States
| |
Collapse
|
16
|
Cambiasso MJ, Cisternas CD, Ruiz-Palmero I, Scerbo MJ, Arevalo MA, Azcoitia I, Garcia-Segura LM. Interaction of sex chromosome complement, gonadal hormones and neuronal steroid synthesis on the sexual differentiation of mammalian neurons. J Neurogenet 2017; 31:300-306. [DOI: 10.1080/01677063.2017.1390572] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Maria Julia 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
| | - Carla Daniela 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
| | - Isabel Ruiz-Palmero
- CSIC, Instituto Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Julia Scerbo
- 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
| | - Maria Angeles Arevalo
- CSIC, Instituto Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Iñigo Azcoitia
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Faculty of Biology, Universidad Complutense, Ciudad Universitaria, Madrid, Spain
| | - Luis M. Garcia-Segura
- CSIC, Instituto Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Morishita M, Maejima S, Tsukahara S. Gonadal Hormone-Dependent Sexual Differentiation of a Female-Biased Sexually Dimorphic Cell Group in the Principal Nucleus of the Bed Nucleus of the Stria Terminalis in Mice. Endocrinology 2017; 158:3512-3525. [PMID: 28977609 DOI: 10.1210/en.2017-00240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/27/2017] [Indexed: 11/19/2022]
Abstract
We recently reported a female-biased sexually dimorphic area in the mouse brain in the boundary region between the preoptic area and the bed nucleus of the stria terminalis (BNST). We reexamined this area and found that it is a ventral part of the principal nucleus of the BNST (BNSTp). The BNSTp is a male-biased sexually dimorphic nucleus, but the ventral part of the BNSTp (BNSTpv) exhibits female-biased sex differences in volume and neuron number. The volume and neuron number of the BNSTpv were increased in males by neonatal orchiectomy and decreased in females by treatment with testosterone, dihydrotestosterone, or estradiol within 5 days after birth. Sex differences in the volume and neuron number of the BNSTpv emerged before puberty. These sex differences became prominent in adulthood with increasing volume in females and loss of neurons in males during the pubertal/adolescent period. Prepubertal orchiectomy did not affect the BNSTpv, although prepubertal ovariectomy reduced the volume increase and induced loss of neurons in the female BNSTpv. In contrast, the volume and neuron number of male-biased sexually dimorphic nuclei that are composed of mainly calbindin neurons and are located in the preoptic area and BNST were decreased by prepubertal orchiectomy but not affected by prepubertal ovariectomy. Testicular testosterone during the postnatal period may defeminize the BNSTpv via binding directly to the androgen receptor and indirectly to the estrogen receptor after aromatization, although defeminization may proceed independently of testicular hormones in the pubertal/adolescent period. Ovarian hormones may act to feminize the BNSTpv during the pubertal/adolescent period.
Collapse
Affiliation(s)
- Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Sho Maejima
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| |
Collapse
|
18
|
Abstract
The prevalence, age of onset, and clinical symptoms of many neuropsychiatric diseases substantially differ between males and females. Factors influencing the relationships between brain development and function and sex or gender may help us understand the differences between males and females in terms of risk or resilience factors in brain diseases.
Collapse
Affiliation(s)
- Florence Thibaut
- University Hospital Cochin (Site Tarnier), Faculty of Medicine Paris Descartes), INSERM U 894, CNP, Paris, France
| |
Collapse
|
19
|
Stoney PN, Rodrigues D, Helfer G, Khatib T, Ashton A, Hay EA, Starr R, Kociszewska D, Morgan P, McCaffery P. A seasonal switch in histone deacetylase gene expression in the hypothalamus and their capacity to modulate nuclear signaling pathways. Brain Behav Immun 2017; 61:340-352. [PMID: 27993690 PMCID: PMC5325119 DOI: 10.1016/j.bbi.2016.12.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 12/01/2016] [Accepted: 12/13/2016] [Indexed: 12/28/2022] Open
Abstract
Seasonal animals undergo changes in physiology and behavior between summer and winter conditions. These changes are in part driven by a switch in a series of hypothalamic genes under transcriptional control by hormones and, of recent interest, inflammatory factors. Crucial to the control of transcription are histone deacetylases (HDACs), generally acting to repress transcription by local histone modification. Seasonal changes in hypothalamic HDAC transcripts were investigated in photoperiod-sensitive F344 rats by altering the day-length (photoperiod). HDAC4, 6 and 9 were found to change in expression. The potential influence of HDACs on two hypothalamic signaling pathways that regulate transcription, inflammatory and nuclear receptor signaling, was investigated. For inflammatory signaling the focus was on NF-κB because of the novel finding made that its expression is seasonally regulated in the rat hypothalamus. For nuclear receptor signaling it was discovered that expression of retinoic acid receptor beta was regulated seasonally. HDAC modulation of NF-κB-induced pathways was examined in a hypothalamic neuronal cell line and primary hypothalamic tanycytes. HDAC4/5/6 inhibition altered the control of gene expression (Fos, Prkca, Prkcd and Ptp1b) by inducers of NF-κB that activate inflammation. These inhibitors also modified the action of nuclear receptor ligands thyroid hormone and retinoic acid. Thus seasonal changes in HDAC4 and 6 have the potential to epigenetically modify multiple gene regulatory pathways in the hypothalamus that could act to limit inflammatory pathways in the hypothalamus during long-day summer-like conditions.
