1
|
Konishi K, Jacobs EG, Aroner S, De Vivo I, Smith B, Scribner-Weiss B, Makris N, Seitz-Holland J, Remington A, Aizley H, Kubicki M, Goldstein JM. Leukocyte telomere length and memory circuitry and cognition in early aging: Impact of sex and menopausal status. Horm Behav 2024; 165:105631. [PMID: 39232410 PMCID: PMC11438173 DOI: 10.1016/j.yhbeh.2024.105631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Telomere length (TL) is an important cellular marker of biological aging impacting the brain and heart. However, how it is related to the brain (e.g., cognitive function and neuroanatomic architecture), and how these relationships may vary by sex and reproductive status, is not well established. Here we assessed the association between leukocyte TL and memory circuitry regional brain volumes and memory performance in early midlife, in relation to sex and reproductive status. Participants (N = 198; 95 females, 103 males; ages 45-55) underwent structural MRI and neuropsychological assessments of verbal, associative, and working memory. Overall, shorter TL was associated with smaller white matter volume in the parahippocampal gyrus and dorsolateral prefrontal cortex. In males, shorter TL was associated with worse working memory performance and corresponding smaller white matter volumes in the parahippocampal gyrus, anterior cingulate cortex, and dorsolateral prefrontal cortex. In females, the impact of cellular aging was revealed over the menopausal transition. In postmenopausal females, shorter TL was associated with poor associative memory performance and smaller grey matter volume in the right hippocampus. In contrast, TL was not related to memory performance or grey and white matter volumes in any memory circuitry region in pre/perimenopausal females. Results demonstrated that shorter TL is associated with worse memory function and smaller volume in memory circuitry regions in early midlife, an association that differs by sex and reproductive status. Taken together, TL may serve as an early indicator of sex-dependent brain abnormalities in early midlife.
Collapse
Affiliation(s)
- Kyoko Konishi
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara 93111, United States of America
| | - Sarah Aroner
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Immaculata De Vivo
- Department of Epidemiology, T.H. Chan School of Public Health, Boston, MA 02120, United States of America
| | - Brianna Smith
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Blair Scribner-Weiss
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States of America
| | - Nikos Makris
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Johanna Seitz-Holland
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Anne Remington
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Harlyn Aizley
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Marek Kubicki
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Jill M Goldstein
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
2
|
Sharma DR, Cheng B, Sahu R, Zhang X, Mehdizadeh R, Singh D, Iacobas D, Ballabh P. Oestrogen treatment restores dentate gyrus development in premature newborns by IGF1 regulation. J Cell Mol Med 2023; 27:2467-2481. [PMID: 37594177 PMCID: PMC10468667 DOI: 10.1111/jcmm.17816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 08/19/2023] Open
Abstract
Prematurely-born infants cared for in the neonatal units suffer from memory and learning deficits. Prematurity diminishes neurogenesis and synaptogenesis in the hippocampal dentate gyrus (DG). This dysmaturation of neurons is attributed to elevated PSD95, NMDR2A, and IGF1 levels. Since oestrogen treatment plays key roles in the development and plasticity of DG, we hypothesized that 17β-estradiol (E2) treatment would ameliorate neurogenesis and synaptogenesis in the DG, reversing cognitive deficits in premature newborns. Additionally, E2-induced recovery would be mediated by IGF1 signalling. These hypotheses were tested in a rabbit model of prematurity and nonmaternal care, in which premature kits were gavage-fed and reared by laboratory personnel. We compared E2- and vehicle-treated preterm kits for morphological, molecular, and behavioural parameters. We also treated kits with oestrogen degrader, RAD1901, and assessed IGF1 signalling. We found that E2 treatment increased the number of Tbr2+ and DCX+ neuronal progenitors and increased the density of glutamatergic synapses in the DG. E2 treatment restored PSD95 and NMDAR2A levels and cognitive function in preterm kits. Transcriptomic analyses showed that E2 treatment contributed to recovery by influencing interactions between IGF1R and neurodegenerative, as well as glutamatergic genes. ERα expression was reduced on completion of E2 treatment at D7, followed by D30 elevation. E2-induced fluctuation in ERα levels was associated with a reciprocal elevation in IGF1/2 expression at D7 and reduction at D30. ERα degradation by RAD1901 treatment enhanced IGF1 levels, suggesting ERα inhibits IGF1 expression. E2 treatment alleviates the prematurity-induced maldevelopment of DG and cognitive dysfunctions by regulating ERα and IGF1 levels.
Collapse
Affiliation(s)
- Deep R. Sharma
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Bokun Cheng
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Rauhin Sahu
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Xusheng Zhang
- Computational Genomics CoreAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Rana Mehdizadeh
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Divya Singh
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Dumitru Iacobas
- Dominick P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
- Personalized Genomics Laboratory, Texas Undergraduate Medical AcademyPrairie View A&M UniversityPrairie ViewTexasUSA
| | - Praveen Ballabh
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
3
|
Effects of circulating estradiol on physiological, behavioural, and subjective correlates of anxiety: A double-blind, randomized, placebo-controlled trial. Psychoneuroendocrinology 2022; 138:105682. [PMID: 35123210 DOI: 10.1016/j.psyneuen.2022.105682] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
Anxiety-related behaviours as well as the prevalence of anxiety disorders show a large sex difference in humans. Clinical studies in humans as well as behavioural studies in rodents suggest that estradiol may have anxiolytic properties. In line with this, anxiety symptoms fluctuate with estradiol levels along the menstrual cycle. However, the influence of estradiol on subjective, behavioural, as well as physiological correlates of anxiety has never been systematically addressed in humans. We ran a double-blind, randomized, placebo-controlled study (N = 126) to investigate the effects of estradiol on anxiety in men and women. In healthy volunteers, circulating estradiol levels were elevated through estradiol administration over two consecutive days to simulate the rise in estradiol levels around ovulation. Subjective, behavioral, as well as, physiological correlates of anxiety were assessed using a virtual reality elevated plus-maze (EPM). Estradiol treatment reduced the physiological stress response with blunted heart rate response and lower cortisol levels compared to placebo treatment in both sexes. In contrast, respiration frequency was only reduced in women after estradiol treatment. Behavioural measures of anxiety as well as subjective anxiety on the EPM were not affected by estradiol treatment. In general, women showed more avoidance and less approach behavior and reported higher subjective anxiety levels on the EPM than men. These results highlight the limited anxiolytic properties of circulating levels of estradiol in humans, which influence physiological markers of anxiety but not approach and avoidance behaviour or subjective anxiety levels.
Collapse
|
4
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Blankers SA, Galea LA. Androgens and Adult Neurogenesis in the Hippocampus. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:203-215. [PMID: 35024692 PMCID: PMC8744005 DOI: 10.1089/andro.2021.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/12/2022]
Abstract
Adult neurogenesis in the hippocampus is modulated by steroid hormones, including androgens, in male rodents. In this review, we summarize research showing that chronic exposure to androgens, such as testosterone and dihydrotestosterone, enhances the survival of new neurons in the dentate gyrus of male, but not female, rodents, via the androgen receptor. However, the neurogenesis promoting the effect of androgens in the dentate gyrus may be limited to younger adulthood as it is not evident in middle-aged male rodents. Although direct exposure to androgens in adult or middle age does not significantly influence neurogenesis in female rodents, the aromatase inhibitor letrozole enhances neurogenesis in the hippocampus of middle-aged female mice. Unlike other androgens, androgenic anabolic steroids reduce neurogenesis in the hippocampus of male rodents. Collectively, the research indicates that the ability of androgens to enhance hippocampal neurogenesis in adult rodents is dependent on dose, androgen type, sex, duration, and age. We discuss these findings and how androgens may be influencing neuroprotection, via neurogenesis in the hippocampus, in the context of health and disease.
Collapse
Affiliation(s)
- Samantha A. Blankers
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| | - Liisa A.M. Galea
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
- Department of Psychology, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Luzio A, Figueiredo M, Matos MM, Coimbra AM, Álvaro AR, Monteiro SM. Effects of short-term exposure to genistein and overfeeding diet on the neural and retinal progenitor competence of adult zebrafish (Danio rerio). Neurotoxicol Teratol 2021; 88:107030. [PMID: 34506931 DOI: 10.1016/j.ntt.2021.107030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Neurogenesis is a process that occurs throughout the life of a vertebrate. Among the different factors that may affect the natural occurrence of neurogenesis, obesity seems to decrease the proliferation capacity of progenitor neuronal cells. Conversely, the phytoestrogen genistein is known to attenuate some obesity effects beyond its neuroprotective action. Aiming to improve the understanding of how obesity and genistein trigger an impact on the neural and retinal progenitor competence of adult zebrafish, fish were exposed to genistein (GEN - 2 μg L-1) alone or combined with two dietary groups (control and overfeed - OFD) for up to 9 weeks. Zebrafish were fed once per day with Artemia sp. in the control and GEN (2% of BW, control diet), and three times per day in the OFD and OFD + GEN groups (12% BW, overfeeding diet). To assess obesity induction, BMI, biometric parameters, and PPAR-γ protein were quantified. Afterwards, qRT-PCR and immunohistochemistry were performed to determine the cell proliferation and the presence of stem cells through PCNA and Sox-2. Our findings proved that overfeeding adult zebrafish increased the general growth and induced the development of fatty liver. However, for OFD + GEN, this effect was assuaged through the anti-adipogenic effect of GEN. This finding suggests that phytoestrogens could be beneficial to reduce the negative effects of obesity. Moreover, OF induced negative effects on retinal and brain homeostasis, decreasing the proliferation capacity of progenitor neuronal cells. With regard to retinal progenitor competence, genistein seems to mitigate the negative impacts of obesity, whereas the effects of obesity on the brain were exacerbated by this phytoestrogen which negatively influenced the homeostasis of zebrafish neural progenitor competence. This study highlighted the fact that the effects of phytoestrogens in adult neural progenitor competence are complex and could exhibit dissimilar effects depending on the tissue.
