1
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. eLife 2024; 12:RP90893. [PMID: 39446467 PMCID: PMC11501206 DOI: 10.7554/elife.90893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2Baxfl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild-type females or males, which is notable because loss of these two hilar cell types is implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, University of SaskatchewanSaskatoonCanada
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Neuroscience & Physiology, Psychiatry, and the Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
2
|
Jain S, LaFrancois JJ, Gerencer K, Botterill JJ, Kennedy M, Criscuolo C, Scharfman HE. Increasing adult-born neurons protects mice from epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.08.548217. [PMID: 37502909 PMCID: PMC10369878 DOI: 10.1101/2023.07.08.548217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Neurogenesis occurs in the adult brain in the hippocampal dentate gyrus, an area that contains neurons which are vulnerable to insults and injury, such as severe seizures. Previous studies showed that increasing adult neurogenesis reduced neuronal damage after these seizures. Because the damage typically is followed by chronic life-long seizures (epilepsy), we asked if increasing adult-born neurons would prevent epilepsy. Adult-born neurons were selectively increased by deleting the pro-apoptotic gene Bax from Nestin-expressing progenitors. Tamoxifen was administered at 6 weeks of age to conditionally delete Bax in Nestin-CreERT2 Bax fl/fl mice. Six weeks after tamoxifen administration, severe seizures (status epilepticus; SE) were induced by injection of the convulsant pilocarpine. After mice developed epilepsy, seizure frequency was quantified for 3 weeks. Mice with increased adult-born neurons exhibited fewer chronic seizures. Postictal depression was reduced also. These results were primarily in female mice, possibly because they were the more affected by Bax deletion than males, consistent with sex differences in Bax. The female mice with enhanced adult-born neurons also showed less neuronal loss of hilar mossy cells and hilar somatostatin-expressing neurons than wild type females or males, which is notable because these two hilar cell types are implicated in epileptogenesis. The results suggest that selective Bax deletion to increase adult-born neurons can reduce experimental epilepsy, and the effect shows a striking sex difference. The results are surprising in light of past studies showing that suppressing adult-born neurons can also reduce chronic seizures.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, The University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Meghan Kennedy
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Chiara Criscuolo
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016
- Departments of Neuroscience & Physiology, Psychiatry, and the New York University, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
3
|
Roggenbuck EC, Hall EA, Hanson IB, Roby AA, Zhang KK, Alkatib KA, Carter JA, Clewner JE, Gelfius AL, Gong S, Gordon FR, Iseler JN, Kotapati S, Li M, Maysun A, McCormick EO, Rastogi G, Sengupta S, Uzoma CU, Wolkov MA, Clowney EJ. Let's talk about sex: Mechanisms of neural sexual differentiation in Bilateria. WIREs Mech Dis 2024; 16:e1636. [PMID: 38185860 DOI: 10.1002/wsbm.1636] [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: 05/09/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 01/09/2024]
Abstract
In multicellular organisms, sexed gonads have evolved that facilitate release of sperm versus eggs, and bilaterian animals purposefully combine their gametes via mating behaviors. Distinct neural circuits have evolved that control these physically different mating events for animals producing eggs from ovaries versus sperm from testis. In this review, we will describe the developmental mechanisms that sexually differentiate neural circuits across three major clades of bilaterian animals-Ecdysozoa, Deuterosomia, and Lophotrochozoa. While many of the mechanisms inducing somatic and neuronal sex differentiation across these diverse organisms are clade-specific rather than evolutionarily conserved, we develop a common framework for considering the developmental logic of these events and the types of neuronal differences that produce sex-differentiated behaviors. This article is categorized under: Congenital Diseases > Stem Cells and Development Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Emma C Roggenbuck
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Elijah A Hall
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Isabel B Hanson
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Alyssa A Roby
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine K Zhang
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Kyle A Alkatib
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph A Carter
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jarred E Clewner
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna L Gelfius
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Shiyuan Gong
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Finley R Gordon
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jolene N Iseler
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Samhita Kotapati
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Marilyn Li
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Areeba Maysun
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Elise O McCormick
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Geetanjali Rastogi
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Srijani Sengupta
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Chantal U Uzoma
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - Madison A Wolkov
- MCDB 464 - Cellular Diversity: Sex Differentiation of the Brain, University of Michigan, Ann Arbor, Michigan, USA
| | - E Josephine Clowney
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Neuroscience Institute Affiliate, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
McCarthy MM. Neural Control of Sexually Dimorphic Social Behavior: Connecting Development to Adulthood. Annu Rev Neurosci 2023; 46:321-339. [PMID: 37001242 DOI: 10.1146/annurev-neuro-121522-110856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Rapid advances in the neural control of social behavior highlight the role of interconnected nodes engaged in differential information processing to generate behavior. Many innate social behaviors are essential to reproductive fitness and therefore fundamentally different in males and females. Programming these differences occurs early in development in mammals, following gonadal differentiation and copious androgen production by the fetal testis during a critical period. Early-life programming of social behavior and its adult manifestation are separate but yoked processes, yet how they are linked is unknown. This review seeks to highlight that gap by identifying four core mechanisms (epigenetics, cell death, circuit formation, and adult hormonal modulation) that could connect developmental changes to the adult behaviors of mating and aggression. We further propose that a unique social behavior, adolescent play, bridges the preweaning to the postpubertal brain by engaging the same neural networks underpinning adult reproductive and aggressive behaviors.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
5
|
Park CJ, Minabe S, Hess RA, Lin PCP, Zhou S, Bashir ST, Barakat R, Gal A, Ko CJ. Single neonatal estrogen implant sterilizes female animals by decreasing hypothalamic KISS1 expression. Sci Rep 2023; 13:9627. [PMID: 37316510 DOI: 10.1038/s41598-023-36727-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023] Open
Abstract
Reproductive sterilization by surgical gonadectomy is strongly advocated to help manage animal populations, especially domesticated pets, and to prevent reproductive behaviors and diseases. This study explored the use of a single-injection method to induce sterility in female animals as an alternative to surgical ovariohysterectomy. The idea was based on our recent finding that repetitive daily injection of estrogen into neonatal rats disrupted hypothalamic expression of Kisspeptin (KISS1), the neuropeptide that triggers and regulates pulsatile secretion of GnRH. Neonatal female rats were dosed with estradiol benzoate (EB) either by daily injections for 11 days or by subcutaneous implantation of an EB-containing silicone capsule designed to release EB over 2-3 weeks. Rats treated by either method did not exhibit estrous cyclicity, were anovulatory, and became infertile. The EB-treated rats had fewer hypothalamic Kisspeptin neurons, but the GnRH-LH axis remained responsive to Kisspeptin stimulation. Because it would be desirable to use a biodegradable carrier that is also easier to handle, an injectable EB carrier was developed from PLGA microspheres to provide pharmacokinetics comparable to the EB-containing silicone capsule. A single neonatal injection of EB-microspheres at an equivalent dosage resulted in sterility in the female rat. In neonatal female Beagle dogs, implantation of an EB-containing silicone capsule also reduced ovarian follicle development and significantly inhibited KISS1 expression in the hypothalamus. None of the treatments produced any concerning health effects, other than infertility. Therefore, further development of this technology for sterilization in domestic female animals, such as dogs and cats is worthy of investigation.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Epivara, Inc, Champaign, IL, 61820, USA
| | - Shiori Minabe
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Epivara, Inc, Champaign, IL, 61820, USA
| | - Po-Ching Patrick Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | | | - Shah Tauseef Bashir
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Radwa Barakat
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Benha University, Qalyubia, 13518, Egypt
| | - Arnon Gal
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - CheMyong Jay Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
- Epivara, Inc, Champaign, IL, 61820, USA.
| |
Collapse
|
6
|
Cortes LR, Forger NG. DNA methylation and demethylation shape sexual differentiation of neurochemical phenotype. Horm Behav 2023; 151:105349. [PMID: 37001316 PMCID: PMC10133097 DOI: 10.1016/j.yhbeh.2023.105349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/24/2023] [Accepted: 03/13/2023] [Indexed: 04/28/2023]
Abstract
Some of the best-studied neural sex differences depend on differential cell death in males and females, but other sex differences persist even if cell death is prevented. These include sex differences in neurochemical phenotype (i.e., stable patterns of gene expression). Work in our laboratory over the last several years has tested the hypothesis that sex differences in DNA methylation early in life underlie sexual differentiation of neuronal phenotype. We have shown that 1) expression of enzymes that place or remove DNA methylation marks is greatest during the first week of life in the mouse brain and overlaps with the perinatal critical period of sexual differentiation; 2) a transient inhibition of DNA methylation during neonatal life abolishes several sex differences in cell phenotype in the mouse hypothalamus; 3) both DNA methylation and de-methylation contribute to the development of neural sex differences; and 4) the effects of DNA methylation and de-methylation are brain region- and cell type-specific.