Collapse
Affiliation(s)
- Patrick N. Stoney
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Diana Rodrigues
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Gisela Helfer
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, Scotland, UK,Faculty of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, UK
| | - Thabat Khatib
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Anna Ashton
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Elizabeth A. Hay
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Robert Starr
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Dagmara Kociszewska
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Peter Morgan
- Rowett Institute of Nutrition and Health, University of Aberdeen, Greenburn Road, Bucksburn, Aberdeen AB21 9SB, Scotland, UK
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
20
|
Moreno A, Gan C, Zasler ND. Neurosexuality: A transdisciplinary approach to sexuality in neurorehabilitation. NeuroRehabilitation 2017; 41:255-259. [PMID: 29036838 DOI: 10.3233/nre-001480] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Alexander Moreno
- Faculty of Human Sciences, Department of Sexology, Université du Québec à Montréal (UQÀM), Canada
- Center for Interdisciplinary Research in Rehabilitation of Greater Montreal, Canada
| | - Caron Gan
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Nathan D Zasler
- Concussion Care Centre of Virginia, Ltd. and Tree of Life Services, Inc., Henrico, VA, USA
| |
Collapse
|
21
|
Walker DM, Gore AC. Epigenetic impacts of endocrine disruptors in the brain. Front Neuroendocrinol 2017; 44:1-26. [PMID: 27663243 PMCID: PMC5429819 DOI: 10.1016/j.yfrne.2016.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/05/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
The acquisition of reproductive competence is organized and activated by steroid hormones acting upon the hypothalamus during critical windows of development. This review describes the potential role of epigenetic processes, particularly DNA methylation, in the regulation of sexual differentiation of the hypothalamus by hormones. We examine disruption of these processes by endocrine-disrupting chemicals (EDCs) in an age-, sex-, and region-specific manner, focusing on how perinatal EDCs act through epigenetic mechanisms to reprogram DNA methylation and sex steroid hormone receptor expression throughout life. These receptors are necessary for brain sexual differentiation and their altered expression may underlie disrupted reproductive physiology and behavior. Finally, we review the literature on histone modifications and non-coding RNA involvement in brain sexual differentiation and their perturbation by EDCs. By putting these data into a sex and developmental context we conclude that perinatal EDC exposure alters the developmental trajectory of reproductive neuroendocrine systems in a sex-specific manner.
Collapse
Affiliation(s)
- Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1065, New York, NY 10029, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, and The University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
22
|
Schlenker EH. Sexual dimorphism of cardiopulmonary regulation in the arcuate nucleus of the hypothalamus. Respir Physiol Neurobiol 2016; 245:37-44. [PMID: 27756648 DOI: 10.1016/j.resp.2016.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 11/19/2022]
Abstract
The arcuate nucleus of the hypothalamus (ANH) interacts with other hypothalamic nuclei, forebrain regions, and downstream brain sites to affect autonomic nervous system outflow, energy balance, temperature regulation, sleep, arousal, neuroendocrine function, reproduction, and cardiopulmonary regulation. Compared to studies of other ANH functions, how the ANH regulates cardiopulmonary function is less understood. Importantly, the ANH exhibits structural and functional sexually dimorphic characteristics and contains numerous neuroactive substances and receptors including leptin, neuropeptide Y, glutamate, acetylcholine, endorphins, orexin, kisspeptin, insulin, Agouti-related protein, cocaine and amphetamine-regulated transcript, dopamine, somatostatin, components of renin-angiotensin system and gamma amino butyric acid that modulate physiological functions. Moreover, several clinically relevant disorders are associated with ANH ventilatory control dysfunction. This review highlights how ANH neurotransmitter systems and receptors modulate breathing differently in male and female rodents. Results highlight the significance of the ANH in cardiopulmonary regulation. The paucity of studies in this area that will hopefully spark investigations of sexually dimorphic ANH-modulation of breathing.
