1
|
Esfandiarei M, Anwar F, Nuthi M, Harrison A, Barrameda ME, Curry T, Pull K, Currier Thomas T, Jadavji NM. Apoptosis is increased in cortical neurons of female Marfan Syndrome mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618278. [PMID: 39464057 PMCID: PMC11507763 DOI: 10.1101/2024.10.14.618278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Marfan Syndrome (MFS) is an autosomal dominant genetic disorder that affects connective tissue throughout the body due to mutations in the FBN1 gene. Individuals with MFS display symptoms in different organs, particularly in the vasculature, but the mechanisms of this multi-system dysfunction are still under investigation. There is still a gap in our understanding of the impact of monogenic connective tissue aberrations on the brain. This study aims to determine the impact of MFS on neurodegeneration, in cortical brain tissue of male and female MFS mice. Brain tissue of 6-month-old female and male mice with the FBN1 C1041G/+ mutation and wildtype litter mates was collected and stained for active caspase-3 (ac3), brain derived neurotrophic factor (BDNF), and neuronal nuclei (NeuN) or with TUNEL and DAPI. Data revealed increased levels of ac3 in neurons within the sensory and motor cortical areas of female MFS mice compared to sex- and age-matched controls. We confirm increased levels of apoptosis in MFS mice using TUNEL staining within the same brain areas. We also report increased levels of neuronal BDNF levels in cortical brain tissue of male and female MFS mice. These results indicate a heightened susceptibility for neurodegeneration in the mouse model of MFS.
Collapse
Affiliation(s)
- Mitra Esfandiarei
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
- Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA
- Department of Anesthesiology, Pharmacology, & Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Faizan Anwar
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Manogna Nuthi
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Alisha Harrison
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | | | - Tala Curry
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
- Department of Child Health, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Kasey Pull
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Biomedical Sciences, Midwestern University, Glendale, AZ, USA
- Department of Child Health, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Nafisa M. Jadavji
- Department of Child Health, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA
- Department of Physiology, Southern Illinois University, Carbondale, IL, USA
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
2
|
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
|
3
|
Ugrumov MV, Pavlova EN, Kolacheva AA, Dil’mukhametova LK, Bogdanov VV, Blokhin V, Pronina TS. The Periventricular Nucleus as a Brain Center Containing Dopaminergic Neurons and Neurons Expressing Individual Enzymes of Dopamine Synthesis. Int J Mol Sci 2022; 23:ijms23126739. [PMID: 35743179 PMCID: PMC9224269 DOI: 10.3390/ijms23126739] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/27/2022] Open
Abstract
Since the 1980s, the concept of dopamine-rich brain centers as clusters of only dopaminergic neurons has been fundamentally revised. It has been shown that, in addition to dopaminergic neurons, most of these centers contain neurons expressing one of the enzymes of dopamine synthesis: tyrosine hydroxylase (TH) or aromatic L-amino acid decarboxylase (AADC). We have obtained convincing evidence that in rats, the hypothalamic periventricular nucleus (PeVN) is one of the largest dopamine-rich centers, containing dopaminergic and monoenzymatic neurons. Indeed, using double immunostaining for TH and AADC, the PeVN was shown to contain almost three thousand dopaminergic and monoenzymatic neurons. According to high-performance liquid chromatography, PeVN contains L-DOPA and dopamine, which, apparently, are synthesized in monoenzymatic TH neurons and bienzymatic neurons, respectively. According to confocal microscopy, neurons (cell bodies, fibers), which were immunopositive only to TH, only to AADC, or both, are in close topographic relationships with each other and with the 3rd ventricle. These data suggest the mutual regulation of the neurons, as well as the delivery of dopamine and L-DOPA to the third ventricle, which is confirmed by their detection in the cerebrospinal fluid. Thus, evidence has been obtained that PeVN is one of the largest dopamine-rich centers of the brain, containing dopaminergic and monoenzymatic neurons.
Collapse
|
4
|
Deep conservation and co-option of programmed cell death facilitates evolution of alternative phenotypes at multiple biological levels. Semin Cell Dev Biol 2022; 145:28-41. [PMID: 35654666 DOI: 10.1016/j.semcdb.2022.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 03/04/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
Alternative phenotypes, such as polyphenisms and sexual dimorphisms, are widespread in nature and appear at all levels of biological organization, from genes and cells to morphology and behavior. Yet, our understanding of the mechanisms through which alternative phenotypes develop and how they evolve remains understudied. In this review, we explore the association between alternative phenotypes and programmed cell death, a mechanism responsible for the elimination of superfluous cells during development. We discuss the ancient origins and deep conservation of programmed cell death (its function, forms and underlying core regulatory gene networks), and propose that it was co-opted repeatedly to generate alternative phenotypes at the level of cells, tissues, organs, external morphology, and even individuals. We review several examples from across the tree of life to explore the conditions under which programmed cell death is likely to facilitate the evolution of alternative phenotypes.
Collapse
|
5
|
Beikoghli Kalkhoran S, Kararigas G. Oestrogenic Regulation of Mitochondrial Dynamics. Int J Mol Sci 2022; 23:ijms23031118. [PMID: 35163044 PMCID: PMC8834780 DOI: 10.3390/ijms23031118] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023] Open
Abstract
Biological sex influences disease development and progression. The steroid hormone 17β-oestradiol (E2), along with its receptors, is expected to play a major role in the manifestation of sex differences. E2 exerts pleiotropic effects in a system-specific manner. Mitochondria are one of the central targets of E2, and their biogenesis and respiration are known to be modulated by E2. More recently, it has become apparent that E2 also regulates mitochondrial fusion–fission dynamics, thereby affecting cellular metabolism. The aim of this article is to discuss the regulatory pathways by which E2 orchestrates the activity of several components of mitochondrial dynamics in the cardiovascular and nervous systems in health and disease. We conclude that E2 regulates mitochondrial dynamics to maintain the mitochondrial network promoting mitochondrial fusion and attenuating mitochondrial fission in both the cardiovascular and nervous systems.
Collapse
|
6
|
Kolmogorova D, Ah-Yen EG, Taylor BC, Vaggas T, Liang J, Davis T, Ismail N. Sex-specific responses of the pubertal neuroimmune axis in CD-1 mice. Brain Behav Immun Health 2021; 13:100229. [PMID: 34589744 PMCID: PMC8474685 DOI: 10.1016/j.bbih.2021.100229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022] Open
Abstract
The mechanistic relationship between the sexually dimorphic neuroimmune system and the sex-specific outcomes of a pubertal immune challenge is unclear. Therefore, we examined sex differences in the progression of cytotoxic microglial responses and blood-brain barrier (BBB) disruption to a peripubertal lipopolysaccharide (LPS) treatment in brain regions relevant to stress responses and cognitive function. Six-week-old (i.e., stress-sensitive pubertal period) male and female CD-1 mice were treated with LPS (1.5 mg/kg body weight, ip) or 0.9% saline (LPS-matched volume, ip). Sex and treatment differences in microglial (Iba1+) and apoptotic neuronal (caspase-3+/NeuN+) and non-neuronal (caspase-3+/NeuN−) expression were examined in the hippocampus, medial prefrontal cortex (mPFC), and paraventricular nucleus 24 h (sickness), one week (symptomatic recovery) and four weeks (early adulthood) post-treatment (n = 8/group). Microglial morphology was quantified with fractal analyses. Group differences in BBB permeability to 14C-sucrose were examined 24 h (whole-brain, hippocampus, prefrontal cortex, hypothalamus, and cerebellum) and one week (whole-brain) post-treatment. The acute effects of pubertal LPS were specific to females (i.e., global BBB disruption, altered microglial expression and morphology in the mPFC and hippocampus, increased hippocampal apoptosis). The residual effects of pubertal LPS-induced sickness observed in microglia persisted into adulthood in a sex- and region-specific manner. In addition to highlighting these sex-specific responses of the pubertal neuroimmune system, we report baseline region-specific sex differences in microglia spanning puberty through adulthood. We propose that these sex differences in neuroimmune-neurovascular interactions during the stress-sensitive pubertal period create sex biases in stress-related disorders of brain and behaviour. Pubertal LPS alters baseline sex differences in microglial numbers and morphology. Pubertal CD-1 mice mount sexually dimorphic neuroimmune responses to systemic LPS. Treatment effects on microglial expression and morphology differ by sex and region. The acute LPS-induced effects were specific to females.