Collapse
Affiliation(s)
- L R Cortes
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - N G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Smiley JF, Bleiwas C, Marino BM, Vaddi P, Canals-Baker S, Wilson DA, Saito M. Estimates of total neuron number show that neonatal ethanol causes immediate and lasting neuron loss in cortical and subcortical areas. Front Neurosci 2023; 17:1186529. [PMID: 37205048 PMCID: PMC10185770 DOI: 10.3389/fnins.2023.1186529] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
In neonatal brain development there is a period of normal apoptotic cell death that regulates adult neuron number. At approximately the same period, ethanol exposure can cause a dramatic spike in apoptotic cell death. While ethanol-induced apoptosis has been shown to reduce adult neuron number, questions remain about the regional selectivity of the ethanol effect, and whether the brain might have some capacity to overcome the initial neuron loss. The present study used stereological cell counting to compare cumulative neuron loss 8 h after postnatal day 7 (P7) ethanol treatment to that of animals left to mature to adulthood (P70). Across several brain regions we found that the reduction of total neuron number after 8 h was as large as that of adult animals. Comparison between regions revealed that some areas are more vulnerable, with neuron loss in the anterior thalamic nuclei > the medial septum/vertical diagonal band, dorsal subiculum, and dorsal lateral geniculate nucleus > the mammillary bodies and cingulate cortex > whole neocortex. In contrast to estimates of total neuron number, estimates of apoptotic cell number in Nissl-stained sections at 8 h after ethanol treatment provided a less reliable predictor of adult neuron loss. The findings show that ethanol-induced neonatal apoptosis often causes immediate neuron deficits that persist in adulthood, and furthermore suggests that the brain may have limited capacity to compensate for ethanol-induced neuron loss.
Collapse
Affiliation(s)
- John F. Smiley
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, School of Medicine, New York University, New York, NY, United States
- *Correspondence: John F. Smiley,
| | - Cynthia Bleiwas
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Brandon M. Marino
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Prerana Vaddi
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | | | - Donald A. Wilson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, School of Medicine, New York University, New York, NY, United States
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, United States
| | - Mariko Saito
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, School of Medicine, New York University, New York, NY, United States
| |
Collapse
|
8
|
Edwards PD, Toor I, Holmes MM. The Curious Case of the Naked Mole-Rat: How Extreme Social and Reproductive Adaptations Might Influence Sex Differences in the Brain. Curr Top Behav Neurosci 2023; 62:47-70. [PMID: 35301704 DOI: 10.1007/7854_2022_310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Research in the neurobiology of sex differences is inherently influenced by the study species that are used. Some traditional animal research models, such as rats and mice, show certain sex differences in the brain that have been foundational to neurobiological research. However, subsequent work has demonstrated that these differences are not always generalizable, especially to species with different social structures and sex-associated roles or behaviors. One such example is the naked mole-rat (Heterocephalus glaber), which has an unusual social structure among mammals. Naked mole-rats live in large groups where reproduction is restricted to a dominant female, called the "queen," and often only one breeding male. All other animals in the group, the "subordinates," are socially suppressed from reproduction and remain in a prepubescent state as adults, unless they are removed from the presence of the queen. These subordinates show little to no sex differences in external morphology, neural morphology, or behavior. However, there are a suite of neurobiological differences between subordinate and breeding naked mole-rats. After naked mole-rats attain breeding status, many of the classically sexually differentiated brain regions increase in volume (paraventricular nucleus, medial amygdala, bed nucleus of the stria terminalis). There are additionally social status differences in sex hormone receptor expression in the brain, as well as other changes in gene expression, some of which also show sex differences - though not always in the predicted direction based on other rodent studies. Data from naked mole-rats show that it is critical to consider the evolved social structure of a species when studying sex differences in the brain.
Collapse
Affiliation(s)
- Phoebe D Edwards
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Ilapreet Toor
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| |
Collapse
|
9
|
Abstract
Across vertebrate species, gonadal hormones coordinate physiology with behavior to facilitate social interactions essential for reproduction and survival. In adulthood, these hormones activate neural circuits that regulate behaviors presenting differently in females and males, such as parenting and territorial aggression. Yet long before sex-typical behaviors emerge at puberty, transient hormone production during sensitive periods of neurodevelopment establish the circuits upon which adult hormones act. How transitory waves of early-life hormone signaling exert lasting effects on the brain remains a central question. Here we discuss how perinatal estradiol signaling organizes cellular and molecular sex differences in the rodent brain. We review classic anatomic studies revealing sex differences in cell number, volume, and neuronal projections, and consider how single-cell sequencing methods enable distinction between sex-biased cell-type abundance and gene expression. Finally, we highlight the recent discovery of a gene regulatory program activated by estrogen receptor α (ERα) following the perinatal hormone surge. A subset of this program displays sustained sex-biased gene expression and chromatin accessibility throughout the postnatal sensitive period, demonstrating a bona fide epigenetic mechanism. We propose that ERα-expressing neurons throughout the social behavior network use similar gene regulatory programs to coordinate brain sexual differentiation.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
10
|
Laue HE, Karagas MR, Coker MO, Bellinger DC, Baker ER, Korrick SA, Madan JC. Sex-specific relationships of the infant microbiome and early-childhood behavioral outcomes. Pediatr Res 2022; 92:580-591. [PMID: 34732816 PMCID: PMC9065210 DOI: 10.1038/s41390-021-01785-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND A link between the gut microbiome and behavior is hypothesized, but most previous studies are cross-sectional or in animal models. The modifying role of host sex is poorly characterized. We aimed to identify sex-specific prospective associations between the early-life gut microbiome and preschool-age neurobehavior. METHODS In a prospective cohort, gut microbiome diversity and taxa were estimated with 16S rRNA sequencing at 6 weeks, 1 year, and 2 years. Species and gene pathways were inferred from metagenomic sequencing at 6 weeks and 1 year. When subjects were 3 years old, parents completed the Behavioral Assessment System for Children, second edition (BASC-2). A total of 260 children contributed 523 16S rRNA and 234 metagenomics samples to analysis. Models adjusted for sociodemographic characteristics. RESULTS Higher diversity at 6 weeks was associated with better internalizing problems among boys, but not girls [βBoys = -1.86 points/SD Shannon diversity; 95% CI (-3.29, -0.42), pBoys = 0.01, βGirls = 0.22 (-1.43, 1.87), pGirls = 0.8, pinteraction = 0.06]. Among other taxa-specific associations, Bifidobacterium at 6 weeks was associated with Adaptive Skills scores in a sex-specific manner. We observed relationships between functional features and BASC-2 scores, including vitamin B6 biosynthesis pathways and better Depression scores. CONCLUSIONS This study advances our understanding of microbe-host interactions with implications for childhood behavioral health. IMPACT This is one of the first studies to examine the early-life microbiome and neurobehavior, and the first to examine prospective sex-specific associations. Infant and early-childhood microbiomes relate to neurobehavior including anxiety, depression, hyperactivity, and social behaviors in a time- and sex-specific manner. Our findings suggest future studies should evaluate whether host sex impacts the relationship between the gut microbiome and behavioral health outcomes.
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA.
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
| | - Modupe O Coker
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
- School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - David C Bellinger
- Department of Neurology, Harvard Medical School and Boston Children's Hospital, Boston, MA, USA
| | - Emily R Baker
- Department of Obstetrics and Gynecology, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Susan A Korrick
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Juliette C Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
- Departments of Pediatrics and Psychiatry, Children's Hospital at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
11
|
Paletta P, Bass N, Aspesi D, Choleris E. Sex Differences in Social Cognition. Curr Top Behav Neurosci 2022; 62:207-234. [PMID: 35604571 DOI: 10.1007/7854_2022_325] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In this review we explore the sex differences underlying various types of social cognition. Particular focus will be placed on the behaviors of social recognition, social learning, and aggression. Known similarities and differences between sexes in the expressions of these behaviors and the known brain regions where these behaviors are mediated are discussed. The role that the sex hormones (estrogens and androgens) have as well as possible interactions with other neurochemicals, such as oxytocin, vasopressin, and serotonin is reviewed as well. Finally, implications about these findings on the mediation of social cognition are mediated and the sex differences related to humans are considered.
Collapse
Affiliation(s)
- Pietro Paletta
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Noah Bass
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Dario Aspesi
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
12
|
Lee EB, Dilower I, Marsh CA, Wolfe MW, Masumi S, Upadhyaya S, Rumi MAK. Sexual Dimorphism in Kisspeptin Signaling. Cells 2022; 11:1146. [PMID: 35406710 PMCID: PMC8997554 DOI: 10.3390/cells11071146] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin (KP) and kisspeptin receptor (KPR) are essential for the onset of puberty, development of gonads, and maintenance of gonadal function in both males and females. Hypothalamic KPs and KPR display a high degree of sexual dimorphism in expression and function. KPs act on KPR in gonadotropin releasing hormone (GnRH) neurons and induce distinct patterns of GnRH secretion in males and females. GnRH acts on the anterior pituitary to secrete gonadotropins, which are required for steroidogenesis and gametogenesis in testes and ovaries. Gonadal steroid hormones in turn regulate the KP neurons. Gonadal hormones inhibit the KP neurons within the arcuate nucleus and generate pulsatile GnRH mediated gonadotropin (GPN) secretion in both sexes. However, the numbers of KP neurons in the anteroventral periventricular nucleus and preoptic area are greater in females, which release a large amount of KPs in response to a high estrogen level and induce the preovulatory GPN surge. In addition to the hypothalamus, KPs and KPR are also expressed in various extrahypothalamic tissues including the liver, pancreas, fat, and gonads. There is a remarkable difference in circulating KP levels between males and females. An increased level of KPs in females can be linked to increased numbers of KP neurons in female hypothalamus and more KP production in the ovaries and adipose tissues. Although the sexually dimorphic features are well characterized for hypothalamic KPs, very little is known about the extrahypothalamic KPs. This review article summarizes current knowledge regarding the sexual dimorphism in hypothalamic as well as extrahypothalamic KP and KPR system in primates and rodents.