Collapse
Affiliation(s)
- Evelyn H Schlenker
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, 414 East Clark St., Vermillion, SD, 57069, United States.
| |
Collapse
|
23
|
Romano E, Cosentino L, Laviola G, De Filippis B. Genes and sex hormones interaction in neurodevelopmental disorders. Neurosci Biobehav Rev 2016; 67:9-24. [DOI: 10.1016/j.neubiorev.2016.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
|
24
|
A genome-wide search for epigenetically [corrected] regulated genes in zebra finch using MethylCap-seq and RNA-seq. Sci Rep 2016; 6:20957. [PMID: 26864856 PMCID: PMC4750092 DOI: 10.1038/srep20957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/14/2016] [Indexed: 01/20/2023] Open
Abstract
Learning and memory formation are known to require dynamic CpG (de)methylation and gene expression changes. Here, we aimed at establishing a genome-wide DNA methylation map of the zebra finch genome, a model organism in neuroscience, as well as identifying putatively epigenetically regulated genes. RNA- and MethylCap-seq experiments were performed on two zebra finch cell lines in presence or absence of 5-aza-2′-deoxycytidine induced demethylation. First, the MethylCap-seq methodology was validated in zebra finch by comparison with RRBS-generated data. To assess the influence of (variable) methylation on gene expression, RNA-seq experiments were performed as well. Comparison of RNA-seq and MethylCap-seq results showed that at least 357 of the 3,457 AZA-upregulated genes are putatively regulated by methylation in the promoter region, for which a pathway analysis showed remarkable enrichment for neurological networks. A subset of genes was validated using Exon Arrays, quantitative RT-PCR and CpG pyrosequencing on bisulfite-treated samples. To our knowledge, this study provides the first genome-wide DNA methylation map of the zebra finch genome as well as a comprehensive set of genes of which transcription is under putative methylation control.
Collapse
|
25
|
Abstract
Brain development is an organized, but constantly adaptive, process in which genetic and epigenetic signals allow neurons to differentiate, to migrate, and to develop correct connections. Gender specific prenatal sex hormone milieu participates in the dimorphic development of many neuronal networks. Environmental cues may interfere with these developmental programs, producing adverse outcomes. Bisphenol A (BPA), an estrogenic/antiandrogenic endocrine disruptor widely diffused in the environment, produces adverse effects at levels below the acceptable daily intake. This review analyzes the recent literature on the consequences of perinatal exposure to BPA environmental doses on the development of a dimorphic brain. The BPA interference with the development and function of the neuroendocrine hypothalamus and of the nuclei controlling energy balance, and with the hippocampal memory processing is also discussed. The detrimental action of BPA appears complex, involving different hormonal and epigenetic pathways activated, often in a dimorphic way, within clearcut susceptibility windows. To date, discrepancies in experimental approaches and in related outcomes make unfeasible to translate the available information into clear dose-response models for human risk assessment. Evaluation of BPA brain levels in relation to the appearance of adverse effects in future basic studies will certainly give better definition of the warning threshold for human health.
Collapse
Affiliation(s)
- P Negri-Cesi
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, INBB Research Unit, Milano, Italy
| |
Collapse
|
26
|
Kuhn C. Emergence of sex differences in the development of substance use and abuse during adolescence. Pharmacol Ther 2015; 153:55-78. [PMID: 26049025 DOI: 10.1016/j.pharmthera.2015.06.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 04/29/2015] [Indexed: 12/24/2022]
Abstract
Substance use and abuse begin during adolescence. Male and female adolescent humans initiate use at comparable rates, but males increase use faster. In adulthood, more men than women use and abuse addictive drugs. However, some women progress more rapidly from initiation of use to entry into treatment. In animal models, adolescent males and females consume addictive drugs similarly. However, reproductively mature females acquire self-administration faster, and in some models, escalate use more. Sex/gender differences exist in neurobiologic factors mediating both reinforcement (dopamine, opioids) and aversiveness (CRF, dynorphin), as well as intrinsic factors (personality, psychiatric co-morbidities) and extrinsic factors (history of abuse, environment especially peers and family) which influence the progression from initial use to abuse. Many of these important differences emerge during adolescence, and are moderated by sexual differentiation of the brain. Estradiol effects which enhance both dopaminergic and CRF-mediated processes contribute to the female vulnerability to substance use and abuse. Testosterone enhances impulsivity and sensation seeking in both males and females. Several protective factors in females also influence initiation and progression of substance use including hormonal changes of pregnancy as well as greater capacity for self-regulation and lower peak levels of impulsivity/sensation seeking. Same sex peers represent a risk factor more for males than females during adolescence, while romantic partners increase risk for women during this developmental epoch. In summary, biologic factors, psychiatric co-morbidities as well as personality and environment present sex/gender-specific risks as adolescents begin to initiate substance use.