Collapse
Affiliation(s)
- Daria Kolmogorova
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily Grace Ah-Yen
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Tiffany Vaggas
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Jacky Liang
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tama Davis
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Choe HN, Jarvis ED. The role of sex chromosomes and sex hormones in vocal learning systems. Horm Behav 2021; 132:104978. [PMID: 33895570 DOI: 10.1016/j.yhbeh.2021.104978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Vocal learning is the ability to imitate and modify sounds through auditory experience, a rare trait found in only a few lineages of mammals and birds. It is a critical component of human spoken language, allowing us to verbally transmit speech repertoires and knowledge across generations. In many vocal learning species, the vocal learning trait is sexually dimorphic, where it is either limited to males or present in both sexes to different degrees. In humans, recent findings have revealed subtle sexual dimorphism in vocal learning/spoken language brain regions and some associated disorders. For songbirds, where the neural mechanisms of vocal learning have been well studied, vocal learning appears to have been present in both sexes at the origin of the lineage and was then independently lost in females of some subsequent lineages. This loss is associated with an interplay between sex chromosomes and sex steroid hormones. Even in species with little dimorphism, like humans, sex chromosomes and hormones still have some influence on learned vocalizations. Here we present a brief synthesis of these studies, in the context of sex determination broadly, and identify areas of needed investigation to further understand how sex chromosomes and sex steroid hormones help establish sexually dimorphic neural structures for vocal learning.
Collapse
Affiliation(s)
- Ha Na Choe
- Duke University Medical Center, The Rockefeller University, Howard Hughes Medical Institute, United States of America.
| | - Erich D Jarvis
- Duke University Medical Center, The Rockefeller University, Howard Hughes Medical Institute, United States of America.
| |
Collapse
|
8
|
Drastichova Z, Rudajev V, Pallag G, Novotny J. Proteome profiling of different rat brain regions reveals the modulatory effect of prolonged maternal separation on proteins involved in cell death-related processes. Biol Res 2021; 54:4. [PMID: 33557947 PMCID: PMC7871601 DOI: 10.1186/s40659-021-00327-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/25/2021] [Indexed: 01/08/2023] Open
Abstract
Background Early-life stress in the form of maternal separation can be associated with alterations in offspring neurodevelopment and brain functioning. Here, we aimed to investigate the potential impact of prolonged maternal separation on proteomic profiling of prefrontal cortex, hippocampus and cerebellum of juvenile and young adult rats. A special attention was devoted to proteins involved in the process of cell death and redox state maintenance. Methods Long-Evans pups were separated from their mothers for 3 h daily over the first 3 weeks of life (during days 2–21 of age). Brain tissue samples collected from juvenile (22-day-old) and young adult (90-day-old) rats were used for label-free quantitative (LFQ) proteomic analysis. In parallel, selected oxidative stress markers and apoptosis-related proteins were assessed biochemically and by Western blot, respectively. Results In total, 5526 proteins were detected in our proteomic analysis of rat brain tissue. Approximately one tenth of them (586 proteins) represented those involved in cell death processes or regulation of oxidative stress balance. Prolonged maternal separation caused changes in less than half of these proteins (271). The observed alterations in protein expression levels were age-, sex- and brain region-dependent. Interestingly, the proteins detected by mass spectrometry that are known to be involved in the maintenance of redox state were not markedly altered. Accordingly, we did not observe any significant differences between selected oxidative stress markers, such as the levels of hydrogen peroxide, reduced glutathione, protein carbonylation and lipid peroxidation in brain samples from rats that underwent maternal separation and from the corresponding controls. On the other hand, a number of changes were found in cell death-associated proteins, mainly in those involved in the apoptotic and autophagic pathways. However, there were no detectable alterations in the levels of cleaved products of caspases or Bcl-2 family members. Taken together, these data indicate that the apoptotic and autophagic cell death pathways were not activated by maternal separation either in adolescent or young adult rats. Conclusion Prolonged maternal separation can distinctly modulate expression profiles of proteins associated with cell death pathways in prefrontal cortex, hippocampus and cerebellum of juvenile rats and the consequences of early-life stress may last into adulthood and likely participate in variations in stress reactivity. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00327-5.
Collapse
Affiliation(s)
- Zdenka Drastichova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Gergely Pallag
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
9
|
Enriquez KD, Gupta AR, Hoffman EJ. Signaling Pathways and Sex Differential Processes in Autism Spectrum Disorder. Front Psychiatry 2021; 12:716673. [PMID: 34690830 PMCID: PMC8531220 DOI: 10.3389/fpsyt.2021.716673] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopmental disorders associated with deficits in social communication and restrictive, repetitive patterns of behavior, that affect up to 1 in 54 children. ASDs clearly demonstrate a male bias, occurring ~4 times more frequently in males than females, though the basis for this male predominance is not well-understood. In recent years, ASD risk gene discovery has accelerated, with many whole-exome sequencing studies identifying genes that converge on common pathways, such as neuronal communication and regulation of gene expression. ASD genetics studies have suggested that there may be a "female protective effect," such that females may have a higher threshold for ASD risk, yet its etiology is not well-understood. Here, we review common biological pathways implicated by ASD genetics studies as well as recent analyses of sex differential processes in ASD using imaging genomics, transcriptomics, and animal models. Additionally, we discuss recent investigations of ASD risk genes that have suggested a potential role for estrogens as modulators of biological pathways in ASD, and highlight relevant molecular and cellular pathways downstream of estrogen signaling as potential avenues for further investigation.
Collapse
Affiliation(s)
- Kristen D Enriquez
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States
| | - Abha R Gupta
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Ellen J Hoffman
- Program on Neurogenetics, Child Study Center, Yale University School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
10
|
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
|
11
|
Vaillant C, Gueguen MM, Feat J, Charlier TD, Coumailleau P, Kah O, Brion F, Pellegrini E. Neurodevelopmental effects of natural and synthetic ligands of estrogen and progesterone receptors in zebrafish eleutheroembryos. Gen Comp Endocrinol 2020; 288:113345. [PMID: 31812531 DOI: 10.1016/j.ygcen.2019.113345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/03/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022]
Abstract
Natural and synthetic estrogens and progestins are widely used in human and veterinary medicine and are detected in waste and surface waters. Our previous studies have clearly shown that a number of these substances targets the brain to induce the estrogen-regulated brain aromatase expression but the consequences on brain development remain virtually unexplored. The aim of the present study was therefore to investigate the effect of estradiol (E2), progesterone (P4) and norethindrone (NOR), a 19-nortestosterone progestin, on zebrafish larval neurogenesis. We first demonstrated using real-time quantitative PCR that nuclear estrogen and progesterone receptor brain expression is impacted by E2, P4 and NOR. We brought evidence that brain proliferative and apoptotic activities were differentially affected depending on the steroidal hormone studied, the concentration of steroids and the region investigated. Our findings demonstrate for the first time that steroid compounds released in aquatic environment have the capacity to disrupt key cellular events involved in brain development in zebrafish embryos further questioning the short- and long-term consequences of this disruption on the physiology and behavior of organisms.
Collapse
Affiliation(s)
- Colette Vaillant
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Marie-Madeleine Gueguen
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Justyne Feat
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Thierry D Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Pascal Coumailleau
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Olivier Kah
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - François Brion
- Institut National de l'Environnement Industriel et des Risques INERIS, Unité d'Ecotoxicologie, 60550, Verneuil-en-Halatte, France
| | - Elisabeth Pellegrini
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
12
|
Role of 17 β-Estradiol on Cell Proliferation and Mitochondrial Fitness in Glioblastoma Cells. JOURNAL OF ONCOLOGY 2020; 2020:2314693. [PMID: 32148493 PMCID: PMC7042539 DOI: 10.1155/2020/2314693] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/22/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Gliomas are the most common primary tumors of the central nervous system (CNS) in the adult. Previous data showed that estrogen affects cancer cells, but its effect is cell-type-dependent and controversial. The present study aimed to analyze the effects of estradiol (E2, 5 nM) in human glioblastoma multiforme U87-MG cells and how it may impact on cell proliferation and mitochondrial fitness. We monitored cell proliferation by xCELLigence technology and mitochondrial fitness by assessing the expression of genes involved in mitochondrial biogenesis (PGC1α, SIRT1, and TFAM), oxidative phosphorylation (ND4, Cytb, COX-II, COX IV, NDUFA6, and ATP synthase), and dynamics (OPA1, MNF2, MNF1, and FIS1). Finally, we evaluated Nrf2 nuclear translocation by immunocytochemical analysis. Our results showed that E2 resulted in a significant increase in cell proliferation, with a significant increase in the expression of genes involved in various mechanisms of mitochondrial fitness. Finally, E2 treatment resulted in a significant increase of Nrf2 nuclear translocation with a significant increase in the expression of one of its target genes (i.e., heme oxygenase-1). Our results suggest that E2 promotes proliferation in glioblastoma cells and regulate the expression of genes involved in mitochondrial fitness and chemoresistance pathway.