Collapse
Affiliation(s)
- A Luzio
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB and Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal; Department of Biology and Environment, Life Sciences and Environment School, University of Trás-os-Montes e Alto Douro, Apt. 1013, 5000-801 Vila Real, Portugal
| | - M Figueiredo
- Department of Biology and Environment, Life Sciences and Environment School, University of Trás-os-Montes e Alto Douro, Apt. 1013, 5000-801 Vila Real, Portugal
| | - M M Matos
- Department of Genetics and Biotechnology, Life Sciences and Environment School, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Biosystems & Integrative Sciences Institute (BioISI), Sciences Faculty, University of Lisbon, Lisbon, Portugal
| | - A M Coimbra
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB and Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal; Department of Genetics and Biotechnology, Life Sciences and Environment School, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - A R Álvaro
- Center for Neuroscience and Cell Biology, University of Coimbra (CNBC-UC), 3004-504 Coimbra, Portugal.
| | - S M Monteiro
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB and Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal; Department of Biology and Environment, Life Sciences and Environment School, University of Trás-os-Montes e Alto Douro, Apt. 1013, 5000-801 Vila Real, Portugal.
| |
Collapse
|
7
|
Abotalebi H, Ebrahimi B, Shahriyari R, Shafieian R. Sex steroids-induced neurogenesis in adult brain: a better look at mechanisms and mediators. Horm Mol Biol Clin Investig 2021; 42:209-221. [PMID: 34058796 DOI: 10.1515/hmbci-2020-0036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 01/14/2021] [Indexed: 11/15/2022]
Abstract
Adult neurogenesis is the production of new nerve cells in the adult brain. Neurogenesis is a clear example of the neuroplasticity phenomenon which can be observed in most of mammalian species, including human beings. This phenomenon occurs, at least, in two regions of the brain: the subgranular zone of the dentate gyrus in hippocampus and the ventricular zone of lateral ventricles. Numerous studies have investigated the relationship between sex steroid hormones and neurogenesis of adult brain; of which, mostly concentrated on the role of estradiol. It has been shown that estrogen plays a significant role in this process through both classic and non-classic mechanisms, including a variety of different growth factors. Therefore, the objective of this review is to investigate the role of female sex steroids with an emphasis on estradiol and also its potential implications for regulating the neurogenesis in the adult brain.
Collapse
Affiliation(s)
- Hamideh Abotalebi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Raziyeh Shahriyari
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Shafieian
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Jiang H, Xiao L, Jin K, Shao B. Estrogen administration attenuates post-stroke depression by enhancing CREB/BDNF/TrkB signaling in the rat hippocampus. Exp Ther Med 2021; 21:433. [PMID: 33747172 PMCID: PMC7967838 DOI: 10.3892/etm.2021.9850] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/09/2020] [Indexed: 12/27/2022] Open
Abstract
A previous study demonstrated that 17β-estradiol (E2), which is an antidepressant, can ameliorate post-stroke depression (PSD); however, the underlying mechanisms governing this remain largely unknown. Therefore, the present study developed a PSD model in rats, which was induced by left middle cerebral artery occlusion followed by exposure to chronic mild stress for 2 weeks. The results revealed that the activity of the cAMP response element-binding protein (CREB), a cellular transcription factor, and the associated brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) signaling were all attenuated in the hippocampus in PSD rats. The depression-like behaviors were significantly improved after treatment with E2, along with increased CREB and the BDNF/TrkB signaling activity. These results provide novel insight into the molecular basis of PSD, and suggest the potential involvement of CREB/BDNF/TrkB signaling in E2-mediated improvement of PSD in rats.
Collapse
Affiliation(s)
- Huigang Jiang
- Department of Neurology, Yiwu City Center Hospital, Wenzhou Medical University, Yiwu, Zhejiang 322000, P.R. China
| | - Li Xiao
- Department of Neurology, Shaoyang City Center Hospital, Shaoyang, Hunan 422000, P.R. China
| | - Kunlin Jin
- Department of Neurology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
9
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
10
|
Chronic adolescent stress causes sustained impairment of cognitive flexibility and hippocampal synaptic strength in female rats. Neurobiol Stress 2021; 14:100303. [PMID: 33614865 PMCID: PMC7876631 DOI: 10.1016/j.ynstr.2021.100303] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/13/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Females that experience chronic stress during development, particularly adolescence, are the most vulnerable group to stress-induced disease. While considerable attention has been devoted to stress-induced manifestation of anxiety, depression, and PTSD, evidence indicates that a history of chronic stress is also a risk factor for cognitive decline and dementia - with females again in a higher risk group. This interplay between sex and stress history indicates specific mechanisms drive neural dysfunction across the lifespan. The presence of sex and stress steroid receptors in the hippocampus provides a point of influence for these variables to drive changes in cognitive function. Here, we used a rodent model of chronic adolescent stress (CAS) to determine the extent to which CAS modifies glutamatergic signaling resulting in cognitive dysfunction. Male and female Wistar rats born in-house remained non-stressed (NS), unmanipulated aside from standard cage cleaning, or were exposed to either physical restraint (60 min) or social defeat (CAS) each day (6 trials each), along with social isolation, throughout the adolescent period (PND 35-47). Cognition was assessed in adult (PND 80-130) male and female rats (n = 10-12) using the Barnes Maze task and the Attention Set-Shift task. Whole hippocampi were extracted from a second cohort of male and female rats (NS and CAS; n = 9-10) and processed for RNA sequencing. Brain tissue from the first cohort (n = 6) was processed for density of glutamatergic synaptic markers (GluA1, NMDA1a, and synaptophysin) or whole-cell patch clamping (n = 4) to determine glutamatergic activity in the hippocampus. Females with a history of chronic stress had shorter latencies to locate the goal box than NS controls during acquisition learning but showed an increased latency to locate the new goal box during reversal learning. This reversal deficit persisted across domains as females with a history of stress required more trials to reach criterion during the reversal phases of the Attention Set-Shift task compared to controls. Ovariectomy resulted in greater performance variability overall during reversal learning with CAS females showing worse performance. Males showed no effects of CAS history on learning or memory performance. Bioinformatic prediction using gene ontology categorization indicated that in females, postsynaptic membrane gene clusters, specifically genes related to glutamatergic synapse remodeling, were enriched with a history of stress. Structural analysis indicated that CAS did not alter glutamate receptor density in females. However, functionally, CAS females had a decreased AMPA/NMDA-dependent current ratio compared to controls indicating a weakening in synaptic strength in the hippocampus. Males showed only a slight change in density of NMDA1a labeling in the CA3 region with a history of stress. The data observed here suggest that females are at risk for impaired cognitive flexibility following a history of adolescent stress, possibly driven by changes in glutamatergic signaling.
Collapse
|
11
|
Sardar R, Zandieh Z, Namjoo Z, Soleimani M, Shirazi R, Hami J. Laterality and sex differences in the expression of brain-derived neurotrophic factor in developing rat hippocampus. Metab Brain Dis 2021; 36:133-144. [PMID: 32975719 PMCID: PMC8206061 DOI: 10.1007/s11011-020-00620-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023]
Abstract
Brain-derived neurotrophic factor (BDNF), as a member of neurotrophin family, plays an important role in neurogenesis, neuronal survival and synaptic plasticity. BDNF is strongly expressed in the hippocampus, where has been associated with memory consolidation, learning, and cognition. In this study, Real-time PCR, immunohistochemistry, and stereology were used to evaluate the gender differences and left-right asymmetries in the expression of BDNF in the developing rat hippocampus during the neurogenesis-active period, at postnatal days P0, P7 and P14. We found the lowest expression of BDNF in the right side and the highest in the left side hippocampi of both male and female neonates at P14 (P ≤ 0.05 each). At the same time, there were significant differences in the hippocampal expression of BDNF between males and females (P ≤ 0.05 each). No important differences in the number of BDNF expressing neurons in different subregions of right/left hippocampus were observed between male and female animals at P0 and P7 (P > 0.05). Furthermore, the highest numerical density of BDNF positive cells was detected in the both sides hippocampal CA1 in the male/female offspring at P7, and in the CA2, CA3 and dentate gyrus at P14 (P ≤ 0.05 each). Based on these findings, it can be concluded that there are prominent sex and interhemispheric differences in the expression of BDNF in the developing rat hippocampus, suggesting a probable mechanism for the control of gender and laterality differences in development, structure, and function of the hippocampus.
Collapse
Affiliation(s)
- Reza Sardar
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Namjoo
- Department of Anatomical Science, school of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mansoureh Soleimani
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Shirazi
- Department of Health and Medical Sciences, Faculty of Health, Arts and Design, Swinburn University, Hawthorn, Melbourne, Australia
| | - Javad Hami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
- Institute for Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany.
| |
Collapse
|
12
|
Trova S, Bovetti S, Pellegrino G, Bonzano S, Giacobini P, Peretto P. HPG-Dependent Peri-Pubertal Regulation of Adult Neurogenesis in Mice. Front Neuroanat 2020; 14:584493. [PMID: 33328903 PMCID: PMC7732626 DOI: 10.3389/fnana.2020.584493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/29/2020] [Indexed: 11/13/2022] Open
Abstract
Adult neurogenesis, a striking form of neural plasticity, is involved in the modulation of social stimuli driving reproduction. Previous studies on adult neurogenesis have shown that this process is significantly modulated around puberty in female mice. Puberty is a critical developmental period triggered by increased secretion of the gonadotropin releasing hormone (GnRH), which controls the activity of the hypothalamic-pituitary-gonadal axis (HPG). Secretion of HPG-axis factors at puberty participates to the refinement of neural circuits that govern reproduction. Here, by exploiting a transgenic GnRH deficient mouse model, that progressively loses GnRH expression during postnatal development (GnRH::Cre;Dicer loxP/loxP mice), we found that a postnatally-acquired dysfunction in the GnRH system affects adult neurogenesis selectively in the subventricular-zone neurogenic niche in a sexually dimorphic way. Moreover, by examining adult females ovariectomized before the onset of puberty, we provide important evidence that, among the HPG-axis secreting factors, the circulating levels of gonadal hormones during pre-/peri-pubertal life contribute to set-up the proper adult subventricular zone-olfactory bulb neurogenic system.