Collapse
Affiliation(s)
- Cynthia Kuhn
- Department of Pharmacology and Cancer Biology, Box 3813, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
27
|
Bhandari RK, Deem SL, Holliday DK, Jandegian CM, Kassotis CD, Nagel SC, Tillitt DE, Vom Saal FS, Rosenfeld CS. Effects of the environmental estrogenic contaminants bisphenol A and 17α-ethinyl estradiol on sexual development and adult behaviors in aquatic wildlife species. Gen Comp Endocrinol 2015; 214:195-219. [PMID: 25277515 DOI: 10.1016/j.ygcen.2014.09.014] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/08/2014] [Accepted: 09/20/2014] [Indexed: 12/12/2022]
Abstract
Endocrine disrupting chemicals (EDCs), including the mass-produced component of plastics, bisphenol A (BPA) are widely prevalent in aquatic and terrestrial habitats. Many aquatic species, such as fish, amphibians, aquatic reptiles and mammals, are exposed daily to high concentrations of BPA and ethinyl estradiol (EE2), estrogen in birth control pills. In this review, we will predominantly focus on BPA and EE2, well-described estrogenic EDCs. First, the evidence that BPA and EE2 are detectable in almost all bodies of water will be discussed. We will consider how BPA affects sexual and neural development in these species, as these effects have been the best characterized across taxa. For instance, such chemicals have been in many cases reported to cause sex-reversal of males to females. Even if these chemicals do not overtly alter the gonadal sex, there are indications that several EDCs might demasculinize male-specific behaviors that are essential for attracting a mate. In so doing, these chemicals may reduce the likelihood that these males reproduce. If exposed males do reproduce, the concern is that they will then be passing on compromised genetic fitness to their offspring and transmitting potential transgenerational effects through their sperm epigenome. We will thus consider how diverse epigenetic changes might be a unifying mechanism of how BPA and EE2 disrupt several processes across species. Such changes might also serve as universal species diagnostic biomarkers of BPA and other EDCs exposure. Lastly, the evidence that estrogenic EDCs-induced effects in aquatic species might translate to humans will be considered.
Collapse
Affiliation(s)
- Ramji K Bhandari
- Biological Sciences, University of Missouri, Columbia, MO 65211, USA; Columbia Environmental Research Center, U.S. Geological Survey, Columbia, MO 65201, USA
| | - Sharon L Deem
- Institute for Conservation Medicine, Saint Louis Zoo, Saint Louis, MO 63110, USA; Veterinary Clinical Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Dawn K Holliday
- Department of Biology and Environmental Science, Westminster College, Fulton, MO 65251, USA; Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Caitlin M Jandegian
- Columbia Environmental Research Center, U.S. Geological Survey, Columbia, MO 65201, USA; Institute for Conservation Medicine, Saint Louis Zoo, Saint Louis, MO 63110, USA; Masters in Public Health Program, University of Missouri, Columbia, MO 65211, USA
| | | | - Susan C Nagel
- Biological Sciences, University of Missouri, Columbia, MO 65211, USA; Obstetrics, Gynecology, & Women's Health, University of Missouri, Columbia, MO 65211, USA
| | - Donald E Tillitt
- Columbia Environmental Research Center, U.S. Geological Survey, Columbia, MO 65201, USA
| | | | - Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Genetics Area Program Faculty Member, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
28
|
Matsuda KI. Epigenetic changes in the estrogen receptor α gene promoter: implications in sociosexual behaviors. Front Neurosci 2014; 8:344. [PMID: 25389384 PMCID: PMC4211403 DOI: 10.3389/fnins.2014.00344] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 10/09/2014] [Indexed: 11/23/2022] Open
Abstract
Estrogen action through estrogen receptor α (ERα) is involved in the control of sexual and social behaviors in adult mammals. Alteration of ERα gene activity mediated by epigenetic mechanisms, such as histone modifications and DNA methylation, in particular brain areas appears to be crucial for determining the extents of these behaviors between the sexes and among individuals within the same sex. This review provides a summary of the epigenetic changes in the ERα gene promoter that correlate with sociosexual behaviors.