Collapse
|
13
|
Does Birth Trigger Cell Death in the Developing Brain? eNeuro 2020; 7:ENEURO.0517-19.2020. [PMID: 32015098 PMCID: PMC7031855 DOI: 10.1523/eneuro.0517-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Developmental cell death eliminates half of the neurons initially generated in the mammalian brain, and occurs perinatally in many species. It is possible that the timing of neuronal cell death is developmentally programmed, and only coincidentally associated with birth. Alternatively, birth may play a role in shaping cell death. To test these competing hypotheses, we experimentally advanced or delayed birth by 1 d in mice (within the normal range of gestation for the species) and examined effects on the temporal pattern and magnitude (amount) of neuronal cell death, using immunohistochemical detection of activated caspase-3 as a cell death marker. In order to detect effects of subtle changes in birth timing, we focused on brain areas that exhibit sharp postnatal peaks in cell death. We find that advancing birth advances peak cell death, supporting the hypothesis that birth triggers cell death. However, a delay of birth does not delay cell death. Thus, birth can advance cell death, but if postponed, a developmental program governs. Advancing or delaying birth also caused region-specific changes in the overall magnitude of cell death. Our findings shed light on the long-standing question of what controls the timing and magnitude of developmental neuronal cell death, and position birth as an orchestrator of brain development. Because humans across the world now routinely alter birth timing, these findings may have implications for current obstetric practices.
Collapse
|
14
|
Abstract
In the past decennia, our understanding of the sexual differentiation of the mammalian brain has dramatically changed. The simple model according to which testosterone masculinizes the brain of males away from a default female form, was replaced with a complex scenario, according to which sex effects on the brain of both females and males are exerted by genetic, hormonal, and environmental factors. These factors act via multiple partly independent mechanisms that may vary according to internal and external factors. These observations led to the "mosaic" hypothesis-the expectation of high variability in the degree of "maleness"/"femaleness" of different features within a single brain. Here, we briefly review animal data that form the basis of current understanding of sexual differentiation; present, in this context, the results of co-analyses of human brain measures obtained by magnetic resonance imaging or postmortem; discuss criticisms and controversies of the mosaic hypothesis and implications for research; and conclude that co-analysis of several (preferably, many) features and going back from the group level to that of the individual would advance our understanding of the relations between sex and the brain in health and disease.
Collapse
Affiliation(s)
- Daphna Joel
- School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Alicia Garcia-Falgueras
- Netherlands Institute for Neuroscience, Amsterdam, An Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Amsterdam, the Netherlands
| | - Dick Swaab
- Netherlands Institute for Neuroscience, Amsterdam, An Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Amsterdam, the Netherlands
| |
Collapse
|
15
|
Barrientos RM, Brunton PJ, Lenz KM, Pyter L, Spencer SJ. Neuroimmunology of the female brain across the lifespan: Plasticity to psychopathology. Brain Behav Immun 2019; 79:39-55. [PMID: 30872093 PMCID: PMC6591071 DOI: 10.1016/j.bbi.2019.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/09/2019] [Indexed: 02/06/2023] Open
Abstract
The female brain is highly dynamic and can fundamentally remodel throughout the normal ovarian cycle as well as in critical life stages including perinatal development, pregnancy and old-age. As such, females are particularly vulnerable to infections, psychological disorders, certain cancers, and cognitive impairments. We will present the latest evidence on the female brain; how it develops through the neonatal period; how it changes through the ovarian cycle in normal individuals; how it adapts to pregnancy and postpartum; how it responds to illness and disease, particularly cancer; and, finally, how it is shaped by old age. Throughout, we will highlight female vulnerability to and resilience against disease and dysfunction in the face of environmental challenges.
Collapse
Affiliation(s)
- R M Barrientos
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Chronic Brain Injury Program, Discovery Themes Initiative, The Ohio State University, Columbus, OH 43210, United States
| | - P J Brunton
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, Scotland, UK; Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, International Campus, Haining, Zhejiang 314400, PR China
| | - K M Lenz
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychology, Department of Neuroscience, The Ohio State University, Columbus, OH 43210, United States
| | - L Pyter
- Institute for Behavioral Medicine Research, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States; Department of Psychiatry and Behavioral Health, Wexner Medical Centre, The Ohio State University, Columbus, OH 43210, United States
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic. 3083, Australia.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Sominsky L, Jasoni CL, Twigg HR, Spencer SJ. Hormonal and nutritional regulation of postnatal hypothalamic development. J Endocrinol 2018; 237:R47-R64. [PMID: 29545398 DOI: 10.1530/joe-17-0722] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is a key centre for regulation of vital physiological functions, such as appetite, stress responsiveness and reproduction. Development of the different hypothalamic nuclei and its major neuronal populations begins prenatally in both altricial and precocial species, with the fine tuning of neuronal connectivity and attainment of adult function established postnatally and maintained throughout adult life. The perinatal period is highly susceptible to environmental insults that, by disrupting critical developmental processes, can set the tone for the establishment of adult functionality. Here, we review the most recent knowledge regarding the major postnatal milestones in the development of metabolic, stress and reproductive hypothalamic circuitries, in the rodent, with a particular focus on perinatal programming of these circuitries by hormonal and nutritional influences. We also review the evidence for the continuous development of the hypothalamus in the adult brain, through changes in neurogenesis, synaptogenesis and epigenetic modifications. This degree of plasticity has encouraging implications for the ability of the hypothalamus to at least partially reverse the effects of perinatal mal-programming.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical SciencesRMIT University, Melbourne, Victoria, Australia
| | - Christine L Jasoni
- School of Biomedical SciencesCentre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Hannah R Twigg
- School of Biomedical SciencesCentre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Sarah J Spencer
- School of Health and Biomedical SciencesRMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Kamitakahara A, Bouyer K, Wang CH, Simerly R. A critical period for the trophic actions of leptin on AgRP neurons in the arcuate nucleus of the hypothalamus. J Comp Neurol 2017; 526:133-145. [PMID: 28891045 DOI: 10.1002/cne.24327] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 12/11/2022]
Abstract
In the developing hypothalamus, the fat-derived hormone leptin stimulates the growth of axons from the arcuate nucleus of the hypothalamus (ARH) to other regions that control energy balance. These projections are significantly reduced in leptin deficient (Lepob/ob ) mice and this phenotype is largely rescued by neonatal leptin treatments. However, treatment of mature Lepob/ob mice is ineffective, suggesting that the trophic action of leptin is limited to a developmental critical period. To temporally delineate closure of this critical period for leptin-stimulated growth, we treated Lepob/ob mice with exogenous leptin during a variety of discrete time periods, and measured the density of Agouti-Related Peptide (AgRP) containing projections from the ARH to the ventral part of the dorsomedial nucleus of the hypothalamus (DMHv), and to the medial parvocellular part of the paraventricular nucleus (PVHmp). The results indicate that leptin loses its neurotrophic potential at or near postnatal day 28. The duration of leptin exposure appears to be important, with 9- or 11-day treatments found to be more effective than shorter (5-day) treatments. Furthermore, leptin treatment for 9 days or more was sufficient to restore AgRP innervation to both the PVHmp and DMHv in Lepob/ob females, but only to the DMHv in Lepob/ob males. Together, these findings reveal that the trophic actions of leptin are contingent upon timing and duration of leptin exposure, display both target and sex specificity, and that modulation of leptin-dependent circuit formation by each of these factors may carry enduring consequences for feeding behavior, metabolism, and obesity risk.