Collapse
Affiliation(s)
- Sara Trova
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy.,Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Giuliana Pellegrino
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| | - Paolo Giacobini
- Univ.Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, Laboratory of the Development and Plasticity of Neuroendocrine Brain, Lille, France
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Orbassano, Italy
| |
Collapse
|
13
|
Jorgensen C, Wang Z. Hormonal Regulation of Mammalian Adult Neurogenesis: A Multifaceted Mechanism. Biomolecules 2020; 10:biom10081151. [PMID: 32781670 PMCID: PMC7465680 DOI: 10.3390/biom10081151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis—resulting in adult-generated functioning, integrated neurons—is still one of the most captivating research areas of neuroplasticity. The addition of new neurons in adulthood follows a seemingly consistent multi-step process. These neurogenic stages include proliferation, differentiation, migration, maturation/survival, and integration of new neurons into the existing neuronal network. Most studies assessing the impact of exogenous (e.g., restraint stress) or endogenous (e.g., neurotrophins) factors on adult neurogenesis have focused on proliferation, survival, and neuronal differentiation. This review will discuss the multifaceted impact of hormones on these various stages of adult neurogenesis. Specifically, we will review the evidence for hormonal facilitation (via gonadal hormones), inhibition (via glucocorticoids), and neuroprotection (via recruitment of other neurochemicals such as neurotrophin and neuromodulators) on newly adult-generated neurons in the mammalian brain.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, UT 84058, USA
- Correspondence:
| | - Zuoxin Wang
- Psychology Department and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
14
|
Sex Differences in Maturation and Attrition of Adult Neurogenesis in the Hippocampus. eNeuro 2020; 7:ENEURO.0468-19.2020. [PMID: 32586842 PMCID: PMC7369314 DOI: 10.1523/eneuro.0468-19.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sex differences exist in the regulation of adult neurogenesis in the hippocampus in response to hormones and cognitive training. Here, we investigated the trajectory and maturation rate of adult-born neurons in the dentate gyrus (DG) of male and female rats. Sprague Dawley rats were perfused 2 h, 24 h, one week (1w), 2w, or 3w after bromodeoxyuridine (BrdU) injection, a DNA synthesis marker that labels dividing progenitor cells and their progeny. Adult-born neurons (BrdU/NeuN-ir) matured faster in males compared with females. Males had a greater density of neural stem cells (Sox2-ir) in the dorsal, but not in the ventral, DG and had higher levels of cell proliferation (Ki67-ir) than non-proestrous females. However, males showed a greater reduction in neurogenesis between 1week and 2weeks after mitosis, whereas females showed similar levels of neurogenesis throughout the weeks. The faster maturation and greater attrition of new neurons in males compared with females suggests greater potential for neurogenesis to respond to external stimuli in males and emphasizes the importance of studying sex on adult hippocampal neurogenesis.
Collapse
|
15
|
Duarte-Guterman P, Lieblich SE, Qiu W, Splinter JEJ, Go KA, Casanueva-Reimon L, Galea LAM. Oxytocin has sex-specific effects on social behaviour and hypothalamic oxytocin immunoreactive cells but not hippocampal neurogenesis in adult rats. Horm Behav 2020; 122:104734. [PMID: 32169583 DOI: 10.1016/j.yhbeh.2020.104734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/06/2020] [Accepted: 02/28/2020] [Indexed: 10/24/2022]
Abstract
Oxytocin regulates social behaviours, pair bonding and hippocampal neurogenesis but most studies have used adult males. Our study investigated the effects of oxytocin on social investigation and adult hippocampal neurogenesis in male and female rats. Oxytocin has poor penetration of the blood-brain barrier, therefore we tested a nanoparticle drug, TRIOZAN™ (Ovensa Inc.), which permits greater blood-brain-barrier penetration. Adult male and female rats were injected daily (i.p.) for 10 days with either: oxytocin in PBS (0.5 or 1.0 mg/kg), oxytocin in TRIOZAN™ (0.5 or 1.0 mg/kg), or vehicle (PBS) and tested for social investigation. Oxytocin decreased body mass and increased social investigation and number of oxytocin-immunoreactive cells in the supraoptic nucleus (SON) of the hypothalamus in male rats only. In both sexes, oxytocin decreased the number of immature neurons (doublecortin+ cells) in the ventral hippocampus and reduced plasma 17β-estradiol levels in a dose- and delivery-dependent way. Oxytocin in TRIOZAN™ reduced "sedation" observed post-injection and increased certain central effects (oxytocin levels in the hypothalamus and neurogenesis in the ventral hippocampus) relative to oxytocin in PBS, indicating that the nanoparticle may be used as an alternative brain delivery system. We showed that oxytocin has sex-specific effects on social investigation, body mass, "sedation", and the oxytocin system. In contrast, similar effects were observed in both sexes in neurogenesis and plasma 17β-estradiol. Our work suggests that sex differences in oxytocin regulation of brain endpoints is region-specific (hypothalamus versus hippocampus) and that oxytocin does not promote social investigation in females.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Jared E J Splinter
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Kimberly A Go
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Laura Casanueva-Reimon
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
16
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
17
|
Konishi K, Cherkerzian S, Aroner S, Jacobs EG, Rentz DM, Remington A, Aizley H, Hornig M, Klibanski A, Goldstein JM. Impact of BDNF and sex on maintaining intact memory function in early midlife. Neurobiol Aging 2019; 88:137-149. [PMID: 31948671 DOI: 10.1016/j.neurobiolaging.2019.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/30/2019] [Accepted: 12/14/2019] [Indexed: 01/06/2023]
Abstract
Sex steroid hormones and neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), play a significant neuroprotective role in memory circuitry aging. Here, we present findings characterizing the neuroprotective effects of BDNF on memory performance, as a function of sex and reproductive status in women. Participants (N = 191; mean age = 50.03 ± 2.10) underwent clinical and cognitive testing, fMRI scanning, and hormonal assessments of menopausal staging. Memory performance was assessed with the 6-Trial Selective Reminding Test and the Face-Name Associative Memory Exam. Participants also performed a working memory (WM) N-back task during fMRI scanning. Results revealed significant interactions between menopausal status and BDNF levels. Only in postmenopausal women, lower plasma BDNF levels were associated with significantly worse memory performance and altered function in the WM circuitry. BDNF had no significant impact on memory performance or WM function in pre/perimenopausal women or men. These results suggest that in postmenopausal women, BDNF is associated with memory performance and memory circuitry function, thus providing evidence of potential sex-dependent factors of risk and resilience for early intervention.
Collapse
Affiliation(s)
- Kyoko Konishi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara Cherkerzian
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sarah Aroner
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Dorene M Rentz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Remington
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harlyn Aizley
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mady Hornig
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Anne Klibanski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill M Goldstein
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Wang S, Zheng Z. Differential cell proliferation and cell death during the urethral groove formation in guinea pig model. Pediatr Res 2019; 86:452-459. [PMID: 30467344 DOI: 10.1038/s41390-018-0236-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/28/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Urethral groove (UG) formation is an important step in penile formation. Because commonly used animal models do not have UG, the mechanisms of UG formation have never been discovered. We aim to discover the cellular mechanism of the UG formation using guinea pig model. METHODS Histology was used to study the ontogeny of UG. BrdU immunofluorescence was used to label proliferating cells, cell death was determined using LysoTracker Red and TUNEL staining, and stereology was used for quantification. To reveal Shh mRNA expression patterns, in situ hybridization was performed in guinea pig genital tubercles (GTs) and ShhGFPcre-LacZ-reporter mice were used for comparison. RESULTS Cell proliferation in the outer layers and programmed cell death in the inner layers of urethral epithelium played key roles during urethral canal movement from dorsal to ventral aspect and final opening to form UG. Shh mRNA expression domain shifted out to the ventral surface of GT from proximal throughout to distal in guinea pigs, but was excluded from the ventral surface epithelium in midshaft and distal of mouse GT. CONCLUSION Differential cell proliferation and cell death in developing urethral epithelium lead to UG formation and Shh expression in ventral surface epithelium of GT may play an important role.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL, 62901, USA
| | - Zhengui Zheng
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL, 62901, USA.
| |
Collapse
|
19
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
20
|
Sheppard PAS, Choleris E, Galea LAM. Structural plasticity of the hippocampus in response to estrogens in female rodents. Mol Brain 2019; 12:22. [PMID: 30885239 PMCID: PMC6423800 DOI: 10.1186/s13041-019-0442-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 01/05/2023] Open
Abstract
It is well established that estrogens affect neuroplasticity in a number of brain regions. In particular, estrogens modulate and mediate spine and synapse formation as well as neurogenesis in the hippocampal formation. In this review, we discuss current research exploring the effects of estrogens on dendritic spine plasticity and neurogenesis with a focus on the modulating factors of sex, age, and pregnancy. Hormone levels, including those of estrogens, fluctuate widely across the lifespan from early life to puberty, through adulthood and into old age, as well as with pregnancy and parturition. Dendritic spine formation and modulation are altered both by rapid (likely non-genomic) and classical (genomic) actions of estrogens and have been suggested to play a role in the effects of estrogens on learning and memory. Neurogenesis in the hippocampus is influenced by age, the estrous cycle, pregnancy, and parity in female rodents. Furthermore, sex differences exist in hippocampal cellular and molecular responses to estrogens and are briefly discussed throughout. Understanding how structural plasticity in the hippocampus is affected by estrogens and how these effects can influence function and be influenced by other factors, such as experience and sex, is critical and can inform future treatments in conditions involving the hippocampus.