Collapse
Affiliation(s)
- Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Iman Dilower
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Courtney A. Marsh
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (C.A.M.); (M.W.W.)
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Sameer Upadhyaya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| | - Mohammad A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (I.D.); (S.M.); (S.U.)
| |
Collapse
|
13
|
Bennett C, Green J, Ciancio M, Goral J, Pitstick L, Pytynia M, Meyer A, Kwatra N, Jadavji NM. Dietary folic acid deficiency impacts hippocampal morphology and cortical acetylcholine metabolism in adult male and female mice. Nutr Neurosci 2021; 25:2057-2065. [PMID: 34042561 DOI: 10.1080/1028415x.2021.1932242] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE One-carbon (1C) metabolism is a metabolic network that integrates nutritional signals with biosynthesis, redox homeostasis, and epigenetics. There are sex differences in hepatic 1C metabolism, however, it is unclear whether sex differences in 1C impact the brain. The aim of this study was to investigate if sex modulates the effects of dietary folic acid deficiency, the main component of 1C, in brain tissue using a mouse model. METHODS Male and female C57Bl/6J mice were placed on a folic acid deficient (FD) or control diet (CD) at six weeks until six months of aged. After which brain tissue and serum were collected for analysis. In brain tissue, hippocampal volume, morphology, and apoptosis as well as cortical acetylcholine metabolism were measured. RESULTS Male and female FD mice had reduced serum levels of folate. Both males and females maintained on a FD showed a decrease in the thickness of the hippocampal CA1-CA3 region. Interestingly, there was a sex difference in the levels of active caspase-3 within the CA3 region of the hippocampus. In cortical tissue, there were increased levels of neuronal ChAT and reduced levels of AChE in FD females and male mice. CONCLUSIONS The results indicated that FD impacts hippocampal morphology and cortical neuronal acetylcholine metabolism. The data from our study indicate that there was only one sex difference and that was in hippocampal apoptosis. Our study provides little evidence that sex modulates the effects of dietary folate deficiency on hippocampal morphology and cortical neuronal acetylcholine metabolism.
Collapse
Affiliation(s)
- Calli Bennett
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ, USA.,College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Jacalyn Green
- Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Mae Ciancio
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Joanna Goral
- Department of Anatomy, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Lenore Pitstick
- Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Matthew Pytynia
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Alice Meyer
- Department of Anatomy, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Neha Kwatra
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ, USA.,College of Dental Medicine, Midwestern University, Glendale, AZ, USA
| | - Nafisa M Jadavji
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ, USA.,College of Veterinary Medicine, Midwestern University, Glendale, AZ, USA.,Department of Neuroscience, Carleton University, Ottawa, Canada
| |
Collapse
|
14
|
Raznahan A, Disteche CM. X-chromosome regulation and sex differences in brain anatomy. Neurosci Biobehav Rev 2021; 120:28-47. [PMID: 33171144 PMCID: PMC7855816 DOI: 10.1016/j.neubiorev.2020.10.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 01/08/2023]
Abstract
Humans show reproducible sex-differences in cognition and psychopathology that may be contributed to by influences of gonadal sex-steroids and/or sex-chromosomes on regional brain development. Gonadal sex-steroids are well known to play a major role in sexual differentiation of the vertebrate brain, but far less is known regarding the role of sex-chromosomes. Our review focuses on this latter issue by bridging together two literatures that have to date been largely disconnected. We first consider "bottom-up" genetic and molecular studies focused on sex-chromosome gene content and regulation. This literature nominates specific sex-chromosome genes that could drive developmental sex-differences by virtue of their sex-biased expression and their functions within the brain. We then consider the complementary "top down" view, from magnetic resonance imaging studies that map sex- and sex chromosome effects on regional brain anatomy, and link these maps to regional gene-expression within the brain. By connecting these top-down and bottom-up approaches, we emphasize the potential role of X-linked genes in driving sex-biased brain development and outline key goals for future work in this field.
Collapse
Affiliation(s)
- Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, 20892, USA.
| | - Christine M Disteche
- Department of Pathology and Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
15
|
Tsukahara S, Morishita M. Sexually Dimorphic Formation of the Preoptic Area and the Bed Nucleus of the Stria Terminalis by Neuroestrogens. Front Neurosci 2020; 14:797. [PMID: 32848568 PMCID: PMC7403479 DOI: 10.3389/fnins.2020.00797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/07/2020] [Indexed: 01/08/2023] Open
Abstract
Testicular androgens during the perinatal period play an important role in the sexual differentiation of the brain of rodents. Testicular androgens transported into the brain act via androgen receptors or are the substrate of aromatase, which synthesizes neuroestrogens that act via estrogen receptors. The latter that occurs in the perinatal period significantly contributes to the sexual differentiation of the brain. The preoptic area (POA) and the bed nucleus of the stria terminalis (BNST) are sexually dimorphic brain regions that are involved in the regulation of sex-specific social behaviors and the reproductive neuroendocrine system. Here, we discuss how neuroestrogens of testicular origin act in the perinatal period to organize the sexually dimorphic structures of the POA and BNST. Accumulating data from rodent studies suggest that neuroestrogens induce the sex differences in glial and immune cells, which play an important role in the sexually dimorphic formation of the dendritic synapse patterning in the POA, and induce the sex differences in the cell number of specific neuronal cell groups in the POA and BNST, which may be established by controlling the number of cells dying by apoptosis or the phenotypic organization of living cells. Testicular androgens in the peripubertal period also contribute to the sexual differentiation of the POA and BNST, and thus their aromatization to estrogens may be unnecessary. Additionally, we discuss the notion that testicular androgens that do not aromatize to estrogens can also induce significant effects on the sexually dimorphic formation of the POA and BNST.
Collapse
Affiliation(s)
- Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| |
Collapse
|
16
|
Rosinger ZJ, De Guzman RM, Jacobskind JS, Saglimbeni B, Malone M, Fico D, Justice NJ, Forni PE, Zuloaga DG. Sex-dependent effects of chronic variable stress on discrete corticotropin-releasing factor receptor 1 cell populations. Physiol Behav 2020; 219:112847. [PMID: 32081812 DOI: 10.1016/j.physbeh.2020.112847] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/25/2020] [Accepted: 02/13/2020] [Indexed: 12/19/2022]
Abstract
Anxiety and depression are strikingly more prevalent in women compared with men. Dysregulation of corticotropin-releasing factor (CRF) binding to its cognate receptor (CRFR1) is thought to play a critical role in the etiology of these disorders. In the present study, we investigated whether there were sex differences in the effects of chronic variable stress (CVS) on CRFR1 cells using CRFR1-GFP reporter mice experiencing a 9-day CVS paradigm. Brains were collected from CVS and stress naïve female and male mice following exposure to the open field test. This CVS paradigm effectively increased anxiety-like behavior in female and male mice. In addition, we assessed changes in activation of CRFR1 cells (co-localization with c-Fos and phosphorylated CREB (pCREB)) in stress associated brain structures, including two sexually dimorphic CRFR1 cell groups in the anteroventral periventricular nucleus (AVPV/PeN; F>M) and paraventricular hypothalamus (PVN; M>F). CVS increased CRFR1-GFP cell number as well as the number of CRFR1/pCREB co-expressing cells in the female but not male AVPV/PeN. In the PVN, the number of CRFR1/pCREB co-expressing cells was overall greater in males regardless of treatment and CVS resulted in a male-specific reduction of CRFR1/c-Fos cells. In addition, CVS induced a female-specific reduction in CRFR1/c-Fos cells within the anteroventral bed nucleus of the stria terminalis and both sexes exhibited a reduction in CRFR1/c-Fos co-expressing cells following CVS within the ventral basolateral amygdala. Overall, these sex-specific effects of CVS on CRFR1 populations may have implications for sex differences in stress-induction of mood disorders.
Collapse
Affiliation(s)
- Zachary J Rosinger
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Rose M De Guzman
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Jason S Jacobskind
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Brianna Saglimbeni
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Margaret Malone
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Danielle Fico
- Department of Psychology, University at Albany, Albany, NY 12222, United States
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, United States
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, United States
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, NY 12222, United States.
| |
Collapse
|
17
|
Kammel LG, Correa SM. Selective sexual differentiation of neurone populations may contribute to sex-specific outputs of the ventromedial nucleus of the hypothalamus. J Neuroendocrinol 2020; 32:e12801. [PMID: 31605642 PMCID: PMC6982598 DOI: 10.1111/jne.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/26/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
Sex differences among neurones in the ventrolateral region of the ventromedial hypothalamic nucleus (VMHvl) allow for the display of a diversity of sex-typical behaviours and physiological responses, ranging from mating behaviour to metabolism. Here, we review recent studies that interrogate the relationship between sex-typical responses and changes in cellular phenotypes. We discuss technologies that increase the resolution of molecular profiling or targeting of cell populations, including single-cell transcriptional profiling and conditional viral genetic approaches to manipulate neurone survival or activity. Overall, emerging studies indicate that sex-typical functions of the VMH may be mediated by phenotypically distinct and sexually differentiated neurone populations within the VMHvl. Future studies in this and other brain regions could exploit cell-type-specific tools to reveal the cell populations and molecular mediators that modulate sex-typical responses. Furthermore, cell-type-specific analyses of the effects of sexually differentiating factors, including sex hormones, can test the hypothesis that distinct cell types within a single brain region vary with respect to sexual differentiation.