Collapse
Affiliation(s)
- Ken Ichi Matsuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto, Japan
| |
Collapse
|
29
|
Tsukahara S, Kanaya M, Yamanouchi K. Neuroanatomy and sex differences of the lordosis-inhibiting system in the lateral septum. Front Neurosci 2014; 8:299. [PMID: 25278832 PMCID: PMC4166118 DOI: 10.3389/fnins.2014.00299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022] Open
Abstract
Female sexual behavior in rodents, termed lordosis, is controlled by facilitatory and inhibitory systems in the brain. It has been well demonstrated that a neural pathway from the ventromedial hypothalamic nucleus (VMN) to the midbrain central gray (MCG) is essential for facilitatory regulation of lordosis. The neural pathway from the arcuate nucleus to the VMN, via the medial preoptic nucleus, in female rats mediates transient suppression of lordosis, until female sexual receptivity is induced. In addition to this pathway, other regions are involved in inhibitory regulation of lordosis in female rats. The lordosis-inhibiting systems exist not only in the female brain but also in the male brain. The systems contribute to suppression of heterotypical sexual behavior in male rats, although they have the potential ability to display lordosis. The lateral septum (LS) exerts an inhibitory influence on lordosis in both female and male rats. This review focuses on the neuroanatomy and sex differences of the lordosis-inhibiting system in the LS. The LS functionally and anatomically links to the MCG to exert suppression of lordosis. Neurons of the intermediate part of the LS (LSi) serve as lordosis-inhibiting neurons and project axons to the MCG. The LSi-MCG neural connection is sexually dimorphic, and formation of the male-like LSi-MCG neural connection is affected by aromatized testosterone originating from the testes in the postnatal period. The sexually dimorphic LSi-MCG neural connection may reflect the morphological basis of sex differences in the inhibitory regulation of lordosis in rats.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Moeko Kanaya
- Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Korehito Yamanouchi
- Department of Human Behavior and Environment Sciences, Faculty of Human Sciences, Waseda University Saitama, Japan
| |
Collapse
|
30
|
Wall EH, Hewitt SC, Case LK, Lin CY, Korach KS, Teuscher C. The role of genetics in estrogen responses: a critical piece of an intricate puzzle. FASEB J 2014; 28:5042-54. [PMID: 25212221 DOI: 10.1096/fj.14-260307] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The estrogens are female sex hormones that are involved in a variety of physiological processes, including reproductive development and function, wound healing, and bone growth. They are mainly known for their roles in reproductive tissues--specifically, 17β-estradiol (E2), the primary estrogen, which is secreted by the ovaries and induces cellular proliferation and growth of the uterus and mammary glands. In addition to the role of estrogens in promoting tissue growth and development during normal physiological states, they have a well-established role in determining susceptibility to disease, particularly cancer, in reproductive tissues. The responsiveness of various tissues to estrogen is genetically controlled, with marked quantitative variation observed across multiple species, including humans. This variation presents both researchers and clinicians with a veritable physiological puzzle, the pieces of which--many of them unknown--are complex and difficult to fit together. Although genetics is known to play a major role in determining sensitivity to estrogens, there are other factors, including parent of origin and the maternal environment, that are intimately linked to heritable phenotypes but do not represent genotype, per se. The objectives of this review article were to summarize the current knowledge of the role of genotype, and uterine and neonatal environments, in phenotypic variation in the response to estrogens; to discuss recent findings and the potential mechanisms involved; and to highlight exciting research opportunities for the future.