Collapse
Affiliation(s)
- Paul A. S. Sheppard
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Elena Choleris
- Department of Psychology & Neuroscience Program, University of Guelph, Guelph, ON N1G 2W1 Canada
| | - Liisa A. M. Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
21
|
Hyer MM, Phillips LL, Neigh GN. Sex Differences in Synaptic Plasticity: Hormones and Beyond. Front Mol Neurosci 2018; 11:266. [PMID: 30108482 PMCID: PMC6079238 DOI: 10.3389/fnmol.2018.00266] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/13/2018] [Indexed: 12/24/2022] Open
Abstract
Notable sex-differences exist between neural structures that regulate sexually dimorphic behaviors such as reproduction and parenting. While anatomical differences have been well-characterized, advancements in neuroimaging and pharmacology techniques have allowed researchers to identify differences between males and females down to the level of the synapse. Disparate mechanisms at the synaptic level contribute to sex-specific neuroplasticity that is reflected in sex-dependent behaviors. Many of these synaptic differences are driven by the endocrine system and its impact on molecular signaling and physiology. While sex-dependent modifications exist at baseline, further differences emerge in response to stimuli such as stressors. While some of these mechanisms are unifying between sexes, they often have directly opposing consequences in males and females. This variability is tied to gonadal steroids and their interactions with intra- and extra-cellular signaling mechanisms. This review article focuses on the various mechanisms by which sex can alter synaptic plasticity, both directly and indirectly, through steroid hormones such as estrogen and testosterone. That sex can drive neuroplasticity throughout the brain, highlights the importance of understanding sex-dependent neural mechanisms of the changing brain to enhance interpretation of results regarding males and females. As mood and stress responsivity are characterized by significant sex-differences, understanding the molecular mechanisms that may be altering structure and function can improve our understanding of these behavioral and mental characteristics.
Collapse
Affiliation(s)
- Molly M Hyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
22
|
Geary DC. Evolutionary perspective on sex differences in the expression of neurological diseases. Prog Neurobiol 2018; 176:33-53. [PMID: 29890214 DOI: 10.1016/j.pneurobio.2018.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 04/25/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022]
Abstract
Sex-specific brain and cognitive deficits emerge with malnutrition, some infectious and neurodegenerative diseases, and often with prenatal or postnatal toxin exposure. These deficits are described in disparate literatures and are generally not linked to one another. Sexual selection may provide a unifying framework that integrates our understanding of these deficits and provides direction for future studies of sex-specific vulnerabilities. Sexually selected traits are those that have evolved to facilitate competition for reproductive resources or that influence mate choices, and are often larger and more complex than other traits. Critically, malnutrition, disease, chronic social stress, and exposure to man-made toxins compromise the development and expression of sexually selected traits more strongly than that of other traits. The fundamental mechanism underlying vulnerability might be the efficiency of mitochondrial energy capture and control of oxidative stress that in turn links these traits to current advances in neuroenergetics, stress endocrinology, and toxicology. The key idea is that the elaboration of these cognitive abilities, with more underlying gray matter or more extensive inter-modular white matter connections, makes them particularly sensitive to disruptions in mitochondrial functioning and oxidative stress. A framework of human sexually selected cognitive abilities and underlying brain systems is proposed and used to organize what is currently known about sex-specific vulnerabilities.
Collapse
Affiliation(s)
- David C Geary
- Department of Psychological Sciences, Interdisciplinary Neuroscience, University of Missouri, MO, 65211-2500, Columbia, United States.
| |
Collapse
|
23
|
Sadaie MR, Farhoudi M, Zamanlu M, Aghamohammadzadeh N, Amouzegar A, Rosenbaum RE, Thomas GA. What does the research say about androgen use and cerebrovascular events? Ther Adv Drug Saf 2018; 9:439-455. [PMID: 30364888 DOI: 10.1177/2042098618773318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
Many studies have investigated the benefits of androgen therapy and neurosteroids in aging men, while concerns remain about the potential associations of exogenous steroids and incidents of cerebrovascular events and ischemic stroke (IS). Testosterone is neuroprotective, neurotrophic and a potent stimulator of neuroplasticity. These benefits are mediated primarily through conversion of a small amount of testosterone to estradiol by the catalytic activity of estrogen synthetase (aromatase cytochrome P450 enzyme). New studies suggest that abnormal serum levels of the nonaromatized potent metabolite of testosterone, either high or low dihydrotestosterone (DHT), is a risk factor for stroke. Associations between pharmacologic androgen use and the incidence of IS are questionable, because a significant portion of testosterone is converted to DHT. There is also insufficient evidence to reject a causal relationship between the pro-testosterone adrenal androgens and incidence of IS. Moreover, vascular intima-media thickness, which is a predictor of stroke and myocardial symptoms, has correlations with sex hormones. Current diagnostic and treatment criteria for androgen therapy for cerebrovascular complications are unclear. Confounding variables, including genetic and metabolic alterations of the key enzymes of steroidogenesis, ought to be considered. Information extracted from pharmacogenetic testing may aid in expounding the protective-destructive properties of neurosteroids, as well as the prognosis of androgen therapy, in particular their cerebrovascular outcomes. This investigative review article addresses relevant findings of the clinical and experimental investigations of androgen therapy, emphasizes the significance of genetic testing of androgen responsiveness towards individualized therapy in post-IS injuries as well as identifying pertinent questions.
Collapse
Affiliation(s)
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masumeh Zamanlu
- Neurosciences Research Center (NSRC), Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Aghamohammadzadeh
- Department of Endocrinology, Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atieh Amouzegar
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Gary A Thomas
- Penn State Hershey Neurology, Penn State University, PA, USA
| |
Collapse
|
24
|
Gheorghe A, Qiu W, Galea LAM. Hormonal Regulation of Hippocampal Neurogenesis: Implications for Depression and Exercise. Curr Top Behav Neurosci 2018; 43:379-421. [PMID: 30414016 DOI: 10.1007/7854_2018_62] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adult hippocampal neurogenesis exists in all mammalian species, including humans, and although there has been considerable research investigating the function and regulation of neurogenesis, there remain many open questions surrounding the complexity of this phenomenon. This stems partially from the fact that neurogenesis is a multistage process that involves proliferation, differentiation, migration, survival, and eventual integration of new cells into the existing hippocampal circuitry, each of which can be independently influenced. The function of adult neurogenesis in the hippocampus is related to stress regulation, behavioral efficacy of antidepressants, long-term spatial memory, forgetting, and pattern separation. Steroid hormones influence the regulation of hippocampal neurogenesis, stress regulation, and cognition and differently in males and females. In this chapter, we will briefly tap into the complex network of steroid hormone modulation of neurogenesis in the hippocampus with specific emphasis on stress, testosterone, and estrogen. We examine the possible role of neurogenesis in the etiology of depression and influencing treatment by examining the influence of both pharmacological (selective serotonin reuptake inhibitors, tricyclic antidepressants) treatments and non-pharmacological (exercise) remedies.
Collapse
Affiliation(s)
- Ana Gheorghe
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada. .,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada. .,Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
25
|
Hyer MM, Khantsis S, Venezia AC, Madison FN, Hallgarth L, Adekola E, Glasper ER. Estrogen-dependent modifications to hippocampal plasticity in paternal California mice (Peromyscus californicus). Horm Behav 2017; 96:147-155. [PMID: 28954216 DOI: 10.1016/j.yhbeh.2017.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 11/22/2022]
Abstract
In many biparental species, mothers and fathers experience similar modifications to circulating hormones. With these modifications come alterations in neural structure and function suggesting that neuroendocrine mechanisms may underlie postpartum plasticity in both males and females. In the biparental California mouse (Peromyscus californicus), adult neurogenesis is maintained and anxiety-like behavior is attenuated in fathers during the mid-postpartum period. Given a causal relationship between estrogen and regulation of both adult neurogenesis and anxiety, we aimed to elucidate the role of estrogen-dependent mechanisms in paternal experience-related modifications to hippocampal neuroplasticity in California mice. In Experiment 1, hippocampal estrogen receptor beta (ERβ) mRNA expression, along with circulating estradiol concentrations, were determined throughout the postpartum period. An upregulation in ERβ expression was observed in postnatal day 16 males compared to virgins. Additionally, a rise in circulating estradiol concentrations was detected on postnatal day 2 compared to virgins; levels began to decline toward virgin levels on postnatal day 16 and postnatal day 30. In Experiment 2, we determined the role of estrogen-dependent mechanisms in adult neurogenesis and anxiety-like behavior by treating virgin and paternal males with saline or the selective estrogen receptor modulator, tamoxifen (TMX), during the time of axon extension (i.e., one week after bromodeoxyuridine injection). While TMX failed to alter elevated plus maze performance, TMX treatment inhibited survival of adult born neurons but only in paternal mice. These findings highlight the potential for estrogen-dependent pathways to mediate hippocampal adult neurogenesis in paternal mice.
Collapse
Affiliation(s)
- Molly M Hyer
- Program in Neuroscience and Cognitive Sciences, University of Maryland, College Park, MD 20742, USA
| | - Sabina Khantsis
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - Andrew C Venezia
- Program in Neuroscience and Cognitive Sciences, University of Maryland, College Park, MD 20742, USA
| | - Farrah N Madison
- Program in Neuroscience and Cognitive Sciences, University of Maryland, College Park, MD 20742, USA; Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - Luke Hallgarth
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - Enoch Adekola
- Department of Psychology, University of Maryland, College Park, MD 20742, USA
| | - Erica R Glasper
- Program in Neuroscience and Cognitive Sciences, University of Maryland, College Park, MD 20742, USA; Department of Psychology, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
26
|
Abdyazdani N, Nourazarian A, Nozad Charoudeh H, Kazemi M, Feizy N, Akbarzade M, Mehdizadeh A, Rezaie J, Rahbarghazi R. The role of morphine on rat neural stem cells viability, neuro-angiogenesis and neuro-steroidgenesis properties. Neurosci Lett 2017; 636:205-212. [PMID: 27845244 DOI: 10.1016/j.neulet.2016.11.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 11/16/2022]
Abstract
A lack of comprehensive data exists on the effect of morphine on neural stem cell neuro-steroidogenesis and neuro-angiogenesis properties. We, herein, investigated the effects of morphine (100μM), naloxone (100μM) and their combination on rat neural stem cells viability, clonogenicity and Ki-67 expression over a period of 72h. Any alterations in the total fatty acids profile under treatment protocols were elucidated by direct transesterification method. We also monitored the expression of p53, aromatase and 5-alpha reductase by real-time PCR assay. To examine angiogenic capacity, in vitro tubulogenesis and the level of VE-cadherin transcript were investigated during neural to endothelial differentiation under the experimental procedure. Cells supplemented with morphine displayed reduced survival (p<0.01) and clonogenicity (p<0.001). Flow cytometric analysis showed a decrease in Ki-67 during 72h. Naloxone potentially blunted morphine-induced all effects. The normal levels of fatty acids, including saturated and unsaturated were altered by naloxone and morphine supplements. Following 48h, the up-regulation of p53, aromatase and 5-alpha reductase genes occurred in morphine-primed cells. Using three-dimensional culture models of angiogenesis and real time PCR assay, we showed morphine impaired the tubulogenesis properties of neural stem cells (p<0.001) by the inhibition of trans-differentiation into vascular cells and led to decrease of in VE-cadherin expression. Collectively, morphine strongly impaired the healthy status of neural stem cells by inducing p53 and concurrent elevation of aromatase and 5-alpha reductase activities especially during early 48h. Also, neural stem cells-being exposed to morphine lost their potency to elicit angiogenesis.