Collapse
Affiliation(s)
- Laura G Kammel
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
- Molecular, Cellular, Integrative Physiology Graduate Program, University of California, Los Angeles, CA, USA
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Cisternas CD, Cortes LR, Golynker I, Castillo-Ruiz A, Forger NG. Neonatal Inhibition of DNA Methylation Disrupts Testosterone-Dependent Masculinization of Neurochemical Phenotype. Endocrinology 2020; 161:5631853. [PMID: 31742329 DOI: 10.1210/endocr/bqz022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/18/2019] [Indexed: 11/19/2022]
Abstract
Many neural sex differences are differences in the number of neurons of a particular phenotype. For example, male rodents have more calbindin-expressing neurons in the medial preoptic area (mPOA) and bed nucleus of the stria terminalis (BNST), and females have more neurons expressing estrogen receptor alpha (ERα) and kisspeptin in the ventromedial nucleus of the hypothalamus (VMH) and the anteroventral periventricular nucleus (AVPV), respectively. These sex differences depend on neonatal exposure to testosterone, but the underlying molecular mechanisms are unknown. DNA methylation is important for cell phenotype differentiation throughout the developing organism. We hypothesized that testosterone causes sex differences in neurochemical phenotype via changes in DNA methylation, and tested this by inhibiting DNA methylation neonatally in male and female mice, and in females given a masculinizing dose of testosterone. Neonatal testosterone treatment masculinized calbindin, ERα and kisspeptin cell number of females at weaning. Inhibiting DNA methylation with zebularine increased calbindin cell number only in control females, thus eliminating sex differences in calbindin in the mPOA and BNST. Zebularine also reduced the sex difference in ERα cell number in the VMH, in this case by increasing ERα neuron number in males and testosterone-treated females. In contrast, the neonatal inhibition of DNA methylation had no effect on kisspeptin cell number. We conclude that testosterone normally increases the number of calbindin cells and reduces ERα cells in males through orchestrated changes in DNA methylation, contributing to, or causing, the sex differences in both cell types.
Collapse
Affiliation(s)
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | - Ilona Golynker
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | | | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA
| |
Collapse
|
19
|
Gegenhuber B, Tollkuhn J. Signatures of sex: Sex differences in gene expression in the vertebrate brain. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e348. [PMID: 31106965 PMCID: PMC6864223 DOI: 10.1002/wdev.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | |
Collapse
|
20
|
Li S, Kendall J, Park S, Wang Z, Alexander J, Moffitt A, Ranade N, Danyko C, Gegenhuber B, Fischer S, Robinson BD, Lepor H, Tollkuhn J, Gillis J, Brouzes E, Krasnitz A, Levy D, Wigler M. Copolymerization of single-cell nucleic acids into balls of acrylamide gel. Genome Res 2019; 30:49-61. [PMID: 31727682 PMCID: PMC6961581 DOI: 10.1101/gr.253047.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/13/2019] [Indexed: 01/06/2023]
Abstract
We show the use of 5′-Acrydite oligonucleotides to copolymerize single-cell DNA or RNA into balls of acrylamide gel (BAGs). Combining this step with split-and-pool techniques for creating barcodes yields a method with advantages in cost and scalability, depth of coverage, ease of operation, minimal cross-contamination, and efficient use of samples. We perform DNA copy number profiling on mixtures of cell lines, nuclei from frozen prostate tumors, and biopsy washes. As applied to RNA, the method has high capture efficiency of transcripts and sufficient consistency to clearly distinguish the expression patterns of cell lines and individual nuclei from neurons dissected from the mouse brain. By using varietal tags (UMIs) to achieve sequence error correction, we show extremely low levels of cross-contamination by tracking source-specific SNVs. The method is readily modifiable, and we will discuss its adaptability and diverse applications.
Collapse
Affiliation(s)
- Siran Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Sarah Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Zihua Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Joan Alexander
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Andrea Moffitt
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Nissim Ranade
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Cassidy Danyko
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Bruno Gegenhuber
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Stephan Fischer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, New York 10021, USA
| | - Herbert Lepor
- Department of Urology, New York University Langone Medical Center, New York, New York 10017, USA
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jesse Gillis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Eric Brouzes
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, USA
| | - Alex Krasnitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Dan Levy
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Michael Wigler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| |
Collapse
|
21
|
Keiser AA, Wood MA. Examining the contribution of histone modification to sex differences in learning and memory. Learn Mem 2019; 26:318-331. [PMID: 31416905 PMCID: PMC6699407 DOI: 10.1101/lm.048850.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/08/2019] [Indexed: 01/04/2023]
Abstract
The epigenome serves as a signal integration platform that encodes information from experience and environment that adds tremendous complexity to the regulation of transcription required for memory, beyond the directions encoded in the genome. To date, our understanding of how epigenetic mechanisms integrate information to regulate gene expression required for memory is primarily obtained from male derived data despite sex-specific life experiences and sex differences in consolidation and retrieval of memory, and in the molecular mechanisms that mediate these processes. In this review, we examine the contribution of chromatin modification to learning and memory in both sexes. We provide examples of how exposure to a number of internal and external factors influence the epigenome in sex-similar and sex-specific ways that may ultimately impact transcription required for memory processes. We also pose a number of key open questions and identify areas requiring further investigation as we seek to understand how histone modifying mechanisms shape memory in females.
Collapse
Affiliation(s)
- Ashley A Keiser
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, USA
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, California 92697, USA
| |
Collapse
|
22
|
Worley NB, Dumais KM, Yuan JC, Newman LE, Alonso AG, Gillespie TC, Hobbs NJ, Breedlove SM, Jordan CL, Bredewold R, Veenema AH. Oestrogen and androgen receptor activation contribute to the masculinisation of oxytocin receptors in the bed nucleus of the stria terminalis of rats. J Neuroendocrinol 2019; 31:e12760. [PMID: 31233647 DOI: 10.1111/jne.12760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 12/29/2022]
Abstract
Oxytocin (OT) often regulates social behaviours in sex-specific ways, and this may be a result of sex differences in the brain OT system. Adult male rats show higher OT receptor (OTR) binding in the posterior bed nucleus of the stria terminalis (pBNST) than adult female rats. In the present study, we investigated the mechanisms that lead to this sex difference. First, we found that male rats have higher OTR mRNA expression in the pBNST than females at postnatal day (P) 35 and P60, which demonstrates the presence of the sex difference in OTR binding density at message level. Second, the sex difference in OTR binding density in the pBNST was absent at P0 and P3, but was present by P5. Third, systemic administration of the oestrogen receptor (ER) antagonist fulvestrant at P0 and P1 dose-dependently reduced OTR binding density in the pBNST of 5-week-old male rats, but did not eliminate the sex difference in OTR binding density. Fourth, pBNST-OTR binding density was lower in androgen receptor (AR) deficient genetic male rats compared to wild-type males, but higher compared to wild-type females. Finally, systemic administration of the histone deacetylase inhibitor valproic acid at P0 and P1 did not alter pBNST-OTR binding density in 5-week-old male and female rats. Interestingly, neonatal ER antagonism, AR deficiency, and neonatal valproic acid treatment each eliminated the sex difference in pBNST size. Overall, we demonstrate a role for neonatal ER and AR activation in setting up the sex difference in OTR binding density in the pBNST, which may underlie sexual differentiation of the pBNST and social behaviour.
Collapse
Affiliation(s)
- Nicholas B Worley
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Kelly M Dumais
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Jingting C Yuan
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Laura E Newman
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Andrea G Alonso
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Tessa C Gillespie
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
| | - Nicholas J Hobbs
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - S Marc Breedlove
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Cynthia L Jordan
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Remco Bredewold
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Alexa H Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, Chestnut Hill, MA, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
23
|
Velasco ER, Florido A, Milad MR, Andero R. Sex differences in fear extinction. Neurosci Biobehav Rev 2019; 103:81-108. [PMID: 31129235 PMCID: PMC6692252 DOI: 10.1016/j.neubiorev.2019.05.020] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/08/2019] [Accepted: 05/19/2019] [Indexed: 12/18/2022]
Abstract
Despite the exponential increase in fear research during the last years, few studies have included female subjects in their design. The need to include females arises from the knowledge gap of mechanistic processes underlying the behavioral and neural differences observed in fear extinction. Moreover, the exact contribution of sex and hormones in relation to learning and behavior is still largely unknown. Insights from this field could be beneficial as fear-related disorders are twice as prevalent in women compared to men. Here, we review an up-to-date summary of animal and human studies in adulthood that report sex differences in fear extinction from a structural and functional approach. Furthermore, we describe how these factors could contribute to the observed sex differences in fear extinction during normal and pathological conditions.