Collapse
Affiliation(s)
- Emma H Wall
- Department of Medicine and Pathology, University of Vermont, Burlington Vermont, USA
| | - Sylvia C Hewitt
- Receptor Biology, National Institute of Environmental Health Science, U.S. National Institutes of Health, Research Triangle Park, North Carolina, USA; and
| | - Laure K Case
- Department of Medicine and Pathology, University of Vermont, Burlington Vermont, USA
| | - Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Kenneth S Korach
- Receptor Biology, National Institute of Environmental Health Science, U.S. National Institutes of Health, Research Triangle Park, North Carolina, USA; and
| | - Cory Teuscher
- Department of Medicine and Pathology, University of Vermont, Burlington Vermont, USA;
| |
Collapse
|
31
|
Handa RJ, McGivern RF. Steroid Hormones, Receptors, and Perceptual and Cognitive Sex Differences in the Visual System. Curr Eye Res 2014; 40:110-27. [DOI: 10.3109/02713683.2014.952826] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
32
|
Berretti R, Santoru F, Locci A, Sogliano C, Calza A, Choleris E, Porcu P, Concas A. Neonatal exposure to estradiol decreases hypothalamic allopregnanolone concentrations and alters agonistic and sexual but not affective behavior in adult female rats. Horm Behav 2014; 65:142-53. [PMID: 24368289 DOI: 10.1016/j.yhbeh.2013.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/03/2013] [Accepted: 12/15/2013] [Indexed: 01/22/2023]
Abstract
Exposure of developing female rats to estradiol during the perinatal period induced long-lasting dysregulation of gonadal axis and decreased cerebrocortical and plasma concentrations of allopregnanolone. We have now examined the effects of neonatal estradiol administration in female rats on hypothalamic allopregnanolone concentrations and on exploratory, affective, agonistic and sexual behaviors as well as social learning. A single administration of β-estradiol 3-benzoate (EB, 10μg) on the day of birth resulted in a delay of vaginal opening, acyclicity and ovarian failure. These alterations were associated with a significant decrease in the concentrations of allopregnanolone in the hypothalamus at 21 and 60days, but not at 7days, after birth. Neonatal administration of EB also increased agonistic behaviors in adult rats, such as dominant behaviors and following of an ovariectomized intruder, while living attacks unaffected. EB-treated rats showed also an increase in anogenital investigation, associated with a drastic reduction in spontaneous and induced female sexual behaviors (receptivity and proceptivity). In contrast, neonatal administration of EB did not affect locomotor activity, anxiety- and mood-related behaviors, the social transmission of flavor preferences, and seizures sensitivity. These effects of estradiol suggest that it plays a major role in regulation of both the abundance of allopregnanolone and the expression of agonistic and sexual behaviors, while failing to influence affective behaviors and social learning. Thus, the pronounced and persistent decrease in hypothalamic allopregnanolone concentration may be related to the manifestation of agonistic and sexual behaviors.
Collapse
Affiliation(s)
- R Berretti
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - F Santoru
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - A Locci
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - C Sogliano
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - A Calza
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - E Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - P Porcu
- Institute of Neuroscience, National Research Council of Italy (CNR), Cagliari, Italy
| | - A Concas
- Department of Life and Environment Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; Institute of Neuroscience, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
33
|
Peruffo A, Giacomello M, Montelli S, Panin M, Cozzi B. Expression profile of the pore-forming subunits α1A and α1D in the foetal bovine hypothalamus: a mammal with a long gestation. Neurosci Lett 2013; 556:124-8. [PMID: 24148303 DOI: 10.1016/j.neulet.2013.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 01/27/2023]
Abstract
This study describes the expression of the voltage operated calcium channels (VOCCs) subunits α1A (typical of the P/Q family) and α1D (of the L family) in the bovine hypothalamus. The expression of both P/Q and L families has been characterized in the brain of adult mammals. However, their distribution and expression during foetal neuronal differentiation have not yet been determined. The expression profile of the α1A and α1D pore-forming subunits was investigated during four embryonic stages in bovine foetuses. Our data suggest that the expressions of α1A and α1D are correlated during development, with an increase only in males that peaks on the last period of gestation. Bovine male hypothalami showed significantly higher α1A and α1D expression values in comparison to female ones during the whole developmental period. In the females, the expression profiles of both genes were constant during all the developmental time. Immunohistochemical studies confirmed the presence of the α1A and α1D protein subunits in foetal hypothalamic neurones starting from the third foetal stage. Our data provide new information on the hypothalamic expression of α1A and α1D subunits during development in a mammal with a long gestation period and a large and convoluted brain.