Collapse
Affiliation(s)
- Nima Abdyazdani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Masoumeh Kazemi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Feizy
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Akbarzade
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
27
|
Geary DC. Evolutionary framework for identifying sex- and species-specific vulnerabilities in brain development and functions. J Neurosci Res 2016; 95:355-361. [DOI: 10.1002/jnr.23794] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 01/20/2023]
Affiliation(s)
- David C. Geary
- Department of Psychological Sciences and Interdisciplinary Neuroscience Program; University of Missouri; Columbia Missouri
| |
Collapse
|
28
|
Pellegrini E, Diotel N, Vaillant-Capitaine C, Pérez Maria R, Gueguen MM, Nasri A, Cano Nicolau J, Kah O. Steroid modulation of neurogenesis: Focus on radial glial cells in zebrafish. J Steroid Biochem Mol Biol 2016; 160:27-36. [PMID: 26151741 DOI: 10.1016/j.jsbmb.2015.06.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/01/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
Estrogens are known as steroid hormones affecting the brain in many different ways and a wealth of data now document effects on neurogenesis. Estrogens are provided by the periphery but can also be locally produced within the brain itself due to local aromatization of circulating androgens. Adult neurogenesis is described in all vertebrate species examined so far, but comparative investigations have brought to light differences between vertebrate groups. In teleost fishes, the neurogenic activity is spectacular and adult stem cells maintain their mitogenic activity in many proliferative areas within the brain. Fish are also quite unique because brain aromatase expression is limited to radial glia cells, the progenitor cells of adult fish brain. The zebrafish has emerged as an interesting vertebrate model to elucidate the cellular and molecular mechanisms of adult neurogenesis, and notably its modulation by steroids. The main objective of this review is to summarize data related to the functional link between estrogens production in the brain and neurogenesis in fish. First, we will demonstrate that the brain of zebrafish is an endogenous source of steroids and is directly targeted by local and/or peripheral steroids. Then, we will present data demonstrating the progenitor nature of radial glial cells in the brain of adult fish. Next, we will emphasize the role of estrogens in constitutive neurogenesis and its potential contribution to the regenerative neurogenesis. Finally, the negative impacts on neurogenesis of synthetic hormones used in contraceptive pills production and released in the aquatic environment will be discussed.
Collapse
Affiliation(s)
- Elisabeth Pellegrini
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France.
| | - Nicolas Diotel
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Inserm UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde F-97490, France; Université de La Réunion, UMR 1188, Sainte-Clotilde F-97490, France
| | - Colette Vaillant-Capitaine
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Rita Pérez Maria
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Laboratorio de Ictiología, Instituto Nacional de Limnología (INALI. CONICET-UNL), Paraje El Pozo, Ciudad Universitaria UNL, 3000 Santa Fe, Argentina
| | - Marie-Madeleine Gueguen
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Ahmed Nasri
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Laboratoire de Biosurveillance de l'Environnement, Unité d'Ecologie côtière et d'Ecotoxicologie, Faculté des Sciences de Bizerte, Zarzouna 7021, Tunisia
| | - Joel Cano Nicolau
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Olivier Kah
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| |
Collapse
|
29
|
Trading new neurons for status: Adult hippocampal neurogenesis in eusocial Damaraland mole-rats. Neuroscience 2016; 324:227-37. [DOI: 10.1016/j.neuroscience.2016.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/19/2016] [Accepted: 03/07/2016] [Indexed: 11/21/2022]
|
30
|
Yuen EY, Wei J, Yan Z. Estrogen in prefrontal cortex blocks stress-induced cognitive impairments in female rats. J Steroid Biochem Mol Biol 2016; 160:221-6. [PMID: 26321384 PMCID: PMC4769981 DOI: 10.1016/j.jsbmb.2015.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/20/2015] [Accepted: 08/24/2015] [Indexed: 11/15/2022]
Abstract
Animal and human studies have found that males and females show distinct stress responses. Recent studies suggest the contribution of estrogen in the brain to this sexual dimorphism. Repeated stress has been found to impair cognitive behaviors via suppressing glutamatergic transmission and glutamate receptor surface expression in pyramidal neurons of prefrontal cortex (PFC) in male rats. On the contrary, female rats exposed to the same stress paradigms show normal synaptic function and PFC-mediated cognition. The level of aromatase, the enzyme for the biosynthesis of estrogen, is significantly higher in the PFC of females than males. The stress-induced glutamatergic deficits and memory impairment are unmasked by blocking estrogen receptors or aromatase in females, suggesting a protective role of estrogen against the detrimental effects of repeated stress.
Collapse
Affiliation(s)
- Eunice Y Yuen
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Jing Wei
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA.
| |
Collapse
|
31
|
Mahmoud R, Wainwright SR, Galea LAM. Sex hormones and adult hippocampal neurogenesis: Regulation, implications, and potential mechanisms. Front Neuroendocrinol 2016; 41:129-52. [PMID: 26988999 DOI: 10.1016/j.yfrne.2016.03.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 11/16/2022]
Abstract
Neurogenesis within the adult hippocampus is modulated by endogenous and exogenous factors. Here, we review the role of sex hormones in the regulation of adult hippocampal neurogenesis in males and females. The review is framed around the potential functional implications of sex hormone regulation of adult hippocampal neurogenesis, with a focus on cognitive function and mood regulation, which may be related to sex differences in incidence and severity of dementia and depression. We present findings from preclinical studies of endogenous fluctuations in sex hormones relating to reproductive function and ageing, and from studies of exogenous hormone manipulations. In addition, we discuss the modulating roles of sex, age, and reproductive history on the relationship between sex hormones and neurogenesis. Because sex hormones have diverse targets in the central nervous system, we overview potential mechanisms through which sex hormones may influence hippocampal neurogenesis. Lastly, we advocate for a more systematic consideration of sex and sex hormones in studying the functional implications of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Rand Mahmoud
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Steven R Wainwright
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada; Centre for Brain Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
32
|
Social behavior, hormones and adult neurogenesis. Front Neuroendocrinol 2016; 41:71-86. [PMID: 26996817 DOI: 10.1016/j.yfrne.2016.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 01/31/2023]
Abstract
A variety of experiences have been shown to affect the production of neurons in the adult hippocampus. These effects may be mediated by experience-driven hormonal changes, which, in turn, interact with factors such as sex, age and life history to alter brain plasticity. Although the effects of physical experience and stress have been extensively characterized, various types of social experience across the lifespan trigger profound neuroendocrine changes in parallel with changes in adult neurogenesis. This review article focuses on the influence of specific social experiences on adult neurogenesis in the dentate gyrus and the potential role of hormones in these effects.
Collapse
|
33
|
Zhang Z, Hong J, Zhang S, Zhang T, Sha S, Yang R, Qian Y, Chen L. Postpartum estrogen withdrawal impairs hippocampal neurogenesis and causes depression- and anxiety-like behaviors in mice. Psychoneuroendocrinology 2016; 66:138-49. [PMID: 26803529 DOI: 10.1016/j.psyneuen.2016.01.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022]
Abstract
Postpartum estrogen withdrawal is known to be a particularly vulnerable time for depressive symptoms. Ovariectomized adult mice (OVX-mice) treated with hormone-simulated pregnancy (HSP mice) followed by a subsequent estradiol benzoate (EB) withdrawal (EW mice) exhibited depression- and anxiety-like behaviors, as assessed by forced swim, tail suspension and elevated plus-maze, while HSP mice, OVX mice or EB-treated OVX mice (OVX/EB mice) did not. The survival and neurite growth of newborn neurons in hippocampal dentate gyrus were examined on day 5 after EW. Compared with controls, the numbers of 28-day-old BrdU(+) and BrdU(+)/NeuN(+) cells were increased in HSP mice but significantly decreased in EW mice; the numbers of 10-day-old BrdU(+) cells were increased in HSP mice and OVX/EB mice; and the density of DCX(+) fibers was reduced in EW mice and OVX mice. The phosphorylation of hippocampal NMDA receptor (NMDAr) NR2B subunit or Src was increased in HSP mice but decreased in EW mice. NMDAr agonist NMDA prevented the loss of 28-day-old BrdU(+) cells and the depression- and anxiety-like behaviors in EW mice. NR2B inhibitor Ro25-6981 or Src inhibitor dasatinib caused depression- and anxiety-like behaviors in HSP mice with the reduction of 28-day-old BrdU(+) cells. The hippocampal BDNF levels were reduced in EW mice and OVX mice. TrkB receptor inhibitor K252a reduced the density of DCX(+) fibers in HSP mice without the reduction of 28-day-old BrdU(+) cells, or the production of affective disorder. Collectively, these results indicate that postpartum estrogen withdrawal impairs hippocampal neurogenesis in mice that show depression- and anxiety-like behaviors.