Collapse
Affiliation(s)
- E R Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - A Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - M R Milad
- Department of Psychiatry, University of Illinois at Chicago, USA
| | - R Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
24
|
John N, Rehman H, Razak S, David M, Ullah W, Afsar T, Almajwal A, Alam I, Jahan S. Comparative study of environmental pollutants bisphenol A and bisphenol S on sexual differentiation of anteroventral periventricular nucleus and spermatogenesis. Reprod Biol Endocrinol 2019; 17:53. [PMID: 31292004 PMCID: PMC6621953 DOI: 10.1186/s12958-019-0491-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bisphenol A is well known endocrine-disrupting chemical while Bisphenol S was considered a safe alternative. The present study aims to examine the comparative effects of xenobiotic bisphenol-A (BPA) and its substitute bisphenol-S (BPS) on spermatogenesis and development of sexually dimorphic nucleus population of dopaminergic neurons in the anteroventral periventricular nucleus (AVPV) of the hypothalamus in male pups. METHODS Sprague Dawley rat's pups were administered subcutaneously at the neonatal stage from postnatal day PND1 to PND 27. Thirty animals were divided into six experimental groups (6 animals/group). The first group served as control and was provided with normal olive oil. The four groups were treated with 2 μg/kg and 200 μg/kg of BPA and BPS, respectively. The sixth group was given with 50 μg/kg of estradiol dissolved in olive oil as a standard to find the development of dopaminergic tyrosine hydroxylase neurons in AVPV regions. Histological analysis for testicular tissues and immunohistochemistry for brain tissues was performed. RESULTS The results revealed adverse histopathological changes in testis after administration of different doses of BPA and BPS. These degenerative changes were marked by highly significant (p < 0.001) decrease in tubular and luminal diameters of seminiferous tubule and epithelial height among bisphenols treated groups as compared to control. Furthermore, significantly increased (p < 0.001) TH-ir cell bodies in the AVPV region of the brain with 200 μg/kg dose of BPA and BPS was evident. CONCLUSION It is concluded that exposure of BPA and BPS during a critical developmental period can structural impairments in testes and affects sexual differentiation of a dimorphic dopaminergic population of AVPV region of hypothalamus in the male brain.
Collapse
Affiliation(s)
- Naham John
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Humaira Rehman
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Suhail Razak
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mehwish David
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Waheed Ullah
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| | - Tayyaba Afsar
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ali Almajwal
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Iftikhar Alam
- 0000 0004 1773 5396grid.56302.32Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sarwat Jahan
- 0000 0001 2215 1297grid.412621.2Reproductive Physiology Lab, Department of Animal Sciences, Quaid- i- Azam University Islamabad, Islamabad, 45320 Pakistan
| |
Collapse
|
25
|
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
|
26
|
Rosinger ZJ, Jacobskind JS, Bulanchuk N, Malone M, Fico D, Justice NJ, Zuloaga DG. Characterization and gonadal hormone regulation of a sexually dimorphic corticotropin-releasing factor receptor 1 cell group. J Comp Neurol 2018; 527:1056-1069. [PMID: 30499109 DOI: 10.1002/cne.24588] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/16/2018] [Accepted: 11/09/2018] [Indexed: 12/11/2022]
Abstract
Corticotropin-releasing factor binds with high affinity to CRF receptor 1 (CRFR1) and is implicated in stress-related mood disorders such as anxiety and depression. Using a validated CRFR1-green fluorescent protein (GFP) reporter mouse, our laboratory recently discovered a nucleus of CRFR1 expressing cells that is prominent in the female rostral anteroventral periventricular nucleus (AVPV/PeN), but largely absent in males. This sex difference is present in the early postnatal period and remains dimorphic into adulthood. The present investigation sought to characterize the chemical composition and gonadal hormone regulation of these sexually dimorphic CRFR1 cells using immunohistochemical procedures. We report that CRFR1-GFP-ir cells within the female AVPV/PeN are largely distinct from other dimorphic cell populations (kisspeptin, tyrosine hydroxylase). However, CRFR1-GFP-ir cells within the AVPV/PeN highly co-express estrogen receptor alpha as well as glucocorticoid receptor. A single injection of testosterone propionate or estradiol benzoate on the day of birth completely eliminates the AVPV/PeN sex difference, whereas adult gonadectomy has no effect on CRFR1-GFP cell number. These results indicate that the AVPV/PeN CRFR1 is regulated by perinatal but not adult gonadal hormones. Finally, female AVPV/PeN CRFR1-GFP-ir cells are activated following an acute 30-min restraint stress, as assessed by co-localization of CRFR1-GFP cells with phosphorylated (p) CREB. CRFR1-GFP/pCREB cells were largely absent in the male AVPV/PeN. Together, these data indicate a stress and gonadal hormone responsive nucleus that is unique to females and may contribute to sex-specific stress responses.
Collapse
Affiliation(s)
| | | | - Nicole Bulanchuk
- Department of Psychology, University at Albany, Albany, New York
| | - Margaret Malone
- Department of Psychology, University at Albany, Albany, New York
| | - Danielle Fico
- Department of Psychology, University at Albany, Albany, New York
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, Texas
| | - Damian G Zuloaga
- Department of Psychology, University at Albany, Albany, New York
| |
Collapse
|
27
|
Hatcher KM, Royston SE, Mahoney MM. Modulation of circadian rhythms through estrogen receptor signaling. Eur J Neurosci 2018; 51:217-228. [PMID: 30270552 DOI: 10.1111/ejn.14184] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/06/2018] [Accepted: 09/17/2018] [Indexed: 01/11/2023]
Abstract
Circadian rhythms are physiological and behavioral processes that exhibit a 24-hr cycle. These daily rhythms are essential for living organisms to align their behavior and physiology with the environment to increase the likelihood of survival. In mammals, circadian rhythms synchronize with the environment primarily by the suprachiasmatic nucleus, a hypothalamic brain region that integrates exogenous and endogenous timing cues. Sex steroid hormones, including estrogens, are thought to modulate sexually dimorphic behaviors through developmental programming of the brain (i.e., organization), as well as acute receptor signaling during adulthood (i.e., activation). Importantly, there are known sex differences in the expression of circadian locomotor activity and molecular organization of the suprachiasmatic nucleus, likely due, in part, to the actions of circulating estrogens. Circadian locomotor rhythms, which are coordinated by the suprachiasmatic nucleus, have been shown to be regulated by developmental and adult levels of circulating estrogens. Further, increasing evidence suggests that estrogens can modulate expression of circadian clock genes that are essential for orchestration of circadian rhythms by the suprachiasmatic nucleus. In this review, we will discuss the organizational and activational modulation of the circadian timekeeping system by estrogens through estrogen receptor signaling.
Collapse
Affiliation(s)
- Katherine M Hatcher
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Sara E Royston
- Department of Anesthesiology, Perioperative Medicine and Pain, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Spine and Pain Management, Christie Clinic, Champaign, Illinois
| | - Megan M Mahoney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
28
|
Ogawa S, Tsukahara S, Choleris E, Vasudevan N. Estrogenic regulation of social behavior and sexually dimorphic brain formation. Neurosci Biobehav Rev 2018; 110:46-59. [PMID: 30392880 DOI: 10.1016/j.neubiorev.2018.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
Abstract
It has long been known that the estrogen, 17β-estradiol (17β-E), plays a central role for female reproductive physiology and behavior. Numerous studies have established the neurochemical and molecular basis of estrogenic induction of female sexual behavior, i.e., lordosis, in animal models. In addition, 17β-E also regulates male-type sexual and aggressive behavior. In males, testosterone secreted from the testes is irreversibly aromatized to 17β-E in the brain. We discuss the contribution of two nuclear receptor isoforms, estrogen receptor (ER)α and ERβ to the estrogenic regulation of sexually dimorphic brain formation and sex-typical expression of these social behaviors. Furthermore, 17β-E is a key player for social behaviors such as social investigation, preference, recognition and memory as well as anxiety-related behaviors in social contexts. Recent studies also demonstrated that not only nuclear receptor-mediated genomic signaling but also membrane receptor-mediated non-genomic actions of 17β-E may underlie the regulation of these behaviors. Finally, we will discuss how rapidly developing research tools and ideas allow us to investigate estrogenic action by emphasizing behavioral neural networks.