Collapse
Affiliation(s)
- Anna Kamitakahara
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.,Program in Neuroscience, University of Southern California, Los Angeles, California
| | - Karine Bouyer
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Chien-Hua Wang
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.,Program in Neuroscience, University of Southern California, Los Angeles, California
| | - Richard Simerly
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
21
|
Rosinger ZJ, Jacobskind JS, Park SG, Justice NJ, Zuloaga DG. Distribution of corticotropin-releasing factor receptor 1 in the developing mouse forebrain: A novel sex difference revealed in the rostral periventricular hypothalamus. Neuroscience 2017; 361:167-178. [PMID: 28823817 DOI: 10.1016/j.neuroscience.2017.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/19/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022]
Abstract
Corticotropin-releasing factor (CRF) signaling through CRF receptor 1 (CRFR1) regulates autonomic, endocrine and behavioral responses to stress and has been implicated in the pathophysiology of several disorders including anxiety, depression, and addiction. Using a validated CRFR1 reporter mouse line (bacterial artificial chromosome identified by green fluorescence protein (BAC GFP-CRFR1)), we investigated the distribution of CRFR1 in the developing mouse forebrain. Distribution of CRFR1 was investigated at postnatal days (P) 0, 4, and 21 in male and female mice. CRFR1 increased with age in several regions including the medial amygdala, arcuate nucleus, paraventricular hypothalamus, medial septum, CA1 hippocampal area, and the lateral habenula. Regions showing decreased CRFR1 expression with increased age include the intermediate portion of the periventricular hypothalamic nucleus, and CA3 hippocampal area. We report a sexually dimorphic expression of CRFR1 within the rostral portion of the anteroventral periventricular nucleus of the hypothalamus (AVPV/PeN), a region known to regulate ovulation, reproductive and maternal behaviors. Females had a greater number of CRFR1-GFP-ir cells at all time points in the AVPV/PeN and CRFR1-GFP-ir was nearly absent in males by P21. Overall, alterations in CRFR1-GFP-ir distribution based on age and sex may contribute to observed age- and sex-dependent differences in stress regulation.
Collapse
Affiliation(s)
| | | | - Shannon G Park
- University at Albany, Department of Psychology, Albany, NY 12222, USA
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, USA.
| |
Collapse
|
22
|
Neuroimmunology and neuroepigenetics in the establishment of sex differences in the brain. Nat Rev Neurosci 2017. [PMID: 28638119 DOI: 10.1038/nrn.2017.61] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The study of sex differences in the brain is a topic of neuroscientific study that has broad reaching implications for culture, society and biomedical science. Recent research in rodent models has led to dramatic shifts in our views of the mechanisms underlying the sexual differentiation of the brain. These include the surprising discoveries of a role for immune cells and inflammatory mediators in brain masculinization and a role for epigenetic suppression in brain feminization. How and to what degree these findings will translate to human brain development will be questions of central importance in future research in this field.
Collapse
|
23
|
Selective estrogen receptor modulator: A novel polysaccharide from Sparganii Rhizoma induces apoptosis in breast cancer cells. Carbohydr Polym 2017; 163:199-207. [DOI: 10.1016/j.carbpol.2017.01.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 01/29/2023]
|
24
|
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
|
25
|
Joel D, McCarthy MM. Incorporating Sex As a Biological Variable in Neuropsychiatric Research: Where Are We Now and Where Should We Be? Neuropsychopharmacology 2017; 42:379-385. [PMID: 27240659 PMCID: PMC5399245 DOI: 10.1038/npp.2016.79] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 05/17/2016] [Accepted: 05/23/2016] [Indexed: 11/09/2022]
Abstract
Understanding the multiplicity of ways in which sex can alter the brain is essential to crafting policies and treatments that are beneficial for all human beings. This is particularly true for the field of neuropsychopharmacology, as many neuropsychiatric disorders exhibit gender bias in the frequency, severity, or response to treatment. The goal of this circumspective is to provide two views on the current state of the art of the relations between sex and the brain, relations that are studied almost exclusively by comparing females and males on specific end points, from gene expression to behavior. We start by suggesting a framework for defining what is being measured and what it means. We suggest that 'sex differences' can be classified on four dimensions: (1) persistent vs transient across the lifespan; (2) context independent vs dependent; (3) dimorphic vs continuous; and (4) a direct vs an indirect consequence of sex. To accurately classify a sex difference along these dimensions, one may need to compare females and males under varied conditions. We next discuss current data on the mechanisms of sexual differentiation of the brain and on sex differences in the brain to conclude that the brain of each male and female is a mosaic of relative masculinization, feminization, and sameness, which theoretically could produce an infinite variety of individuals. We also raise the possibility that sex differences in the brain are canalized, which may act to both enhance and restrain variation between males and females. We end by discussing ways to consider sex when studying neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daphna Joel
- School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel,School of Psychological Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel, Tel: +972 3 640 8996, Fax: +972 3 640 9547, E-mail:
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Guillamon A, Junque C, Gómez-Gil E. A Review of the Status of Brain Structure Research in Transsexualism. ARCHIVES OF SEXUAL BEHAVIOR 2016; 45:1615-48. [PMID: 27255307 PMCID: PMC4987404 DOI: 10.1007/s10508-016-0768-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/22/2015] [Accepted: 04/29/2016] [Indexed: 05/22/2023]
Abstract
The present review focuses on the brain structure of male-to-female (MtF) and female-to-male (FtM) homosexual transsexuals before and after cross-sex hormone treatment as shown by in vivo neuroimaging techniques. Cortical thickness and diffusion tensor imaging studies suggest that the brain of MtFs presents complex mixtures of masculine, feminine, and demasculinized regions, while FtMs show feminine, masculine, and defeminized regions. Consequently, the specific brain phenotypes proposed for MtFs and FtMs differ from those of both heterosexual males and females. These phenotypes have theoretical implications for brain intersexuality, asymmetry, and body perception in transsexuals as well as for Blanchard's hypothesis on sexual orientation in homosexual MtFs. Falling within the aegis of the neurohormonal theory of sex differences, we hypothesize that cortical differences between homosexual MtFs and FtMs and male and female controls are due to differently timed cortical thinning in different regions for each group. Cross-sex hormone studies have reported marked effects of the treatment on MtF and FtM brains. Their results are used to discuss the early postmortem histological studies of the MtF brain.
Collapse
Affiliation(s)
- Antonio Guillamon
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia, c/Juand del Rosal, 10, 28040, Madrid, Spain.
- Academia de Psicología de España, Madrid, Spain.
| | - Carme Junque
- Departamento de Psiquiatría y Psicobiología Clínica, Universidad de Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer, Barcelona, Spain
| | - Esther Gómez-Gil
- Institute of Biomedical Research August Pi i Sunyer, Barcelona, Spain
- Unidad de Identidad de Género, Hospital Clinic, Barcelona, Spain
| |
Collapse
|
27
|
Panzica G, Melcangi RC. Structural and molecular brain sexual differences: A tool to understand sex differences in health and disease. Neurosci Biobehav Rev 2016; 67:2-8. [DOI: 10.1016/j.neubiorev.2016.04.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 02/07/2023]
|
28
|
Sex differences in the brain–an interplay of sex steroid hormones and sex chromosomes. Clin Sci (Lond) 2016; 130:1481-97. [DOI: 10.1042/cs20160299] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022]
Abstract
Although considerable progress has been made in our understanding of brain function, many questions remain unanswered. The ultimate goal of studying the brain is to understand the connection between brain structure and function and behavioural outcomes. Since sex differences in brain morphology were first observed, subsequent studies suggest different functional organization of the male and female brains in humans. Sex and gender have been identified as being a significant factor in understanding human physiology, health and disease, and the biological differences between the sexes is not limited to the gonads and secondary sexual characteristics, but also affects the structure and, more crucially, the function of the brain and other organs. Significant variability in brain structures between individuals, in addition to between the sexes, is factor that complicates the study of sex differences in the brain. In this review, we explore the current understanding of sex differences in the brain, mostly focusing on preclinical animal studies.