Collapse
Affiliation(s)
- Zhuan Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China; Department of Anesthesiology, Jiangsu Province Hospital, Nanjing Medical University, Nanjing 86025, China
| | - Juan Hong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China; Department of Physiology, Nanjing Medical University, Nanjing 86025, China
| | - Suyun Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China
| | - Tingting Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China; Department of Physiology, Nanjing Medical University, Nanjing 86025, China
| | - Sha Sha
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China; Department of Physiology, Nanjing Medical University, Nanjing 86025, China
| | - Rong Yang
- Department of Obstetrics and Gynecology, Hangzhou First People's Hospital, Hangzhou 860571, China
| | - Yanning Qian
- Department of Anesthesiology, Jiangsu Province Hospital, Nanjing Medical University, Nanjing 86025, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 86025, China; Department of Physiology, Nanjing Medical University, Nanjing 86025, China.
| |
Collapse
|
34
|
Gender Differences in the Neurobiology of Anxiety: Focus on Adult Hippocampal Neurogenesis. Neural Plast 2016; 2016:5026713. [PMID: 26885403 PMCID: PMC4738969 DOI: 10.1155/2016/5026713] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/30/2015] [Accepted: 12/06/2015] [Indexed: 12/14/2022] Open
Abstract
Although the literature reports a higher incidence of anxiety disorders in women, the majority of basic research has focused on male rodents, thus resulting in a lack of knowledge on the neurobiology of anxiety in females. Bridging this gap is crucial for the design of effective translational interventions in women. One of the key brain mechanisms likely to regulate anxious behavior is adult hippocampal neurogenesis (AHN). This review paper aims to discuss the evidence on the differences between male and female rodents with regard to anxiety-related behavior and physiology, with a special focus on AHN. The differences between male and female physiologies are greatly influenced by hormonal differences. Gonadal hormones and their fluctuations during the estrous cycle have often been identified as agents responsible for sexual dimorphism in behavior and AHN. During sexual maturity, hormone levels fluctuate cyclically in females more than in males, increasing the stress response and the susceptibility to anxiety. It is therefore of great importance that future research investigates anxiety and other neurophysiological aspects in the female model, so that results can be more accurately applicable to the female population.
Collapse
|
35
|
Pan Y, Liu Y, Lieberwirth C, Zhang Z, Wang Z. Species differences in behavior and cell proliferation/survival in the adult brains of female meadow and prairie voles. Neuroscience 2015; 315:259-70. [PMID: 26708743 DOI: 10.1016/j.neuroscience.2015.12.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 02/03/2023]
Abstract
Microtine rodents display diverse patterns of social organization and behaviors, and thus provide a useful model for studying the effects of the social environment on physiology and behavior. The current study compared the species differences and the effects of oxytocin (OT) on anxiety-like, social affiliation, and social recognition behaviors in female meadow voles (Microtus pennsylvanicus) and prairie voles (Microtus ochrogaster). Furthermore, cell proliferation and survival in the brains of adult female meadow and prairie voles were compared. We found that female meadow voles displayed a higher level of anxiety-like behavior but lower levels of social affiliation and social recognition compared to female prairie voles. In addition, meadow voles showed lower levels of cell proliferation (measured by Ki67 staining) and cell survival (measured by BrdU staining) in the ventromedial hypothalamus (VMH) and amygdala (AMY), but not the dentate gyrus of the hippocampus (DG), than prairie voles. Interestingly, the numbers of new cells in the VMH and AMY, but not DG, also correlated with anxiety-like, social affiliation, and social recognition behaviors in a brain region-specific manner. Finally, central OT treatment (200 ng/kg, icv) did not lead to changes in behavior or cell proliferation/survival in the brain. Together, these data indicate a potential role of cell proliferation/survival in selected brain areas on different behaviors between vole species with distinct life strategies.
Collapse
Affiliation(s)
- Y Pan
- Program in Molecular and Translational Medicine, School of Medicine, Huzhou University, Huzhou 313000, PR China; State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China; Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - Y Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - C Lieberwirth
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA; Behavioral Science Department, Utah Valley University, Orem, UT, 84058, USA
| | - Z Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China
| | - Z Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA.
| |
Collapse
|
36
|
Sha S, Hong J, Qu WJ, Lu ZH, Li L, Yu WF, Chen L. Sex-related neurogenesis decrease in hippocampal dentate gyrus with depressive-like behaviors in sigma-1 receptor knockout mice. Eur Neuropsychopharmacol 2015; 25:1275-86. [PMID: 25983018 DOI: 10.1016/j.euroneuro.2015.04.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 02/14/2015] [Accepted: 04/24/2015] [Indexed: 11/17/2022]
Abstract
Male sigma-1 receptor knockout (σ1R(-/-)) mice showed depressive-like phenotype with deficit in the survival of newly generated neuronal cells in the hippocampal dentate gyrus (DG), but female σ1R(-/-) mice did not. The level of serum estradiol (E2) at proestrus or diestrus did not differ between female σ1R(-/-) mice and wild-type (WT) mice. Ovariectomized (OVX) female σ1R(-/-) mice, but not WT mice, presented the same depressive-like behaviors and neurogenesis decrease as male σ1R(-/-) mice. Treatment of male σ1R(-/-) mice with E2 could alleviate the depressive-like behaviors and rescue the neurogenesis decrease. In addition, E2 could correct the decline in the density of NMDA-activated current (INMDA) in granular cells of DG and the phosphorylation of NMDA receptor (NMDAr) subtype 2B (NR2B) in male σ1R(-/-) mice, which was associated with the elevation of Src phosphorylation. The neuroprotection and antidepressant effects of E2 in male σ1R(-/-) mice were blocked by the inhibitor of Src or NR2B. The NMDAr agonist showed also the neuroprotection and antidepressant effects in male σ1R(-/-) mice, which were insensitive to the Src inhibitor. On the other hand, either the deprivation of E2 or the inhibition of Src in female σ1R(-/-) mice rather than WT mice led to a distinct decline in INMDA and NR2B phosphorylation. Similarly, the Src inhibitor could cause neurogenesis decrease and depressive-like behaviors in female σ1R(-/-) mice, but not in WT mice. These results indicate that the σ1R deficiency impairs neurogenesis leading to a depressive-like phenotype, which is alleviated by the neuroprotection of E2.
Collapse
Affiliation(s)
- Sha Sha
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China; Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Juan Hong
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Wei-Jun Qu
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Zi-Hong Lu
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Lin Li
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Wen-Feng Yu
- The Key Lab of Molecular Biology, Guiyang Medical University, Guiyang 550004, Guizhou, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China; Department of Physiology, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
37
|
Duarte-Guterman P, Yagi S, Chow C, Galea LAM. Hippocampal learning, memory, and neurogenesis: Effects of sex and estrogens across the lifespan in adults. Horm Behav 2015; 74:37-52. [PMID: 26122299 DOI: 10.1016/j.yhbeh.2015.05.024] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/29/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". There are sex differences in hippocampus-dependent cognition and neurogenesis suggesting that sex hormones are involved. Estrogens modulate certain forms of spatial and contextual memory and neurogenesis in the adult female rodent, and to a lesser extent male, hippocampus. This review focuses on the effects of sex and estrogens on hippocampal learning, memory, and neurogenesis in the young and aged adult rodent. We discuss how factors such as the type of estrogen, duration and dose of treatment, timing of treatment, and type of memory influence the effects of estrogens on cognition and neurogenesis. We also address how reproductive experience (pregnancy and mothering) and aging interact with estrogens to modulate hippocampal cognition and neurogenesis in females. Given the evidence that adult hippocampal neurogenesis plays a role in long-term spatial memory and pattern separation, we also discuss the functional implications of regulating neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Shunya Yagi
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Carmen Chow
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Liisa A M Galea
- Department of Psychology, Centre for Brain Health, Program in Neuroscience, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
38
|
Vega-Rivera NM, Fernández-Guasti A, Ramírez-Rodríguez G, Estrada-Camarena E. Effect of sub-optimal doses of fluoxetine plus estradiol on antidepressant-like behavior and hippocampal neurogenesis in ovariectomized rats. Psychoneuroendocrinology 2015; 57:113-24. [PMID: 25917885 DOI: 10.1016/j.psyneuen.2015.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/11/2015] [Accepted: 03/15/2015] [Indexed: 02/06/2023]
Abstract
Estrogens and antidepressants synergize to reduce depressive symptoms and stimulate neurogenesis and neuroplastic events. The aim of this study was to explore whether the antidepressant-like effect induced by the combination of low doses of estradiol (E2) and fluoxetine (FLX) involves changes in cell proliferation, early survival, morphology and dendrite complexity of hippocampal new-immature neurons. The antidepressant-like effects of E2 and/or FLX were evaluated by the forced swimming test (FST), cell proliferation was determined with the endogenous marker Ki67, survival of newborn cells was established with bromo-deoxiuridine (BrdU) and immature neurons were ascertained by doublecortin (DCX) labeling while their dendrite complexity was evaluated with Sholl analysis. Ovariectomized Wistar rats were randomly assigned to one of the following groups: Vehicle (saline/14 days+Oil/-8h before FST); E2 (saline/14 days + E2 2.5 or 10 μg/rat; -8 h before FST); FLX (1.25 or 10 mg/kg for 14 days + oil -8h before FST), and FLX plus E2 (FLX 1.25 mg/kg for 14 days + E2 2.5 μg/rat -8 h before FST). The combination of sub-threshold doses of FLX plus E2 produced antidepressant-like actions similar to those induced by FLX or E2 given independently at optimal doses. Only FLX at an optimal dose and the combination of FLX plus E2 increased cell proliferation, the number of DCX-labeled immature neurons and the complexity of their dendritic tree, suggesting that these events may be responsible for their antidepressant-like effect.