Collapse
Affiliation(s)
- Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura-ku, Saitama City, Saitama 338-8570, Japan
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nandini Vasudevan
- School of Biological Sciences, University of Reading, WhiteKnights Campus, Reading, RG6 6AS, United Kingdom
| |
Collapse
|
29
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
30
|
Cellular fate decisions in the developing female anteroventral periventricular nucleus are regulated by canonical Notch signaling. Dev Biol 2018; 442:87-100. [PMID: 29885287 DOI: 10.1016/j.ydbio.2018.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/05/2018] [Indexed: 01/20/2023]
Abstract
The hypothalamic anteroventral periventricular nucleus (AVPV) is the major regulator of reproductive function within the hypothalamic-pituitary-gonadal (HPG) axis. Despite an understanding of the function of neuronal subtypes within the AVPV, little is known about the molecular mechanisms regulating their development. Previous work from our laboratory has demonstrated that Notch signaling is required in progenitor cell maintenance and formation of kisspeptin neurons of the arcuate nucleus (ARC) while simultaneously restraining POMC neuron number. Based on these findings, we hypothesized that the Notch signaling pathway may act similarly in the AVPV by promoting development of kisspeptin neurons at the expense of other neuronal subtypes. To address this hypothesis, we utilized a genetic mouse model with a conditional loss of Rbpj in Nkx2.1 expressing cells (Rbpj cKO). We noted an increase in cellular proliferation, as marked by Ki-67, in the hypothalamic ventricular zone (HVZ) in Rbpj cKO mice at E13.5. This corresponded to an increase in general neurogenesis and more TH-positive neurons. Additionally, an increase in OLIG2-positive early oligodendrocytic precursor cells was observed at postnatal day 0 in Rbpj cKO mice. By 5 weeks of age in Rbpj cKO mice, TH-positive cells were readily detected in the AVPV but few kisspeptin neurons were present. To elucidate the direct effects of Notch signaling on neuron and glia differentiation, an in vitro primary hypothalamic neurosphere assay was employed. We demonstrated that treatment with the chemical Notch inhibitor DAPT increased mKi67 and Olig2 mRNA expression while decreasing astroglial Gfap expression, suggesting Notch signaling regulates both proliferation and early glial fate decisions. A modest increase in expression of TH in both the cell soma and neurite extensions was observed after extended culture, suggesting that inhibition of Notch signaling alone is enough to bias progenitors towards a dopaminergic fate. Together, these data suggest that Notch signaling restricts early cellular proliferation and differentiation of neurons and oligodendrocytes both in vivo and in vitro and acts as a fate selector of kisspeptin neurons.
Collapse
|
31
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
32
|
Gore AC, Holley AM, Crews D. Mate choice, sexual selection, and endocrine-disrupting chemicals. Horm Behav 2018; 101:3-12. [PMID: 28888817 PMCID: PMC5845777 DOI: 10.1016/j.yhbeh.2017.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/26/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
Humans have disproportionately affected the habitat and survival of species through environmental contamination. Important among these anthropogenic influences is the proliferation of organic chemicals, some of which perturb hormone systems, the latter referred to as endocrine-disrupting chemicals (EDCs). EDCs are widespread in the environment and affect all levels of reproduction, including development of reproductive organs, hormone release and regulation through the life cycle, the development of secondary sexual characteristics, and the maturation and maintenance of adult physiology and behavior. However, what is not well-known is how the confluence of EDC actions on the manifestation of morphological and behavioral sexual traits influences mate choice, a process that requires the reciprocal evaluation of and/or acceptance of a sexual partner. Moreover, the outcomes of EDC-induced perturbations are likely to influence sexual selection; yet this has rarely been directly tested. Here, we provide background on the development and manifestation of sexual traits, reproductive competence, and the neurobiology of sexual behavior, and evidence for their perturbation by EDCs. Selection acts on individuals, with the consequences manifest in populations, and we discuss the implications for EDC contamination of these processes, and the future of species.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, USA.
| | - Amanda M Holley
- Division of Pharmacology and Toxicology, College of Pharmacy, USA; Department of Integrative Biology, College of Natural Sciences, USA
| | - David Crews
- Department of Integrative Biology, College of Natural Sciences, USA.
| |
Collapse
|
33
|
Forger NG. Past, present and future of epigenetics in brain sexual differentiation. J Neuroendocrinol 2018; 30. [PMID: 28585265 DOI: 10.1111/jne.12492] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/22/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
Sexual differentiation has long been considered "epigenetic", although the meaning of that word has shifted over time. Here, we track the evolution of ideas about epigenetics in sexual differentiation, and identify principles that have emerged from recent studies. Experiments manipulating a particular epigenetic mechanism during neonatal life demonstrate a role for both histone acetylation and DNA methylation in the development of sex differences in the brain and behaviour of rodents. In addition, hormone-dependent sex differences in the number of neurones of a particular phenotype may be programmed by differences in DNA methylation early in life. Genome-wide studies suggest that many effects of neonatal testosterone on the brain methylome do not emerge until adulthood, and there may be sex biases in the use of epigenetic marks that do not correlate with differences in gene expression. In other words, even when the transcription of a gene does not differ between males and females, the epigenetic underpinnings of that expression may differ. Finally, recent evidence suggests that sex differences in epigenetic marks may primarily serve to make gene expression more similar in males and females. We discuss the implications of these findings for understanding sex differences in susceptibility to disease, and point to recent conceptual and technical advances likely to influence the field going forward.
Collapse
Affiliation(s)
- N G Forger
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
34
|
Neuronal Loss in the Developing Cerebral Cortex of Normal and Bax-Deficient Mice: Effects of Ethanol Exposure. Neuroscience 2018; 369:278-291. [DOI: 10.1016/j.neuroscience.2017.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 11/21/2022]
|
35
|
Kanaya M, Morishita M, Tsukahara S. Temporal Expression Patterns of Genes Related to Sex Steroid Action in Sexually Dimorphic Nuclei During Puberty. Front Endocrinol (Lausanne) 2018; 9:213. [PMID: 29770127 PMCID: PMC5940742 DOI: 10.3389/fendo.2018.00213] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/16/2018] [Indexed: 01/08/2023] Open
Abstract
Sex steroids play a major role in sexually dimorphic brain development during not only the perinatal period but also the pubertal period. We previously showed that, in male mice, the estrogen receptor-α (Esr1) and aromatase (Cyp19a1) genes are essential to the sexually dimorphic formation of the anteroventral periventricular nucleus (AVPV) and the principal nucleus of the bed nucleus of the stria terminalis (BNSTp), but the estrogen receptor-β (Esr2) gene is not necessary. We also showed that the androgen receptor (Ar) gene is essential to the sexually dimorphic formation of the BNSTp. These genes are expressed in the AVPV and BNSTp of perinatal mice. However, it remains unknown whether these genes are expressed in the AVPV and BNSTp during puberty, and whether the expression, if any, differs by sex, age, and brain region. Here, we dissected the AVPV and BNSTp from Nissl-stained brain sections of male and female mice on postnatal day (PD) 20 (prepuberty), PD30 (puberty onset in females), PD40 (puberty onset in males), and PD60 (young adult) using a laser microdissection system. We then examined the mRNA levels of Esr1, Esr2, Cyp19a1, and Ar in these brain regions. In the AVPV, Esr1 mRNA levels were greater in females than males during PD20-60. Esr2 and Ar mRNA expressions did not differ between sexes. Ar mRNA levels were higher at PD30 than PD20. Cyp19a1 mRNA was not detected in the AVPV at PD20-60. In the BNSTp, Esr1 and Esr2 mRNA levels were higher in females than in males during PD20-60, although the mRNA levels of Cyp19a1 and Ar did not differ between sexes. Additionally, we revealed that orchiectomy at PD20 induced a failure of normal formation of the male BNSTp and testosterone replacement in the prepubertal period rescued the effect of orchiectomy at PD20. Taken together, it is suggested that pubertal testosterone transported to the AVPV is not converted to estradiol there and does not act via ESR1 and ESR2. By contrast, the formation of the male BNSTp may be affected by testicular testosterone during puberty via AR and/or via ESR1 after conversion to estradiol by CYP19A1.
Collapse
|
36
|
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
|
37
|
Morgan CP, Bale TL. Sex differences in microRNA-mRNA networks: examination of novel epigenetic programming mechanisms in the sexually dimorphic neonatal hypothalamus. Biol Sex Differ 2017; 8:27. [PMID: 28810930 PMCID: PMC5558756 DOI: 10.1186/s13293-017-0149-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sexual differentiation of the male brain, and specifically the stress circuitry in the hypothalamus, is primarily driven by estrogen exposure during the perinatal period. Surprisingly, this single hormone promotes diverse programs of sex-specific development that vary widely between different cell types and across the developing male brain. The complexity of this phenomenon suggests that additional layers of gene regulation, including microRNAs (miRNAs), must act downstream of estrogen to mediate this specificity. METHODS To identify noncanonical mediators of estrogen-dependent sex-specific neural development, we assayed the miRNA complement of the mouse PN2 hypothalamus by microarray following an injection of vehicle or the aromatase inhibitor, formestane. Initially, multivariate analyses were used to test the influence of sex and experimental group on the miRNA environment as a whole. Then, we utilized traditional hypothesis testing to identify individual miRNA with significantly sex-biased expression. Finally, we performed a transcriptome-wide mapping of Argonaute footprints by high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (Ago HITS-CLIP) to empirically characterize targeting relationship between estrogen-responsive miRNAs and their messenger RNA (mRNA) targets. RESULTS In this study, we demonstrated that the neonatal hypothalamic miRNA environment has robust sex differences and is dynamically responsive to estrogen. Analyses identified 162 individual miRNAs with sex-biased expression, 92 of which were estrogen-responsive. Examining the genomic distribution of these miRNAs, we found three miRNA clusters encoded within a 175-kb region of chromosome 12 that appears to be co-regulated by estrogen, likely acting broadly to alter the epigenetic programming of this locus. Ago HITS-CLIP analysis uncovered novel miRNA-target interactions within prototypical mediators of estrogen-driven sexual differentiation of the brain, including Esr1 and Cyp19a1. Finally, using Gene Ontology annotations and empirically identified miRNA-mRNA connections, we identified a gene network regulated by estrogen-responsive miRNAs that converge on biological processes relevant to sexual differentiation of the brain. CONCLUSIONS Sexual differentiation of the perinatal brain, and that of stress circuitry in the hypothalamus specifically, seems to be particularly susceptible to environmental programming effects. Integrating miRNA into our conceptualization of factors, directing differentiation of this circuitry could be an informative next step in efforts to understand the complexities behind these processes.