Collapse
|
29
|
Gonadal hormone modulation of intracellular calcium as a mechanism of neuroprotection. Front Neuroendocrinol 2016; 42:40-52. [PMID: 26930421 DOI: 10.1016/j.yfrne.2016.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/22/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.
Collapse
|
30
|
Schiller CE, Johnson SL, Abate AC, Schmidt PJ, Rubinow DR. Reproductive Steroid Regulation of Mood and Behavior. Compr Physiol 2016; 6:1135-60. [PMID: 27347888 PMCID: PMC6309888 DOI: 10.1002/cphy.c150014] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this article, we examine evidence supporting the role of reproductive steroids in the regulation of mood and behavior in women and the nature of that role. In the first half of the article, we review evidence for the following: (i) the reproductive system is designed to regulate behavior; (ii) from the subcellular to cellular to circuit to behavior, reproductive steroids are powerful neuroregulators; (iii) affective disorders are disorders of behavioral state; and (iv) reproductive steroids affect virtually every system implicated in the pathophysiology of depression. In the second half of the article, we discuss the diagnosis of the three reproductive endocrine-related mood disorders (premenstrual dysphoric disorder, postpartum depression, and perimenopausal depression) and present evidence supporting the relevance of reproductive steroids to these conditions. Existing evidence suggests that changes in reproductive steroid levels during specific reproductive states (i.e., the premenstrual phase of the menstrual cycle, pregnancy, parturition, and the menopause transition) trigger affective dysregulation in susceptible women, thus suggesting the etiopathogenic relevance of these hormonal changes in reproductive mood disorders. Understanding the source of individual susceptibility is critical to both preventing the onset of illness and developing novel, individualized treatments for reproductive-related affective dysregulation. © 2016 American Physiological Society. Compr Physiol 6:1135-1160, 2016e.
Collapse
Affiliation(s)
- Crystal Edler Schiller
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah L. Johnson
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anna C. Abate
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Schmidt
- Section on Behavioral Endocrinology, National Institute of Mental Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - David R. Rubinow
- Psychiatry Department, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
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
|
32
|
Syntabulin regulates the trafficking of PICK1-containing vesicles in neurons. Sci Rep 2016; 6:20924. [PMID: 26868290 PMCID: PMC4751430 DOI: 10.1038/srep20924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/08/2016] [Indexed: 11/08/2022] Open
Abstract
PICK1 (protein interacting with C-kinase 1) is a peripheral membrane protein that interacts with diverse membrane proteins. PICK1 has been shown to regulate the clustering and membrane localization of synaptic receptors such as AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors, metabotropic glutamate receptor 7, and ASICs (acid-sensing ion channels). Moreover, recent evidence suggests that PICK1 can mediate the trafficking of various vesicles out from the Golgi complex in several cell systems, including neurons. However, how PICK1 affects vesicle-trafficking dynamics remains unexplored. Here, we show that PICK1 mediates vesicle trafficking by interacting with syntabulin, a kinesin-binding protein that mediates the trafficking of both synaptic vesicles and mitochondria in axons. Syntabulin recruits PICK1 onto microtubule structures and mediates the trafficking of PICK1-containing vesicles along microtubules. In neurons, syntabulin alters PICK1 expression by recruiting PICK1 into axons and regulates the trafficking dynamics of PICK1-containing vesicles. Furthermore, we show that syntabulin forms a complex with PICK1 and ASICs, regulates ASIC protein expression in neurons, and participates in ASIC-induced acidotoxicity.
Collapse
|
33
|
McCarthy MM, Pickett LA, VanRyzin JW, Kight KE. Surprising origins of sex differences in the brain. Horm Behav 2015; 76:3-10. [PMID: 25917865 PMCID: PMC4620061 DOI: 10.1016/j.yhbeh.2015.04.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 11/22/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Discerning the biologic origins of neuroanatomical sex differences has been of interest since they were first reported in the late 60's and early 70's. The centrality of gonadal hormone exposure during a developmental critical window cannot be denied but hormones are indirect agents of change, acting to induce gene transcription or modulate membrane bound signaling cascades. Sex differences in the brain include regional volume differences due to differential cell death, neuronal and glial genesis, dendritic branching and synaptic patterning. Early emphasis on mechanism therefore focused on neurotransmitters and neural growth factors, but by and large these endpoints failed to explain the origins of neural sex differences. More recently evidence has accumulated in favor of inflammatory mediators and immune cells as principle regulators of brain sexual differentiation and reveal that the establishment of dimorphic circuits is not cell autonomous but instead requires extensive cell-to-cell communication including cells of non-neuronal origin. Despite the multiplicity of cells involved the nature of the sex differences in the neuroanatomical endpoints suggests canalization, a process that explains the robustness of individuals in the face of intrinsic and extrinsic variability. We propose that some neuroanatomical endpoints are canalized to enhance sex differences in the brain by reducing variability within one sex while also preventing the sexes from diverging too greatly. We further propose mechanisms by which such canalization could occur and discuss what relevance this may have to sex differences in behavior.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Lindsay A Pickett
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jonathan W VanRyzin
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine E Kight
- Department of Pharmacology, Program in Neuroscience and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
34
|
Neonatal testosterone suppresses a neuroendocrine pulse generator required for reproduction. Nat Commun 2015; 5:3285. [PMID: 24518793 DOI: 10.1038/ncomms4285] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/20/2014] [Indexed: 01/21/2023] Open
Abstract
The pituitary gland releases hormones in a pulsatile fashion guaranteeing signalling efficiency. The determinants of pulsatility are poorly circumscribed. Here we show in magnocellular hypothalamo-neurohypophyseal oxytocin (OT) neurons that the bursting activity underlying the neurohormonal pulses necessary for parturition and the milk-ejection reflex is entirely driven by a female-specific central pattern generator (CPG). Surprisingly, this CPG is active in both male and female neonates, but is inactivated in males after the first week of life. CPG activity can be restored in males by orchidectomy or silenced in females by exogenous testosterone. This steroid effect is aromatase and caspase dependent, and is mediated via oestrogen receptor-α. This indicates the apoptosis of the CPG network during hypothalamic sexual differentiation, explaining why OT neurons do not burst in adult males. This supports the view that stereotypic neuroendocrine pulsatility is governed by CPGs, some of which are subjected to gender-specific perinatal programming.
Collapse
|
35
|
Ferguson SA, Paule MG, He Z. Pre- and postnatal bisphenol A treatment does not alter the number of tyrosine hydroxylase-positive cells in the anteroventral periventricular nucleus (AVPV) of weanling male and female rats. Brain Res 2015. [PMID: 26206302 DOI: 10.1016/j.brainres.2015.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Exposure to Bisphenol A (BPA) may interfere with brain sexual differentiation. Altered numbers of tyrosine hydroxylase (TH) cells in the rodent anteroventral periventricular nucleus (AVPV) after developmental BPA treatment have been reported; however, definitive conclusions are lacking. The current study incorporated many of the guidelines suggested for endocrine disrupter research. Specifically, ethinyl estradiol (EE2) served as a reference estrogen, exogenous environmental estrogen exposure was controlled, BPA was administered orally, and subjects consumed a low phytoestrogen diet. Here, on gestational days 6-21, Sprague-Dawley rats (10-15/group) were gavaged with 2.5 or 25.0 µg BPA/kg/day or 5.0 or 10.0 µg EE2/kg/day or the vehicle (5 ml of 0.3% aqueous carboxymethylcellulose/kg/day). A naïve control group was weighed and restrained, but not gavaged. Beginning on postnatal day (PND) 1 and continuing until PND 21, the 4 pups/sex/litter were orally treated with the same dose their dam had received. On PND 21, 1/sex/litter was perfused and the brain removed. TH immunoreactivity (TH-ir) was counted in 8 images/pup by a technician blind to treatment status. ANOVA results indicated significantly higher TH-ir cells/mm(2) in females (main effect of sex: p<0.01); however, there was no significant effect of treatment or a significant interaction of treatment with sex. In a separate untreated group of PND 21 Sprague-Dawley pups, AVPV volume was quantified and no significant sexual dimorphism was apparent. Similar to our reported results of behavioral assessments, the BPA treatment paradigm used here (2.5 or 25.0 µg BPA/kg/day administered orally) does not appear to cause significant alterations in AVPV TH-ir.