Collapse
Affiliation(s)
- A Peruffo
- Department of Comparative Biomedicine and Food Science, University of Padova, Vialedell'Università 16, 35020 Legnaro, PD, Italy.
| | | | | | | | | |
Collapse
|
34
|
Microarray analysis of perinatal-estrogen-induced changes in gene expression related to brain sexual differentiation in mice. PLoS One 2013; 8:e79437. [PMID: 24223949 PMCID: PMC3817063 DOI: 10.1371/journal.pone.0079437] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 09/23/2013] [Indexed: 11/19/2022] Open
Abstract
Sexual dimorphism of the behaviors or physiological functions in mammals is mainly due to the sex difference of the brain. A number of studies have suggested that the brain is masculinized or defeminized by estradiol converted from testicular androgens in perinatal period in rodents. However, the mechanisms of estrogen action resulting in masculinization/defeminization of the brain have not been clarified yet. The large-scale analysis with microarray in the present study is an attempt to obtain the candidate gene(s) mediating the perinatal estrogen effect causing the brain sexual differentiation. Female mice were injected with estradiol benzoate (EB) or vehicle on the day of birth, and the hypothalamus was collected at either 1, 3, 6, 12, or 24 h after the EB injection. More than one hundred genes down-regulated by the EB treatment in a biphasic manner peaked at 3 h and 12-24 h after the EB treatment, while forty to seventy genes were constantly up-regulated after it. Twelve genes, including Ptgds, Hcrt, Tmed2, Klc1, and Nedd4, whose mRNA expressions were down-regulated by the neonatal EB treatment, were chosen for further examination by semiquantitative RT-PCR in the hypothalamus of perinatal intact male and female mice. We selected the genes based on the known profiles of their potential roles in brain development. mRNA expression levels of Ptgds, Hcrt, Tmed2, and Nedd4 were significantly lower in male mice than females at the day of birth, suggesting that the genes are down-regulated by estrogen converted from testicular androgen in perinatal male mice. Some genes, such as Ptgds encoding prostaglandin D2 production enzyme and Hcrt encording orexin, have been reported to have a role in neuroprotection. Thus, Ptgds and Hcrt could be possible candidate genes, which may mediate the effect of perinatal estrogen responsible for brain sexual differentiation.
Collapse
|
35
|
Rice WR, Friberg U, Gavrilets S. Homosexuality via canalized sexual development: a testing protocol for a new epigenetic model. Bioessays 2013; 35:764-70. [PMID: 23868698 PMCID: PMC3840696 DOI: 10.1002/bies.201300033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We recently synthesized and reinterpreted published studies to advance an epigenetic model for the development of homosexuality (HS). The model is based on epigenetic marks laid down in response to the XX vs. XY karyotype in embryonic stem cells. These marks boost sensitivity to testosterone in XY fetuses and lower it in XX fetuses, thereby canalizing sexual development. Our model predicts that a subset of these canalizing epigenetic marks stochastically carry over across generations and lead to mosaicism for sexual development in opposite-sex offspring--the homosexual phenotype being one such outcome. Here, we begin by outlining why HS has been under-appreciated as a commonplace phenomenon in nature, and how this trend is currently being reversed in the field of neurobiology. We next briefly describe our epigenetic model of HS, develop a set of predictions, and describe how epigenetic profiles of human stem cells can provide for a strong test of the model.
Collapse
Affiliation(s)
- William R Rice
- Department of Ecology, Evolution & Marine Biology, University of California, Santa Barbara, CA, USA.
| | | | | |
Collapse
|
36
|
Puberty dysregulation and increased risk of disease in adult life: possible modes of action. Reprod Toxicol 2013; 44:15-22. [PMID: 23791931 DOI: 10.1016/j.reprotox.2013.06.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 03/12/2013] [Accepted: 06/05/2013] [Indexed: 11/22/2022]
Abstract
Puberty is the developmental window when the final maturation of body systems is orchestrated by hormones; lifelong sex-related differences and capacity to interact with the environment are defined during this life stage. Increased incidence in a number of chronic, multifactorial diseases could be related to environmental exposures during puberty: however, insight on the susceptibility of the peripubertal period is still limited. The estrogen/androgen balance is a crucial axis in harmonizing the whole pubertal development, pointing out the significance of exposures to endocrine disruptors. Besides the reproductive system, endocrine-related perturbations may affect the maturation of skeleton, adipose tissues, brain, immune system, as well as cancer predisposition. Thus, risk assessment of environmental stressors should duly consider specific aspects of the pubertal window. Besides endocrine-related mechanisms, suggested research priorities include signaling molecules (e.g., kisspeptins, dopamine) as xenobiotic targets and disturbances of specific pubertal methylation processes potentially involved in neurobehavioral disorders and cancer risk in adulthood.