Collapse
Affiliation(s)
- Christopher P Morgan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 380 South University Ave, 410F Hill Pavilion, Philadelphia, PA, 19104, USA
| | - Tracy L Bale
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 380 South University Ave, 410F Hill Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
38
|
Zilkha N, Scott N, Kimchi T. Sexual Dimorphism of Parental Care: From Genes to Behavior. Annu Rev Neurosci 2017; 40:273-305. [DOI: 10.1146/annurev-neuro-072116-031447] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Noga Zilkha
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Niv Scott
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
39
|
Baum MJ. Evidence That a Sex Difference in Neonatal DNA Methylation Organizes Two Distinct Phenotypic Characteristics of Neurons in the Murine Forebrain. Endocrinology 2017; 158:1569-1571. [PMID: 28575429 DOI: 10.1210/en.2017-00364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Michael J Baum
- Department of Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
40
|
Mosley M, Weathington J, Cortes LR, Bruggeman E, Castillo-Ruiz A, Xue B, Forger NG. Neonatal Inhibition of DNA Methylation Alters Cell Phenotype in Sexually Dimorphic Regions of the Mouse Brain. Endocrinology 2017; 158:1838-1848. [PMID: 28398586 PMCID: PMC5460944 DOI: 10.1210/en.2017-00205] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/24/2017] [Indexed: 02/06/2023]
Abstract
Many of the best-studied neural sex differences relate to differences in cell number and are due to the hormonal control of developmental cell death. However, several prominent neural sex differences persist even if cell death is eliminated. We hypothesized that these may reflect cell phenotype "decisions" that depend on epigenetic mechanisms, such as DNA methylation. To test this, we treated newborn mice with the DNA methyltransferase (DNMT) inhibitor zebularine, or vehicle, and examined two sexually dimorphic markers at weaning. As expected, control males had more cells immunoreactive for calbindin-D28k (CALB) in the medial preoptic area (mPOA) and fewer cells immunoreactive for estrogen receptor α (ERα) in the ventrolateral portion of the ventromedial nucleus of the hypothalamus (VMHvl) and the mPOA than did females. Neonatal DNMT inhibition markedly increased CALB cell number in both sexes and ERα cell density in males; as a result, the sex differences in ERα in the VMHvl and mPOA were completely eliminated in zebularine-treated animals. Zebularine treatment did not affect developmental cell death or the total density of Nissl-stained cells at weaning. Thus, a neonatal disruption of DNA methylation apparently has long-term effects on the proportion of cells expressing CALB and ERα, and some of these effects are sex specific. We also found that sex differences in CALB in the mPOA and ERα in the VMHvl persist in mice with a neuron-specific depletion of either Dnmt1 or Dnmt3b, indicating that neither DNMT alone is likely to be required for the sexually dimorphic expression of these markers.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Jill Weathington
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Laura R. Cortes
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Emily Bruggeman
- Department of Biology, Neuroscience Institute and Center for Obesity Reversal, Georgia State University, Atlanta, Georgia 30302
| | - Alexandra Castillo-Ruiz
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| | - Bingzhong Xue
- Department of Biology, Neuroscience Institute and Center for Obesity Reversal, Georgia State University, Atlanta, Georgia 30302
| | - Nancy G. Forger
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30302
| |
Collapse
|
41
|
Zettergren A, Karlsson S, Studer E, Sarvimäki A, Kettunen P, Thorsell A, Sihlbom C, Westberg L. Proteomic analyses of limbic regions in neonatal male, female and androgen receptor knockout mice. BMC Neurosci 2017; 18:9. [PMID: 28056817 PMCID: PMC5217640 DOI: 10.1186/s12868-016-0332-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 12/28/2016] [Indexed: 11/10/2022] Open
Abstract
Background It is well-established that organizational effects of sex steroids during early development are fundamental for sex-typical displays of, for example, mating and aggressive behaviors in rodents and other species. Male and female brains are known to differ with respect to neuronal morphology in particular regions of the brain, including the number and size of neurons, and the density and length of dendrites in nuclei of hypothalamus and amygdala. The aim of the present study was to use global proteomics to identify proteins differentially expressed in hypothalamus/amygdala during early development (postnatal day 8) of male, female and conditional androgen receptor knockout (ARNesDel) male mice, lacking androgen receptors specifically in the brain. Furthermore, verification of selected sexually dimorphic proteins was performed using targeted proteomics. Results Our proteomic approach, iTRAQ, allowed us to investigate expression differences in the 2998 most abundantly expressed proteins in our dissected tissues. Approximately 170 proteins differed between the sexes, and 38 proteins between ARNesDel and control males (p < 0.05). In line with previous explorative studies of sexually dimorphic gene expression we mainly detected subtle protein expression differences (fold changes <1.3). The protein MARCKS (myristoylated alanine rich C kinase substrate), having the largest fold change of the proteins selected from the iTRAQ analyses and of known importance for synaptic transmission and dendritic branching, was confirmed by targeted proteomics as differentially expressed between the sexes. Conclusions Overall, our results provide solid evidence that a large number of proteins show sex differences in their brain expression and could potentially be involved in brain sexual differentiation. Furthermore, our finding of a sexually dimorphic expression of MARCKS in the brain during development warrants further investigation on the involvement in sexual differentiation of this protein. Electronic supplementary material The online version of this article (doi:10.1186/s12868-016-0332-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Zettergren
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, POB 431, 405 30, Göteborg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Sara Karlsson
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, POB 431, 405 30, Göteborg, Sweden
| | - Erik Studer
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, POB 431, 405 30, Göteborg, Sweden
| | - Anna Sarvimäki
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, POB 431, 405 30, Göteborg, Sweden
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.,Department of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Annika Thorsell
- The Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Carina Sihlbom
- The Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Lars Westberg
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, POB 431, 405 30, Göteborg, Sweden.
| |
Collapse
|
42
|
Mosley M, Shah C, Morse KA, Miloro SA, Holmes MM, Ahern TH, Forger NG. Patterns of cell death in the perinatal mouse forebrain. J Comp Neurol 2017; 525:47-64. [PMID: 27199256 PMCID: PMC5116296 DOI: 10.1002/cne.24041] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
Abstract
The importance of cell death in brain development has long been appreciated, but many basic questions remain, such as what initiates or terminates the cell death period. One obstacle has been the lack of quantitative data defining exactly when cell death occurs. We recently created a "cell death atlas," using the detection of activated caspase-3 (AC3) to quantify apoptosis in the postnatal mouse ventral forebrain and hypothalamus, and found that the highest rates of cell death were seen at the earliest postnatal ages in most regions. Here we have extended these analyses to prenatal ages and additional brain regions. We quantified cell death in 16 forebrain regions across nine perinatal ages from embryonic day (E) 17 to postnatal day (P) 11 and found that cell death peaks just after birth in most regions. We found greater cell death in several regions in offspring delivered vaginally on the day of parturition compared with those of the same postconception age but still in utero at the time of collection. We also found massive cell death in the oriens layer of the hippocampus on P1 and in regions surrounding the anterior crossing of the corpus callosum on E18 as well as the persistence of large numbers of cells in those regions in adult mice lacking the pro-death Bax gene. Together these findings suggest that birth may be an important trigger of neuronal cell death and identify transient cell groups that may undergo wholesale elimination perinatally. J. Comp. Neurol. 525:47-64, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Charisma Shah
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Kiriana A Morse
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Stephen A Miloro
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Todd H Ahern
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
43
|
Genazzani AR, Giannini A, Simoncini T. Dimorphism of Human Brain: The Basis of the Gender Differences. ISGE SERIES 2017. [DOI: 10.1007/978-3-319-41433-1_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
44
|
Rebuli ME, Gibson P, Rhodes CL, Cushing BS, Patisaul HB. Sex differences in microglial colonization and vulnerabilities to endocrine disruption in the social brain. Gen Comp Endocrinol 2016; 238:39-46. [PMID: 27102938 PMCID: PMC5067172 DOI: 10.1016/j.ygcen.2016.04.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/11/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023]
Abstract
During development, microglia, the resident immune cells of the brain, play an important role in synaptic organization. Microglial colonization of the developing brain is sexually dimorphic in some regions, including nuclei critical for the coordination of social behavior, suggesting steroid hormones have an influencing role, particularly estrogen. By extension, microglial colonization may be vulnerable to endocrine disruption. Concerns have been raised regarding the potential for endocrine disrupting compounds (EDCs) to alter brain development and behavior. Developmental exposure to Bisphenol A (BPA), a ubiquitous EDC, has been associated with altered sociosexual and mood-related behaviors in various animal models and children. Through a comparison of the promiscuous Wistar rat (Rattus norvegicus) and the socially monogamous prairie vole (Microtus ochrogaster), we are the first to observe that developmental exposure to the synthetic estrogen ethinyl estradiol (EE) or BPA alters the sex-specific colonization of the hippocampus and amygdala by microglia.