Collapse
Affiliation(s)
- Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, 3900 NCTR Road, Jefferson, AR 72079, United States.
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, 3900 NCTR Road, Jefferson, AR 72079, United States.
| | - Zhen He
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, 3900 NCTR Road, Jefferson, AR 72079, United States.
| |
Collapse
|
36
|
Chen L, Chu C, Kong X, Huang T, Cai YD. Discovery of new candidate genes related to brain development using protein interaction information. PLoS One 2015; 10:e0118003. [PMID: 25635857 PMCID: PMC4311913 DOI: 10.1371/journal.pone.0118003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/03/2015] [Indexed: 12/18/2022] Open
Abstract
Human brain development is a dramatic process composed of a series of complex and fine-tuned spatiotemporal gene expressions. A good comprehension of this process can assist us in developing the potential of our brain. However, we have only limited knowledge about the genes and gene functions that are involved in this biological process. Therefore, a substantial demand remains to discover new brain development-related genes and identify their biological functions. In this study, we aimed to discover new brain-development related genes by building a computational method. We referred to a series of computational methods used to discover new disease-related genes and developed a similar method. In this method, the shortest path algorithm was executed on a weighted graph that was constructed using protein-protein interactions. New candidate genes fell on at least one of the shortest paths connecting two known genes that are related to brain development. A randomization test was then adopted to filter positive discoveries. Of the final identified genes, several have been reported to be associated with brain development, indicating the effectiveness of the method, whereas several of the others may have potential roles in brain development.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People’s Republic of China
| | - Chen Chu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, People’s Republic of China
- * E-mail: (TH); (YDC)
| | - Yu-Dong Cai
- Institute of Systems Biology, Shanghai University, Shanghai 200444, People’s Republic of China
- * E-mail: (TH); (YDC)
| |
Collapse
|
37
|
Zup SL, Edwards NS, McCarthy MM. Sex- and age-dependent effects of androgens on glutamate-induced cell death and intracellular calcium regulation in the developing hippocampus. Neuroscience 2014; 281:77-87. [PMID: 25264034 DOI: 10.1016/j.neuroscience.2014.09.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 08/31/2014] [Accepted: 09/18/2014] [Indexed: 12/12/2022]
Abstract
Hippocampal neurons must maintain control over cytosolic calcium levels, especially during development, as excitation and calcium flux are necessary for proper growth and function. But excessive calcium can lead to excitotoxic cell death. Previous work suggests that neonatal male and female hippocampal neurons regulate cytosolic calcium differently, thereby leading to differential susceptibility to excitotoxic damage. Hippocampal neurons are also exposed to gonadal hormones during development and express high levels of androgen receptors. Androgens have both neuroprotective and neurotoxic effects in adults and developing animals. The present study sought to examine the effect of androgen on cell survival after an excitatory stimulus in the developing hippocampus, and whether androgen-mediated calcium regulation was the governing mechanism. We observed that glutamate did not induce robust or sexually dimorphic apoptosis in cultured hippocampal neurons at an early neonatal time point, but did 5days later - only in males. Further, pretreatment with the androgen dihydrotestosterone (DHT) protected males from apoptosis during this time, but had no effect on females. Calcium imaging of sex-specific cultures revealed that DHT decreased the peak of intracellular calcium induced by glutamate, but only in males. To determine a possible mechanism for this androgen neuroprotection and calcium regulation, we quantified three calcium regulatory proteins, plasma membrane calcium ATPase1 (PMCA1), sodium/calcium exchanger1 (NCX1), and the sarco/endoplasmic reticulum calcium ATPase 2 (SERCA2). Surprisingly, there was no sex difference in the level of any of the three proteins. Treatment with DHT significantly decreased PMCA1 and NCX1, but increased SERCA2 protein levels in very young animals but not at a later timepoint. Taken together, these data suggest a complex interaction of sex, hormones, calcium regulation and developmental age; however androgens acting during the first week of life are implicated in regulation of hippocampal cell death and may be an underlying mechanism for sexually dimorphic apoptosis.
Collapse
Affiliation(s)
- S L Zup
- Program in Developmental and Brain Sciences, University of Massachusetts Boston, Boston, MA 02125, United States; Department of Psychology, University of Massachusetts Boston, Boston, MA 02125, United States.
| | - N S Edwards
- Department of Pharmacology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - M M McCarthy
- Department of Pharmacology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| |
Collapse
|
38
|
Aromatase, estrogen receptors and brain development in fish and amphibians. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:152-62. [PMID: 25038582 DOI: 10.1016/j.bbagrm.2014.07.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/19/2014] [Accepted: 07/07/2014] [Indexed: 12/20/2022]
Abstract
Estrogens affect brain development of vertebrates, not only by impacting activity and morphology of existing circuits, but also by modulating embryonic and adult neurogenesis. The issue is complex as estrogens can not only originate from peripheral tissues, but also be locally produced within the brain itself due to local aromatization of androgens. In this respect, teleost fishes are quite unique because aromatase is expressed exclusively in radial glial cells, which represent pluripotent cells in the brain of all vertebrates. Expression of aromatase in the brain of fish is also strongly stimulated by estrogens and some androgens. This creates a very intriguing positive auto-regulatory loop leading to dramatic aromatase expression in sexually mature fish with elevated levels of circulating steroids. Looking at the effects of estrogens or anti-estrogens in the brain of adult zebrafish showed that estrogens inhibit rather than stimulate cell proliferation and newborn cell migration. The functional meaning of these observations is still unclear, but these data suggest that the brain of fish is experiencing constant remodeling under the influence of circulating steroids and brain-derived neurosteroids, possibly permitting a diversification of sexual strategies, notably hermaphroditism. Recent data in frogs indicate that aromatase expression is limited to neurons and do not concern radial glial cells. Thus, until now, there is no other example of vertebrates in which radial progenitors express aromatase. This raises the question of when and why these new features were gained and what are their adaptive benefits. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
|
39
|
Lee DA, Yoo S, Pak T, Salvatierra J, Velarde E, Aja S, Blackshaw S. Dietary and sex-specific factors regulate hypothalamic neurogenesis in young adult mice. Front Neurosci 2014; 8:157. [PMID: 24982613 PMCID: PMC4056383 DOI: 10.3389/fnins.2014.00157] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/26/2014] [Indexed: 11/26/2022] Open
Abstract
The hypothalamus is the central regulator of a broad range of homeostatic and instinctive physiological processes, such as the sleep-wake cycle, food intake, and sexually dimorphic behaviors. These behaviors can be modified by various environmental and physiological cues, although the molecular and cellular mechanisms that mediate these effects remain poorly understood. Recently, it has become clear that both the juvenile and adult hypothalamus exhibit ongoing neurogenesis, which serve to modify homeostatic neural circuitry. In this report, we share new findings on the contributions of sex-specific and dietary factors to regulating neurogenesis in the hypothalamic mediobasal hypothalamus, a recently identified neurogenic niche. We report that high fat diet (HFD) selectively activates neurogenesis in the median eminence (ME) of young adult female but not male mice, and that focal irradiation of the ME in HFD-fed mice reduces weight gain in females but not males. These results suggest that some physiological effects of high fat diet are mediated by the stimulation of ME neurogenesis in a sexually dimorphic manner. We discuss these results in the context of recent advances in understanding the cellular and molecular mechanisms that regulate neurogenesis in postnatal and adult hypothalamus.