Collapse
|
37
|
Matsuda KI, Yanagisawa M, Sano K, Ochiai I, Musatov S, Okoshi K, Tsukahara S, Ogawa S, Kawata M. Visualisation and characterisation of oestrogen receptor α-positive neurons expressing green fluorescent protein under the control of the oestrogen receptor α promoter. Eur J Neurosci 2013; 38:2242-9. [PMID: 23601009 DOI: 10.1111/ejn.12227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 03/21/2013] [Indexed: 12/20/2022]
Abstract
Oestrogen receptor (ER)α plays important roles in the development and function of various neuronal systems through activation by its ligands, oestrogens. To visualise ERα-positive neurons, we generated transgenic (tg) mice expressing green fluorescent protein (GFP) under the control of the ERα promoter. In three independent tg lines, GFP-positive neurons were observed in areas previously reported to express ERα mRNA, including the lateral septum, bed nucleus of the stria terminalis, medial preoptic nucleus (MPO), hypothalamus, and amygdala. In these areas, GFP signals mostly overlapped with ERα immunoreactivity. GFP fluorescence was seen in neurites and cell bodies of neurons. In addition, the network and detailed structure of neurites were visible in dissociated and slice cultures of hypothalamic neurons. We examined the effect of oestrogen deprivation by ovariectomy on the structure of the GFP-positive neurons. The area of ERα-positive cell bodies in the bed nucleus of the stria terminalis and MPO was measured by capturing the GFP signal and was found to be significantly smaller in ovariectomy mice than in control mice. When neurons in the MPO were infected with an adeno-associated virus that expressed small hairpin RNA targeting the ERα gene, an apparent induction of GFP was observed in this area, suggesting a negative feedback mechanism in which ERα controls expression of the ERα gene itself. Thus, the ERα promoter-GFP tg mice will be useful to analyse the development and plastic changes of the structure of ERα-expressing neurons and oestrogen and its receptor-mediated neuronal responses.
Collapse
Affiliation(s)
- Ken Ichi Matsuda
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, 465 Kawaramachi Hirokoji, Kamigyoku, Kyoto 602-8566, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Walker DM, Goetz BM, Gore AC. Dynamic postnatal developmental and sex-specific neuroendocrine effects of prenatal polychlorinated biphenyls in rats. Mol Endocrinol 2013; 28:99-115. [PMID: 24284824 DOI: 10.1210/me.2013-1270] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gestational exposures to estrogenic compounds, both endogenous hormones and exogenous endocrine-disrupting chemicals (EDCs), have long-term effects on reproductive physiology and behavior. We tested the hypothesis that prenatal treatment of rats with low doses of Aroclor 1221 (A1221), a weakly estrogenic polychlorinated biphenyl mix previously used in industry, or estradiol benzoate (EB), alters development of the hypothalamus in a sexually dimorphic manner and subsequently perturbs reproductive function. Pregnant Sprague-Dawley rats were injected on embryonic days 16 and 18 with vehicle (dimethylsulfoxide), A1221 (1 mg/kg), or EB (50 μg/kg). Developmental milestones were monitored, and on postnatal days 15, 30, 45, and 90, 1 male and 1 female per litter were euthanized. Because of their key roles in the mediation of steroid actions on reproductive function, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC) were punched for a low-density quantitative PCR array of 48 neuroendocrine genes and analysis of DNA methylation of a subset of genes. Gestational exposure to A1221 or EB delayed the timing of puberty in males and disrupted estrous cyclicity in females. In the AVPV, 28 genes were affected by treatment in a developmental stage-specific manner, mostly in females, which exhibited a masculinized expression profile. This included 2 clock genes, Per2 and Arntl, implicating circadian circuits as being vulnerable to endocrine disruption. DNA methylation analysis of 2 genes, Per2 and Ar, showed no effect of EDCs and suggested alternative mechanisms for the altered mRNA levels. In the ARC, 12 genes were affected by treatment, mostly in males, again with dynamic developmental changes. Bionetwork analysis of relationships among genes, hormones, and physiological markers showed sexually dimorphic effects of estrogenic EDC exposures, with the female AVPV and the male ARC being most vulnerable, and provided novel relationships among hypothalamic genes and postnatal reproductive maturation.
Collapse
Affiliation(s)
- Deena M Walker
- The Institute for Neuroscience (D.M.W., A.C.G.), Center for Computational Biology and Bioinformatics (B.M.G.), Division of Pharmacology and Toxicology (A.C.G.), College of Pharmacy, and Institute for Cell and Molecular Biology (A.C.G.), The University of Texas at Austin, Austin, Texas 78712
| | | | | |
Collapse
|