Collapse
Affiliation(s)
- Nelly M Vega-Rivera
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry, México, D.F., Mexico; Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), México, D.F., Mexico
| | - Alonso Fernández-Guasti
- Department of Pharmacobiology, Center for Research and Advanced Studies (CINVESTAV), México, D.F., Mexico
| | - Gerardo Ramírez-Rodríguez
- Laboratory of Neurogenesis, Division of Clinical Investigations, National Institute of Psychiatry, México, D.F., Mexico
| | - Erika Estrada-Camarena
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry, México, D.F., Mexico.
| |
Collapse
|
39
|
Wang XJ, Song XJ, Gao PH, Zhou XY, Zhou SN. Estradiol prevents Aβ 25 35-inhibited long-term potentiation induction through enhancing survival of newborn neurons in the dentate gyrus. Int J Neurosci 2015; 126:154-62. [PMID: 25567530 DOI: 10.3109/00207454.2014.995267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIM AND METHODS Estradiol (E2) is reported to attenuate β-amyloid (Aβ) accumulation and slow the progression of Alzheimer's disease (AD). This study explored the beneficial effect of E2 in AD using histological examination and electrophysiological recording technique in AD model mice created by intracerebroventricular injection of β-amyloid 25-35 (Aβ 25-35). RESULTS Infusion of Aβ 25-35 reduced the number of newborn neurons in the 2nd week after birth, a critical period for neurite growth, and impaired high-frequency stimulation-dependent long-term potentiation (LTP) induction in perforant path-granular synapses of hippocampal dentate gyrus (DG). Administration of E2 from the 2nd to 4th week after cell birth in Aβ 25-35-mice ameliorated the impairment of newborn neurons and LTP induction in DG. Acute application of E2 failed to increase the newborn neurons and rescue LTP induction in the DG of Aβ 25-35-mice. CONCLUSIONS The effect of E2 in Aβ 25-35-impaired LTP induction depends on its neuroprotection improvement.
Collapse
Affiliation(s)
- Xian-Jun Wang
- a Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, Shandong, China.,b Department of Neurology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xiao-Jie Song
- b Department of Neurology, Linyi People's Hospital, Linyi, Shandong, China
| | - Pei-Hong Gao
- b Department of Neurology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xue-Ying Zhou
- c Department of Neurology, Chinese Medicine Hospital of Shandong, Chinese Medicine University, Jinan, China
| | - Sheng-Nian Zhou
- a Department of Neurology, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, Shandong, China
| |
Collapse
|
40
|
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
|
41
|
Martini M, Calandreau L, Jouhanneau M, Mhaouty-Kodja S, Keller M. Perinatal exposure to methoxychlor enhances adult cognitive responses and hippocampal neurogenesis in mice. Front Behav Neurosci 2014; 8:202. [PMID: 24982620 PMCID: PMC4059339 DOI: 10.3389/fnbeh.2014.00202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/19/2014] [Indexed: 02/02/2023] Open
Abstract
During perinatal life, sex steroids, such as estradiol, have marked effects on the development and function of the nervous system. Environmental estrogens or xenoestrogens are man-made chemicals, which animal and human population encounter in the environment and which are able to disrupt the functioning of the endocrine system. Scientific interest in the effects of exposure to xenoestrogens has focused more on fertility and reproductive behaviors, while the effects on cognitive behaviors have received less attention. Therefore, the present study explored whether the organochlorine insecticide Methoxychlor (MXC), with known xenoestrogens properties, administered during the perinatal period (from gestational day 11 to postnatal day 8) to pregnant-lactating females, at an environmentally relevant dose (20 µg/kg (body weight)/day), would also affect learning and memory functions depending on the hippocampus of male and female offspring mice in adulthood. When tested in adulthood, MXC perinatal exposure led to an increase in anxiety-like behavior and in short-term spatial working memory in both sexes. Emotional learning was also assessed using a contextual fear paradigm and MXC treated male and female mice showed an enhanced freezing behavior compared to controls. These results were correlated with an increased survival of adult generated cells in the adult hippocampus. In conclusion, our results show that perinatal exposure to an environmentally relevant dose of MXC has an organizational effect on hippocampus-dependent memory and emotional behaviors.
Collapse
Affiliation(s)
- Mariangela Martini
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA/CNRS/Université Francois Rabelais Nouzilly, France
| | - Ludovic Calandreau
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA/CNRS/Université Francois Rabelais Nouzilly, France
| | - Mélanie Jouhanneau
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA/CNRS/Université Francois Rabelais Nouzilly, France
| | - Sakina Mhaouty-Kodja
- Physiopathologie des Maladies du Système Nerveux Central, UMR 7224 CNRS/INSERM U 952/Université Pierre et Marie Curie Paris, France
| | - Matthieu Keller
- Physiologie de la Reproduction et des Comportements, UMR 7247 INRA/CNRS/Université Francois Rabelais Nouzilly, France
| |
Collapse
|
42
|
Burger DK, Gulbrandsen T, Saucier DM, Iwaniuk AN. The effects of season and sex on dentate gyrus size and neurogenesis in a wild rodent, Richardson's ground squirrel (Urocitellus richardsonii). Neuroscience 2014; 272:240-51. [PMID: 24813432 DOI: 10.1016/j.neuroscience.2014.04.067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/07/2014] [Accepted: 04/28/2014] [Indexed: 11/19/2022]
Abstract
Sex and reproductive status affect hippocampal neurogenesis and dentate gyrus (DG) size in rodents. Relatively few studies, however, address these two effects simultaneously and even fewer studies address this issue in wild populations. Here, we examined seasonal and sex differences in neurogenesis and DG size in a wild, polygynous and social rodent, Richardson's ground squirrel (Uriocitellus richardsonii). Based on the behavioral ecology of this species, we predicted that both neurogenesis and DG size would be sexually dimorphic and the degree of dimorphism would be greatest in the breeding season. Using unbiased stereology and doublecortin (DCX) immunohistochemistry, we found that brain volume, DG size and number of DCX cells varied significantly between breeding and non-breeding seasons, but only brain volume and the number of DCX labeled cells differed between the sexes. Both sex and seasonal differences likely reflect circulating hormone levels, but the extent to which these differences relate to space use in this species is unclear. Based on the degree of seasonal differences in neurogenesis and the DG, we suggest that ground squirrels could be considered model species in which to examine hippocampal plasticity in an ecologically valid context.
Collapse
Affiliation(s)
- D K Burger
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - T Gulbrandsen
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - D M Saucier
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, ON, Canada
| | - A N Iwaniuk
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.
| |
Collapse
|
43
|
Zhou L, Yin J, Wang C, Liao J, Liu G, Chen L. Lack of seipin in neurons results in anxiety- and depression-like behaviors via down regulation of PPARγ. Hum Mol Genet 2014; 23:4094-102. [PMID: 24651066 DOI: 10.1093/hmg/ddu126] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Libin Zhou
- State Key Laboratory of Reproductive Medicine and Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Jun Yin
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Conghui Wang
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Jiawei Liao
- Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Beijing 100191, China
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Beijing 100191, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine and Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
44
|
Hami J, Kheradmand H, Haghir H. Gender differences and lateralization in the distribution pattern of insulin-like growth factor-1 receptor in developing rat hippocampus: an immunohistochemical study. Cell Mol Neurobiol 2014; 34:215-26. [PMID: 24287499 DOI: 10.1007/s10571-013-0005-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/06/2013] [Indexed: 12/11/2022]
Abstract
Numerous investigators have provided data supporting essential roles for insulin-like growth factor-I (IGF-I) in development of the brain. The aim of this study was to immunohistochemically determine the distinct regional distribution pattern of IGF-1 receptor (IGF-IR) expression in various portions of newborn rat hippocampus on postnatal days 0 (P0), 7 (P7), and 14 (P14), with comparison between male/female and right/left hippocampi. We found an overall significant increase in distribution of IGF-IR-positive (IGF-IR+) cells in CA1 from P0 until P14. Although, no marked changes in distribution of IGF-IR+ cells in areas CA2 and CA3 were observed; IGF-IR+ cells in DG decreased until P14. The smallest number of immunoreactive cells was present in CA2 and the highest number in DG at P0. Moreover, in CA1, CA3, and DG, the number of IGF-IR+ cells was markedly higher in both sides of the hippocampus in females. Our data also showed a higher mean number of IGF-IR+ cells in the left hippocampus of female at P7. By contrast, male pups showed a significantly higher number of IGF-IR+ cells in the DG of the right hippocampus. At P14, the mean number of immunoreactive cells in CA1, CA3, and DG areas found to be significantly increased in left side of hippocampus of males, compared to females. These results indicate the existence of a differential distribution pattern of IGF-IR between left-right and male-female hippocampi. Together with other mechanisms, these differences may underlie sexual dimorphism and left-right asymmetry in the hippocampus.
Collapse
Affiliation(s)
- Javad Hami
- Department of Anatomy, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | | |
Collapse
|
45
|
Hami J, Kheradmand H, Haghir H. Sex differences and laterality of insulin receptor distribution in developing rat hippocampus: an immunohistochemical study. J Mol Neurosci 2014; 54:100-8. [PMID: 24573599 DOI: 10.1007/s12031-014-0255-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/04/2014] [Indexed: 12/19/2022]
Abstract
This study aimed to compare the regional distribution of insulin receptor in various portions of newborn rat hippocampus on postnatal days 0 (P0), 7 (P7), and 14 (P14) between male/female and right/left hippocampi. We found that the number of insulin receptor (InsR)-immunoreactive-positive (InsR+) cells in CA1 continued to increase until P7 and remained unchanged thereafter. A marked increase in distribution of InsR+ cells in CA3 from P0 to P14 was observed, although there was a significant decline in the number of InsR+ cells in dentate gyrus (DG) at the same time. No differences between the right/left and male/female hippocampi were detected at P0 (P > 0.05). Seven-day-old female rats showed a higher number of labeled cells in the left than in the right hippocampus. Moreover, the differences between the number of InsR+ cells in area CA1 and CA3 were statistically significant between males and females (P < 0.05). At P14, the number of InsR+ cells was significantly higher in CA1 and DG of males, especially in the right one (P < 0.05). These results indicate the existence of a differential distribution pattern of InsR between the left/right and male/female hippocampi. Together with other mechanisms, these differences may underlie sexual dimorphism and left/right asymmetry in the hippocampus.