Collapse
Affiliation(s)
- Daniel A Lee
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Division of Biology and Biomedical Engineering, California Institute of Technology Pasadena, CA, USA
| | - Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Thomas Pak
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Juan Salvatierra
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Sciences, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Susan Aja
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Institute for Cell Engineering, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Center for High-Throughput Biology, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
40
|
Zubiaurre-Elorza L, Junque C, Gómez-Gil E, Guillamon A. Effects of Cross-Sex Hormone Treatment on Cortical Thickness in Transsexual Individuals. J Sex Med 2014; 11:1248-61. [DOI: 10.1111/jsm.12491] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Sharif A, Ojeda SR, Prevot V. Neurogenesis and Gliogenesis in the Postnatal Hypothalamus: A New Level of Plasticity for the Regulation of Hypothalamic Function? ENDOGENOUS STEM CELL-BASED BRAIN REMODELING IN MAMMALS 2014. [DOI: 10.1007/978-1-4899-7399-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Li G, Zhang W, Baker MS, Laritsky E, Mattan-Hung N, Yu D, Kunde-Ramamoorthy G, Simerly RB, Chen R, Shen L, Waterland RA. Major epigenetic development distinguishing neuronal and non-neuronal cells occurs postnatally in the murine hypothalamus. Hum Mol Genet 2013; 23:1579-90. [PMID: 24186871 DOI: 10.1093/hmg/ddt548] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Prenatal and early postnatal environment can persistently alter one's risk of obesity. Environmental effects on hypothalamic developmental epigenetics constitute a likely mechanism underlying such 'developmental programming' of energy balance regulation. To advance our understanding of these processes, it is essential to develop approaches to disentangle the cellular and regional heterogeneity of hypothalamic developmental epigenetics. We therefore performed genome-scale DNA methylation profiling in hypothalamic neurons and non-neuronal cells at postnatal day 0 (P0) and P21 and found, surprisingly, that most of the DNA methylation differences distinguishing these two cell types are established postnatally. In particular, neuron-specific increases in DNA methylation occurred extensively at genes involved in neuronal development. Quantitative bisulfite pyrosequencing verified our methylation profiling results in all 15 regions examined, and expression differences were associated with DNA methylation at several genes. We also identified extensive methylation differences between the arcuate (ARH) and paraventricular nucleus of the hypothalamus (PVH). Integrating these two data sets showed that genomic regions with PVH versus ARH differential methylation strongly overlap with those undergoing neuron-specific increases from P0 to P21, suggesting that these developmental changes occur preferentially in either the ARH or PVH. In particular, neuron-specific methylation increases at the 3' end of Shh localized to the ARH and were positively associated with gene expression. Our data indicate a key role for DNA methylation in establishing the gene expression potential of diverse hypothalamic cell types, and provide the novel insight that early postnatal life is a critical period for cell type-specific epigenetic development in the murine hypothalamus.
Collapse
Affiliation(s)
- Ge Li
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kelly DA, Varnum MM, Krentzel AA, Krug S, Forger NG. Differential control of sex differences in estrogen receptor α in the bed nucleus of the stria terminalis and anteroventral periventricular nucleus. Endocrinology 2013; 154:3836-46. [PMID: 24025225 PMCID: PMC3776875 DOI: 10.1210/en.2013-1239] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The principal nucleus of the bed nucleus of the stria terminalis (BNSTp) and anteroventral periventricular nucleus of the hypothalamus (AVPV) are sexually dimorphic, hormone-sensitive forebrain regions. Here we report a profound sex difference in estrogen receptor-α (ERα) immunoreactivity (IR) in the BNSTp, with robust ERα IR in females and the near absence of labeling in males. This sex difference is due to the suppression of ERα IR by testicular hormones in adulthood: it was not present at birth and was not altered by neonatal treatment of females with estradiol; gonadectomy of adult males increased ERα IR to that of females, whereas gonadectomy of adult females had no effect. Treating gonadally intact males with an aromatase inhibitor partially feminized ERα IR in the BNSTp, suggesting that testicular suppression required aromatization. By contrast, in AVPV we found a modest sex difference in ERα IR that was relatively insensitive to steroid manipulations in adulthood. ERα IR in AVPV was, however, masculinized in females treated with estradiol at birth, suggesting that the sex difference is due to organizational effects of estrogens. The difference in ERα IR in the BNSTp of males and females appears to be at least in part due to greater expression of mRNA of the ERα gene (Esr1) in females. The sex difference in message is smaller than the difference in immunoreactivity, however, suggesting that posttranscriptional mechanisms also contribute to the pronounced suppression of ERα IR and presumably to functions mediated by ERα in the male BNSTp.
Collapse
Affiliation(s)
- D A Kelly
- PhD, Department of Psychology, University of Massachusetts, Amherst, Massachusetts 01003.
| | | | | | | | | |
Collapse
|
44
|
Ahern TH, Krug S, Carr AV, Murray EK, Fitzpatrick E, Bengston L, McCutcheon J, De Vries GJ, Forger NG. Cell death atlas of the postnatal mouse ventral forebrain and hypothalamus: effects of age and sex. J Comp Neurol 2013; 521:2551-69. [PMID: 23296992 PMCID: PMC4968939 DOI: 10.1002/cne.23298] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/28/2012] [Accepted: 12/26/2012] [Indexed: 01/21/2023]
Abstract
Naturally occurring cell death is essential to the development of the mammalian nervous system. Although the importance of developmental cell death has been appreciated for decades, there is no comprehensive account of cell death across brain areas in the mouse. Moreover, several regional sex differences in cell death have been described for the ventral forebrain and hypothalamus, but it is not known how widespread the phenomenon is. We used immunohistochemical detection of activated caspase-3 to identify dying cells in the brains of male and female mice from postnatal day (P) 1 to P11. Cell death density, total number of dying cells, and regional volume were determined in 16 regions of the hypothalamus and ventral forebrain (the anterior hypothalamus, arcuate nucleus, anteroventral periventricular nucleus, medial preoptic nucleus, paraventricular nucleus, suprachiasmatic nucleus, and ventromedial nucleus of the hypothalamus; the basolateral, central, and medial amygdala; the lateral and principal nuclei of the bed nuclei of the stria terminalis; the caudate-putamen; the globus pallidus; the lateral septum; and the islands of Calleja). All regions showed a significant effect of age on cell death. The timing of peak cell death varied between P1 to P7, and the average rate of cell death varied tenfold among regions. Several significant sex differences in cell death and/or regional volume were detected. These data address large gaps in the developmental literature and suggest interesting region-specific differences in the prevalence and timing of cell death in the hypothalamus and ventral forebrain.
Collapse
Affiliation(s)
- Todd H. Ahern
- Center for Behavioral Neuroscience, Department of Psychology, Quinnipiac University, Hamden, Connecticut 06518
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Stefanie Krug
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Audrey V. Carr
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Elaine K. Murray
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Emmett Fitzpatrick
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Lynn Bengston
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Jill McCutcheon
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
| | - Geert J. De Vries
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Nancy G. Forger
- Department of Psychology, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| |
Collapse
|
45
|
Iwakura T, Sakoh M, Tsutiya A, Yamashita N, Ohtani A, Tsuda MC, Ogawa S, Tsukahara S, Nishihara M, Shiga T, Goshima Y, Kato T, Ohtani-Kaneko R. Collapsin response mediator protein 4 affects the number of tyrosine hydroxylase-immunoreactive neurons in the sexually dimorphic nucleus in female mice. Dev Neurobiol 2013; 73:502-17. [PMID: 23420586 DOI: 10.1002/dneu.22076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/25/2012] [Accepted: 02/12/2013] [Indexed: 01/04/2023]
Abstract
In the sexually dimorphic anteroventral periventricular nucleus (AVPV) of the hypothalamus, females have a greater number of tyrosine hydroxylase-immunoreactive (TH-ir) and kisspeptin-immunoreactive (kisspeptin-ir) neurons than males. In this study, we used proteomics analysis and gene-deficient mice to identify proteins that regulate the number of TH-ir and kisspeptin-ir neurons in the AVPV. Analysis of protein expressions in the rat AVPV on postnatal day 1 (PD1; the early phase of sex differentiation) using two-dimensional fluorescence difference gel electrophoresis followed by MALDI-TOF-MS identified collapsin response mediator protein 4 (CRMP4) as a protein exhibiting sexually dimorphic expression. Interestingly, this sexually differential expressions of CRMP4 protein and mRNA in the AVPV was not detected on PD6. Prenatal testosterone exposure canceled the sexual difference in the expression of Crmp4 mRNA in the rat AVPV. Next, we used CRMP4-knockout (CRMP4-KO) mice to determine the in vivo function of CRMP4 in the AVPV. Crmp4 knockout did not change the number of kisspeptin-ir neurons in the adult AVPV in either sex. However, the number of TH-ir neurons was increased in the AVPV of adult female CRMP4-KO mice as compared with the adult female wild-type mice. During development, no significant difference in the number of TH-ir neurons was detected between sexes or genotypes on embryonic day 15, but a female-specific increase in TH-ir neurons was observed in CRMP4-KO mice on PD1, when the sex difference was not yet apparent in wild-type mice. These results indicate that CRMP4 regulates the number of TH-ir cell number in the female AVPV.