Collapse
Affiliation(s)
- Meghan E Rebuli
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; W. M. Keck Center for Behavioral Biology, Raleigh, NC 27695, USA
| | - Paul Gibson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Cassie L Rhodes
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Bruce S Cushing
- Department of Biological Sciences, University of Texas at El Paso, El Paso 79968, USA
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; W. M. Keck Center for Behavioral Biology, Raleigh, NC 27695, USA; NCSU Center for Human Health and the Environment, Raleigh, NC 27695, USA.
| |
Collapse
|
45
|
Mohr MA, Garcia FL, DonCarlos LL, Sisk CL. Neurons and Glial Cells Are Added to the Female Rat Anteroventral Periventricular Nucleus During Puberty. Endocrinology 2016; 157:2393-402. [PMID: 27145006 PMCID: PMC4891785 DOI: 10.1210/en.2015-2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anteroventral periventricular nucleus (AVPV) orchestrates the neuroendocrine-positive feedback response that triggers ovulation in female rodents. The AVPV is larger and more cell-dense in females than in males, and during puberty, only females develop the capacity to show a positive feedback response. We previously reported a potential new mechanism to explain this female-specific gain of function during puberty, namely a female-biased sex difference in the pubertal addition of new cells to the rat AVPV. Here we first asked whether this sex difference is due to greater cell proliferation and/or survival in females. Female and male rats received the cell birthdate marker 5-bromo-2'-deoxyuridine (BrdU; 200 mg/kg, ip) on postnatal day (P) 30; brains were collected at short and long intervals after BrdU administration to assess cell proliferation and survival, respectively. Overall, females had more BrdU-immunoreactive cells in the AVPV than did males, with no sex differences in the rate of cell attrition over time. Thus, the sex difference in pubertal addition of AVPV cells appears to be due to greater cell proliferation in females. Next, to determine the phenotype of pubertally born AVPV cells, daily BrdU injections were given to female rats on P28-56, and tissue was collected on P77 to assess colocalization of BrdU and markers for mature neurons or glia. Of the pubertally born AVPV cells, approximately 15% differentiated into neurons, approximately 19% into astrocytes, and approximately 23% into microglia. Thus, both neuro- and gliogenesis occur in the pubertal female rat AVPV and potentially contribute to maturation of female reproductive function.
Collapse
Affiliation(s)
- Margaret A Mohr
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Francisca L Garcia
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Lydia L DonCarlos
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| | - Cheryl L Sisk
- Neuroscience Program (M.A.M., C.L.S.), Michigan State University, East Lansing, Michigan 48824; and Department of Cell and Molecular Physiology (F.L.G., L.L.D.), Loyola University Chicago, Maywood, Illinois 60153
| |
Collapse
|
46
|
Madden AMK, Paul AT, Pritchard RA, Michel R, Zup SL. Serotonin promotes feminization of the sexually dimorphic nucleus of the preoptic area, but not the calbindin cell group. Dev Neurobiol 2016; 76:1241-1253. [DOI: 10.1002/dneu.22386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/30/2015] [Accepted: 02/16/2016] [Indexed: 01/20/2023]
Affiliation(s)
- Amanda M. K. Madden
- Graduate Program in Developmental and Brain Sciences; University of Massachusetts Boston; Boston Massachusetts 02125
- Psychology Department; University of Massachusetts Boston; Boston Massachusetts 02125
| | - Alexandria T. Paul
- Psychology Department; University of Massachusetts Boston; Boston Massachusetts 02125
| | - Rory A. Pritchard
- Psychology Department; University of Massachusetts Boston; Boston Massachusetts 02125
| | - Rebecca Michel
- Psychology Department; University of Massachusetts Boston; Boston Massachusetts 02125
| | - Susan L. Zup
- Graduate Program in Developmental and Brain Sciences; University of Massachusetts Boston; Boston Massachusetts 02125
- Psychology Department; University of Massachusetts Boston; Boston Massachusetts 02125
| |
Collapse
|
47
|
Clarkson J, Herbison AE. Hypothalamic control of the male neonatal testosterone surge. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150115. [PMID: 26833836 PMCID: PMC4785901 DOI: 10.1098/rstb.2015.0115] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2015] [Indexed: 11/12/2022] Open
Abstract
Sex differences in brain neuroanatomy and neurophysiology underpin considerable physiological and behavioural differences between females and males. Sexual differentiation of the brain is regulated by testosterone secreted by the testes predominantly during embryogenesis in humans and the neonatal period in rodents. Despite huge advances in understanding how testosterone, and its metabolite oestradiol, sexually differentiate the brain, little is known about the mechanism that actually generates the male-specific neonatal testosterone surge. This review examines the evidence for the role of the hypothalamus, and particularly the gonadotropin-releasing hormone (GnRH) neurons, in generating the neonatal testosterone surge in rodents and primates. Kisspeptin-GPR54 signalling is well established as a potent and critical regulator of GnRH neuron activity during puberty and adulthood, and we argue here for an equally important role at birth in driving the male-specific neonatal testosterone surge in rodents. The presence of a male-specific population of preoptic area kisspeptin neurons that appear transiently in the perinatal period provide one possible source of kisspeptin drive to neonatal GnRH neurons in the mouse.
Collapse
Affiliation(s)
- Jenny Clarkson
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
48
|
Forger NG. Epigenetic mechanisms in sexual differentiation of the brain and behaviour. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150114. [PMID: 26833835 DOI: 10.1098/rstb.2015.0114] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2015] [Indexed: 11/12/2022] Open
Abstract
Circumstantial evidence alone argues that the establishment and maintenance of sex differences in the brain depend on epigenetic modifications of chromatin structure. More direct evidence has recently been obtained from two types of studies: those manipulating a particular epigenetic mechanism, and those examining the genome-wide distribution of specific epigenetic marks. The manipulation of histone acetylation or DNA methylation disrupts the development of several neural sex differences in rodents. Taken together, however, the evidence suggests there is unlikely to be a simple formula for masculine or feminine development of the brain and behaviour; instead, underlying epigenetic mechanisms may vary by brain region or even by dependent variable within a region. Whole-genome studies related to sex differences in the brain have only very recently been reported, but suggest that males and females may use different combinations of epigenetic modifications to control gene expression, even in cases where gene expression does not differ between the sexes. Finally, recent findings are discussed that are likely to direct future studies on the role of epigenetic mechanisms in sexual differentiation of the brain and behaviour.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30307, USA
| |
Collapse
|
49
|
Dumais KM, Alonso AG, Immormino MA, Bredewold R, Veenema AH. Involvement of the oxytocin system in the bed nucleus of the stria terminalis in the sex-specific regulation of social recognition. Psychoneuroendocrinology 2016; 64:79-88. [PMID: 26630388 PMCID: PMC4698213 DOI: 10.1016/j.psyneuen.2015.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/08/2015] [Accepted: 11/08/2015] [Indexed: 01/03/2023]
Abstract
Sex differences in the oxytocin (OT) system in the brain may explain why OT often regulates social behaviors in sex-specific ways. However, a link between sex differences in the OT system and sex-specific regulation of social behavior has not been tested. Here, we determined whether sex differences in the OT receptor (OTR) or in OT release in the posterior bed nucleus of the stria terminalis (pBNST) mediates sex-specific regulation of social recognition in rats. We recently showed that, compared to female rats, male rats have a three-fold higher OTR binding density in the pBNST, a sexually dimorphic area implicated in the regulation of social behaviors. We now demonstrate that OTR antagonist (5 ng/0.5 μl/side) administration into the pBNST impairs social recognition in both sexes, while OT (100 pg/0.5 μl/side) administration into the pBNST prolongs the duration of social recognition in males only. These effects seem specific to social recognition, as neither treatment altered total social investigation time in either sex. Moreover, baseline OT release in the pBNST, as measured with in vivo microdialysis, did not differ between the sexes. However, males showed higher OT release in the pBNST during social recognition compared to females. These findings suggest a sex-specific role of the OT system in the pBNST in the regulation of social recognition.
Collapse
Affiliation(s)
- Kelly M. Dumais
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA,Corresponding author: Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, McGuinn 300, Chestnut Hill, MA, 02467, USA, , 617-552-6149
| | - Andrea G. Alonso
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Marisa A. Immormino
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Remco Bredewold
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Alexa H. Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| |
Collapse
|
50
|
Forger NG, Strahan JA, Castillo-Ruiz A. Cellular and molecular mechanisms of sexual differentiation in the mammalian nervous system. Front Neuroendocrinol 2016; 40:67-86. [PMID: 26790970 PMCID: PMC4897775 DOI: 10.1016/j.yfrne.2016.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/31/2015] [Accepted: 01/09/2016] [Indexed: 01/16/2023]
Abstract
Neuroscientists are likely to discover new sex differences in the coming years, spurred by the National Institutes of Health initiative to include both sexes in preclinical studies. This review summarizes the current state of knowledge of the cellular and molecular mechanisms underlying sex differences in the mammalian nervous system, based primarily on work in rodents. Cellular mechanisms examined include neurogenesis, migration, the differentiation of neurochemical and morphological cell phenotype, and cell death. At the molecular level we discuss evolving roles for epigenetics, sex chromosome complement, the immune system, and newly identified cell signaling pathways. We review recent findings on the role of the environment, as well as genome-wide studies with some surprising results, causing us to re-think often-used models of sexual differentiation. We end by pointing to future directions, including an increased awareness of the important contributions of tissues outside of the nervous system to sexual differentiation of the brain.
Collapse
Affiliation(s)
- Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | - J Alex Strahan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| | | |
Collapse
|