Collapse
Affiliation(s)
- Javad Hami
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | | |
Collapse
|
46
|
17 β -Estradiol attenuates poststroke depression and increases neurogenesis in female ovariectomized rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:392434. [PMID: 24307996 PMCID: PMC3838842 DOI: 10.1155/2013/392434] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 09/11/2013] [Accepted: 09/18/2013] [Indexed: 11/18/2022]
Abstract
Studies have linked neurogenesis to the beneficial actions of specific antidepressants. However, whether 17β-estradiol (E2), an antidepressant, can ameliorate poststroke depression (PSD) and whether E2-mediated improvement of PSD is associated with neurogenesis are largely unexplored. In the present study, we found that depressive-like behaviors were observed at the first week after focal ischemic stroke in female ovariectomized (OVX) rats, as measured by sucrose preference and open field test, suggesting that focal cerebral ischemia could induce PSD. Three weeks after middle cerebral artery occlusion (MCAO), rats were treated with E2 for consecutive 14 days. We found that E2-treated rats had significantly improving ischemia-induced depression-like behaviors in the forced-swimming test and sucrose preference test, compared to vehicle-treated group. In addition, we also found that BrdU- and doublecortin (DCX)-positive cells in the dentate gyrus of the hippocampus and the subventricular zone (SVZ) were significantly increased in ischemic rats after E2 treatment, compared to vehicle-treated group. Our data suggest that focal cerebral ischemia can induce PSD, and E2 can ameliorate PSD. In addition, newborn neurons in the hippocampus may play an important role in E2-mediated antidepressant like effect after ischemic stroke.
Collapse
|
47
|
Galea LAM, Wainwright SR, Roes MM, Duarte-Guterman P, Chow C, Hamson DK. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of neurogenesis and potential functional implications. J Neuroendocrinol 2013; 25:1039-61. [PMID: 23822747 DOI: 10.1111/jne.12070] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/23/2013] [Accepted: 06/29/2013] [Indexed: 12/12/2022]
Abstract
The hippocampus is an area of the brain that undergoes dramatic plasticity in response to experience and hormone exposure. The hippocampus retains the ability to produce new neurones in most mammalian species and is a structure that is targeted in a number of neurodegenerative and neuropsychiatric diseases, many of which are influenced by both sex and sex hormone exposure. Intriguingly, gonadal and adrenal hormones affect the structure and function of the hippocampus differently in males and females. Adult neurogenesis in the hippocampus is regulated by both gonadal and adrenal hormones in a sex- and experience-dependent way. Sex differences in the effects of steroid hormones to modulate hippocampal plasticity should not be completely unexpected because the physiology of males and females is different, with the most notable difference being that females gestate and nurse the offspring. Furthermore, reproductive experience (i.e. pregnancy and mothering) results in permanent changes to the maternal brain, including the hippocampus. This review outlines the ability of gonadal and stress hormones to modulate multiple aspects of neurogenesis (cell proliferation and cell survival) in both male and female rodents. The function of adult neurogenesis in the hippocampus is linked to spatial memory and depression, and the present review provides early evidence of the functional links between the hormonal modulation of neurogenesis that may contribute to the regulation of cognition and stress.
Collapse
Affiliation(s)
- L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Hamson DK, Wainwright SR, Taylor JR, Jones BA, Watson NV, Galea LAM. Androgens increase survival of adult-born neurons in the dentate gyrus by an androgen receptor-dependent mechanism in male rats. Endocrinology 2013; 154:3294-304. [PMID: 23782943 DOI: 10.1210/en.2013-1129] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gonadal steroids are potent regulators of adult neurogenesis. We previously reported that androgens, such as testosterone (T) and dihydrotestosterone (DHT), but not estradiol, increased the survival of new neurons in the dentate gyrus of the male rat. These results suggest androgens regulate hippocampal neurogenesis via the androgen receptor (AR). To test this supposition, we examined the role of ARs in hippocampal neurogenesis using 2 different approaches. In experiment 1, we examined neurogenesis in male rats insensitive to androgens due to a naturally occurring mutation in the gene encoding the AR (termed testicular feminization mutation) compared with wild-type males. In experiment 2, we injected the AR antagonist, flutamide, into castrated male rats and compared neurogenesis levels in the dentate gyrus of DHT and oil-treated controls. In experiment 1, chronic T increased hippocampal neurogenesis in wild-type males but not in androgen-insensitive testicular feminization mutation males. In experiment 2, DHT increased hippocampal neurogenesis via cell survival, an effect that was blocked by concurrent treatment with flutamide. DHT, however, did not affect cell proliferation. Interestingly, cells expressing doublecortin, a marker of immature neurons, did not colabel with ARs in the dentate gyrus, but ARs were robustly expressed in other regions of the hippocampus. Together these studies provide complementary evidence that androgens regulate adult neurogenesis in the hippocampus via the AR but at a site other than the dentate gyrus. Understanding where in the brain androgens act to increase the survival of new neurons in the adult brain may have implications for neurodegenerative disorders.
Collapse
Affiliation(s)
- D K Hamson
- Department of Psychology, University of British Columbia, Vancouver V6T 1Z4, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Ormerod BK, Hanft SJ, Asokan A, Haditsch U, Lee SW, Palmer TD. PPARγ activation prevents impairments in spatial memory and neurogenesis following transient illness. Brain Behav Immun 2013; 29:28-38. [PMID: 23108061 PMCID: PMC3570721 DOI: 10.1016/j.bbi.2012.10.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/25/2012] [Accepted: 10/19/2012] [Indexed: 11/25/2022] Open
Abstract
The detrimental effects of illness on cognition are familiar to virtually everyone. Some effects resolve quickly while others may linger after the illness resolves. We found that a transient immune response stimulated by lipopolysaccharide (LPS) compromised hippocampal neurogenesis and impaired hippocampus-dependent spatial memory. The immune event caused an ∼50% reduction in the number of neurons generated during the illness and the onset of the memory impairment was delayed and coincided with the time when neurons generated during the illness would have become functional within the hippocampus. Broad spectrum non-steroidal anti-inflammatory drugs attenuated these effects but selective Cox-2 inhibition was ineffective while PPARγ activation was surprisingly effective at protecting both neurogenesis and memory from the effects of LPS-produced transient illness. These data may highlight novel mechanisms behind chronic inflammatory and neuroinflammatory episodes that are known to compromise hippocampus-dependent forms of learning and memory.
Collapse
Affiliation(s)
- Brandi K. Ormerod
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA, 94305,J. Crayton Pruitt Family Department of Biomedical Engineering, McKnight Brain Institute and Neuroscience Department, University of Florida, Gainesville, FL, USA, 32611,To whom correspondence should be addressed: Dr. Brandi K. Ormerod: J. Crayton Pruitt Family Department of Biomedical Engineering, 1600 Center Drive, Room J296, Biomedical Sciences Building, University of Florida, Gainesville, FL, USA, 32611, Phone: 352-273-8125, Fax: 352-273-9222, Or Dr. Theo D. Palmer, Institute for Stem Cell Biology and Regenerative Medicine, Lorey I Lokey Stem Cell Building, Rm1141, 265 Campus Drive, Stanford University, Stanford, CA, USA, 94305. Phone: 650-723-9306, Fax: 650-736-0936,
| | - Simon J. Hanft
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA, 94305
| | - Aditya Asokan
- J. Crayton Pruitt Family Department of Biomedical Engineering, McKnight Brain Institute and Neuroscience Department, University of Florida, Gainesville, FL, USA, 32611
| | - Ursula Haditsch
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA, 94305
| | - Star W. Lee
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA, 94305
| | - Theo D. Palmer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA, 94305,To whom correspondence should be addressed: Dr. Brandi K. Ormerod: J. Crayton Pruitt Family Department of Biomedical Engineering, 1600 Center Drive, Room J296, Biomedical Sciences Building, University of Florida, Gainesville, FL, USA, 32611, Phone: 352-273-8125, Fax: 352-273-9222, Or Dr. Theo D. Palmer, Institute for Stem Cell Biology and Regenerative Medicine, Lorey I Lokey Stem Cell Building, Rm1141, 265 Campus Drive, Stanford University, Stanford, CA, USA, 94305. Phone: 650-723-9306, Fax: 650-736-0936,
| |
Collapse
|
50
|
Diotel N, Vaillant C, Gabbero C, Mironov S, Fostier A, Gueguen MM, Anglade I, Kah O, Pellegrini E. Effects of estradiol in adult neurogenesis and brain repair in zebrafish. Horm Behav 2013; 63:193-207. [PMID: 22521210 DOI: 10.1016/j.yhbeh.2012.04.003] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 12/21/2022]
Abstract
The brain of the adult teleost fish exhibits intense neurogenic activity and an outstanding capability for brain repair. Remarkably, the brain estrogen-synthesizing enzyme, aromatase B, is strongly expressed, particularly in adult fishes, in radial glial cells, which act as progenitors. Using zebrafish, we tested the hypothesis that estrogens affect adult neurogenesis and brain regeneration by modulating the neurogenic activity of radial glial cells. To investigate this, the estrogenic environment was modified through inhibition of aromatase activity, blockade of nuclear estrogen receptors, or estrogenic treatments. Estrogens significantly decreased cell proliferation and migration at the olfactory bulbs/telencephalon junction and in the mediobasal hypothalamus. It also appears that cell survival is reduced at the olfactory bulbs/telencephalon junction. We also developed a model of telencephalic lesion to assess the role of aromatase and estrogens in brain repair. Proliferation increased rapidly immediately after the lesion in the parenchyma of the injured telencephalon, while proliferation at the ventricular surface appeared after 48 h and peaked at 7 days. At this time, most proliferative cells express Sox2, however, none of these Sox2 positive cells correspond to aromatase B-positive radial glial cells. Interestingly, aromatase B expression was significantly reduced 48 h and 7 days after the injury, but surprisingly, at 72 h after lesion, aromatase B expression appeared de novo expressed in parenchyma cells, suggesting a role for this ectopic expression of aromatase in brain repair mechanisms. Altogether these data suggest that estrogens modulate adult, but not reparative neurogenesis, in zebrafish.
Collapse
Affiliation(s)
- Nicolas Diotel
- Neuroendocrine Effects of Endocrine Disruptors, Inserm, IRSET, U1085, Université de Rennes 1, Rennes, France
| | | | | | | | | | | | | | | | | |
Collapse
|