Collapse
Affiliation(s)
- Takashi Iwakura
- Doctoral Program in Kansei, Behavioral and Brain Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8577, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Waddell J, Bowers JM, Edwards NS, Jordan CL, McCarthy MM. Dysregulation of neonatal hippocampal cell genesis in the androgen insensitive Tfm rat. Horm Behav 2013; 64:144-52. [PMID: 23747829 PMCID: PMC3753588 DOI: 10.1016/j.yhbeh.2013.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 05/23/2013] [Accepted: 05/29/2013] [Indexed: 12/20/2022]
Abstract
The first two weeks of life are a critical period for hippocampal development. At this time gonadal steroid exposure organizes sex differences in hippocampal sensitivity to activational effects of steroids, hippocampal cell morphology and hippocampus dependent behaviors. Our laboratory has characterized a robust sex difference in neonatal neurogenesis in the hippocampus that is mediated by estradiol. Here, we extend our knowledge of this sex difference by comparing the male and female hippocampus to the androgen insensitive testicular feminized mutant (Tfm) rat. In the neonatal Tfm rat hippocampus, fewer newly generated cells survive compared to males or females. This deficit in cell genesis is partially recovered with the potent androgen DHT, but is more completely recovered following estradiol administration. Tfm rats do not differ from males or females in the level of endogenous estradiol in the neonatal hippocampus, suggesting other mechanisms mediate a differential sensitivity to estradiol in male, female and Tfm hippocampus. We also demonstrate disrupted performance on a hippocampal-dependent contextual fear discrimination task. Tfm rats generalize fear across contexts, and do not exhibit significant loss of fear during extinction exposure. These results extend prior reports of exaggerated response to stress in Tfm rats, and following gonadectomy in normal male rats.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Pediatrics, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
47
|
The development of kisspeptin circuits in the Mammalian brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:221-52. [PMID: 23550009 DOI: 10.1007/978-1-4614-6199-9_11] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The neuropeptide kisspeptin, encoded by the Kiss1 gene, is required for mammalian puberty and fertility. Examining the development of the kisspeptin system contributes to our understanding of pubertal progression and adult reproduction and sheds light on possible mechanisms underlying the development of reproductive disorders, such as precocious puberty or hypogonadotropic hypogonadism. Recent work, primarily in rodent models, has begun to study the development of kisspeptin neurons and their regulation by sex steroids and other factors at early life stages. In the brain, kisspeptin is predominantly expressed in two areas of the hypothalamus, the anteroventral periventricular nucleus and neighboring periventricular nucleus (pre-optic area in some species) and the arcuate nucleus. Kisspeptin neurons in these two hypothalamic regions are differentially regulated by testosterone and estradiol, both in development and in adulthood, and also display differences in their degree of sexual dimorphism. In this chapter, we discuss what is currently known and not known about the ontogeny, maturation, and sexual differentiation of kisspeptin neurons, as well as their regulation by sex steroids and other factors during development.
Collapse
|
48
|
Poling MC, Kauffman AS. Organizational and activational effects of sex steroids on kisspeptin neuron development. Front Neuroendocrinol 2013; 34:3-17. [PMID: 22728025 PMCID: PMC3725275 DOI: 10.1016/j.yfrne.2012.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 05/17/2012] [Accepted: 06/07/2012] [Indexed: 11/29/2022]
Abstract
Kisspeptin, encoded by the Kiss1 gene, is a neuropeptide required for puberty and adult reproductive function. Understanding the regulation and development of the kisspeptin system provides valuable knowledge about the physiology of puberty and adult fertility, and may provide insights into human pubertal or reproductive disorders. Recent studies, particularly in rodent models, have assessed how kisspeptin neurons develop and how hormonal and non-hormonal factors regulate this developmental process. Exposure to sex steroids (testosterone and estradiol) during critical periods of development can induce organizational (permanent) effects on kisspeptin neuron development, with respect to both sexually dimorphic and non-sexually dimorphic aspects of kisspeptin biology. In addition, sex steroids can also impart activational (temporary) effects on kisspeptin neurons and Kiss1 gene expression at various times during neonatal and peripubertal development, as they do in adulthood. Here, we discuss the current knowledge--and in some cases, lack thereof--of the influence of hormones and other factors on kisspeptin neuronal development.
Collapse
Affiliation(s)
- Matthew C Poling
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
49
|
Morgan CP, Bale TL. Sex differences in microRNA regulation of gene expression: no smoke, just miRs. Biol Sex Differ 2012; 3:22. [PMID: 23009289 PMCID: PMC3507674 DOI: 10.1186/2042-6410-3-22] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 09/22/2012] [Indexed: 11/10/2022] Open
Abstract
Males and females differ widely in morphology, physiology, and behavior leading to disparities in many health outcomes, including sex biases in the prevalence of many neurodevelopmental disorders. However, with the exception of a relatively small number of genes on the Y chromosome, males and females share a common genome. Therefore, sexual differentiation must in large part be a product of the sex biased expression of this shared genetic substrate. microRNAs (miRs) are small non-coding RNAs involved in the post-transcriptional regulation of up to 70% of protein-coding genes. The ability of miRs to regulate such a vast amount of the genome with a high degree of specificity makes them perfectly poised to play a critical role in programming of the sexually dimorphic brain. This review describes those characteristics of miRs that make them particularly amenable to this task, and examines the influences of both the sex chromosome complement as well as gonadal hormones on their regulation. Exploring miRs in the context of sex differences in disease, particularly in sex-biased neurodevelopmental disorders, may provide novel insight into the pathophysiology and potential therapeutic targets in disease treatment and prevention.
Collapse
Affiliation(s)
- Christopher P Morgan
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Ste, 201E, Philadelphia, PA, 19104-6046, USA.
| | | |
Collapse
|
50
|
Szymanski L, Bakker J. Aromatase knockout mice show normal steroid-induced activation of gonadotrophin-releasing hormone neurones and luteinising hormone surges with a reduced population of kisspeptin neurones in the rostral hypothalamus. J Neuroendocrinol 2012; 24:1222-33. [PMID: 22577852 DOI: 10.1111/j.1365-2826.2012.02334.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We recently reported that female aromatase knockout (ArKO) mice show deficits in sexual behaviour and a decreased population of kisspeptin-immunoreactive neurones in the rostral periventricular area of the third ventricle (RP3V), resurrecting the question of whether oestradiol actively contributes to female-typical sexual differentiation. To further address this question, we assessed the capacity of ArKO mice to generate a steroid-induced luteinising hormone (LH) surge. Adult, gonadectomised wild-type (WT) and ArKO mice were given silastic oestradiol implants s.c. and, 1 week later, received s.c. injections of either oestradiol benzoate (EB) followed by progesterone, EB alone, or no additional steroids to activate gonadotrophin-releasing hormone (GnRH) neurones and generate an LH surge. Treatment with EB and progesterone induced significant Fos/GnRH double-labelling and, consequently, an LH surge in female WT and in ArKO mice of both sexes but not in male WT mice. ArKO mice of both sexes had fewer cells expressing Kiss-1 mRNA in the RP3V compared to female WT mice but had more Kiss-1 mRNA-expressing cells compared to WT males, reflecting an incomplete sexual differentiation of this system. To determine the number of cells expressing kisspeptin, the same experimental design was repeated in Experiment 2 with the addition of groups of WT and ArKO mice that were given EB + progesterone and sacrificed 2 h before the expected LH surge. No differences were observed in the number of kisspeptin-immunoreactive cells 2 h before and at the time of the LH surge. The finding that ArKO mice of both sexes have a competent LH surge system suggests that oestradiol has predominantly defeminising actions on the GnRH/LH surge system in males and that the steroid-induced LH surge can occur in females even with a greatly reduced population of kisspeptin neurones in the RP3V.
Collapse
Affiliation(s)
- L Szymanski
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | | |
Collapse
|