1
|
Torres E, Pellegrino G, Granados-Rodríguez M, Fuentes-Fayos AC, Velasco I, Coutteau-Robles A, Legrand A, Shanabrough M, Perdices-Lopez C, Leon S, Yeo SH, Manchishi SM, Sánchez-Tapia MJ, Navarro VM, Pineda R, Roa J, Naftolin F, Argente J, Luque RM, Chowen JA, Horvath TL, Prevot V, Sharif A, Colledge WH, Tena-Sempere M, Romero-Ruiz A. Kisspeptin signaling in astrocytes modulates the reproductive axis. J Clin Invest 2024; 134:e172908. [PMID: 38861336 PMCID: PMC11291270 DOI: 10.1172/jci172908] [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: 06/08/2023] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
Reproduction is safeguarded by multiple, often cooperative, regulatory networks. Kisspeptin signaling, via KISS1R, plays a fundamental role in reproductive control, primarily by regulation of hypothalamic GnRH neurons. We disclose herein a pathway for direct kisspeptin actions in astrocytes that contributes to central reproductive modulation. Protein-protein interaction and ontology analyses of hypothalamic proteomic profiles after kisspeptin stimulation revealed that glial/astrocyte markers are regulated by kisspeptin in mice. This glial-kisspeptin pathway was validated by the demonstrated expression of Kiss1r in mouse astrocytes in vivo and astrocyte cultures from humans, rats, and mice, where kisspeptin activated canonical intracellular signaling-pathways. Cellular coexpression of Kiss1r with the astrocyte markers GFAP and S100-β occurred in different brain regions, with higher percentage in Kiss1- and GnRH-enriched areas. Conditional ablation of Kiss1r in GFAP-positive cells in the G-KiR-KO mouse altered gene expression of key factors in PGE2 synthesis in astrocytes and perturbed astrocyte-GnRH neuronal appositions, as well as LH responses to kisspeptin and LH pulsatility, as surrogate marker of GnRH secretion. G-KiR-KO mice also displayed changes in reproductive responses to metabolic stress induced by high-fat diet, affecting female pubertal onset, estrous cyclicity, and LH-secretory profiles. Our data unveil a nonneuronal pathway for kisspeptin actions in astrocytes, which cooperates in fine-tuning the reproductive axis and its responses to metabolic stress.
Collapse
Affiliation(s)
- Encarnacion Torres
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Giuliana Pellegrino
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Melissa Granados-Rodríguez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Antonio C. Fuentes-Fayos
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Inmaculada Velasco
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Adrian Coutteau-Robles
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Amandine Legrand
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Marya Shanabrough
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Cecilia Perdices-Lopez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Silvia Leon
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Shel H. Yeo
- Reproductive Physiology Group, Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Stephen M. Manchishi
- Reproductive Physiology Group, Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Maria J. Sánchez-Tapia
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Victor M. Navarro
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston,Massachusetts, USA
| | - Rafael Pineda
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Juan Roa
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Jesús Argente
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, and IMDEA-Food Institute, CEI-UAM+CSIC, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raúl M. Luque
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Julie A. Chowen
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, and IMDEA-Food Institute, CEI-UAM+CSIC, Madrid, Spain
| | - Tamas L. Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Ariane Sharif
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - William H. Colledge
- Reproductive Physiology Group, Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Romero-Ruiz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| |
Collapse
|
2
|
Sauve F, Nampoothiri S, Clarke SA, Fernandois D, Ferreira Coêlho CF, Dewisme J, Mills EG, Ternier G, Cotellessa L, Iglesias-Garcia C, Mueller-Fielitz H, Lebouvier T, Perbet R, Florent V, Baroncini M, Sharif A, Ereño-Orbea J, Mercado-Gómez M, Palazon A, Mattot V, Pasquier F, Catteau-Jonard S, Martinez-Chantar M, Hrabovszky E, Jourdain M, Deplanque D, Morelli A, Guarnieri G, Storme L, Robil C, Trottein F, Nogueiras R, Schwaninger M, Pigny P, Poissy J, Chachlaki K, Maurage CA, Giacobini P, Dhillo W, Rasika S, Prevot V. Long-COVID cognitive impairments and reproductive hormone deficits in men may stem from GnRH neuronal death. EBioMedicine 2023; 96:104784. [PMID: 37713808 PMCID: PMC10507138 DOI: 10.1016/j.ebiom.2023.104784] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND We have recently demonstrated a causal link between loss of gonadotropin-releasing hormone (GnRH), the master molecule regulating reproduction, and cognitive deficits during pathological aging, including Down syndrome and Alzheimer's disease. Olfactory and cognitive alterations, which persist in some COVID-19 patients, and long-term hypotestosteronaemia in SARS-CoV-2-infected men are also reminiscent of the consequences of deficient GnRH, suggesting that GnRH system neuroinvasion could underlie certain post-COVID symptoms and thus lead to accelerated or exacerbated cognitive decline. METHODS We explored the hormonal profile of COVID-19 patients and targets of SARS-CoV-2 infection in post-mortem patient brains and human fetal tissue. FINDINGS We found that persistent hypotestosteronaemia in some men could indeed be of hypothalamic origin, favouring post-COVID cognitive or neurological symptoms, and that changes in testosterone levels and body weight over time were inversely correlated. Infection of olfactory sensory neurons and multifunctional hypothalamic glia called tanycytes highlighted at least two viable neuroinvasion routes. Furthermore, GnRH neurons themselves were dying in all patient brains studied, dramatically reducing GnRH expression. Human fetal olfactory and vomeronasal epithelia, from which GnRH neurons arise, and fetal GnRH neurons also appeared susceptible to infection. INTERPRETATION Putative GnRH neuron and tanycyte dysfunction following SARS-CoV-2 neuroinvasion could be responsible for serious reproductive, metabolic, and mental health consequences in long-COVID and lead to an increased risk of neurodevelopmental and neurodegenerative pathologies over time in all age groups. FUNDING European Research Council (ERC) grant agreements No 810331, No 725149, No 804236, the European Union Horizon 2020 research and innovation program No 847941, the Fondation pour la Recherche Médicale (FRM) and the Agence Nationale de la Recherche en Santé (ANRS) No ECTZ200878 Long Covid 2021 ANRS0167 SIGNAL, Agence Nationale de la recherche (ANR) grant agreements No ANR-19-CE16-0021-02, No ANR-11-LABEX-0009, No. ANR-10-LABEX-0046, No. ANR-16-IDEX-0004, Inserm Cross-Cutting Scientific Program HuDeCA, the CHU Lille Bonus H, the UK Medical Research Council (MRC) and National Institute of Health and care Research (NIHR).
Collapse
Affiliation(s)
- Florent Sauve
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Sreekala Nampoothiri
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Sophie A Clarke
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | | | - Julie Dewisme
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France
| | - Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | | | - Helge Mueller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Thibaud Lebouvier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Neurology, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France
| | - Romain Perbet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France
| | - Vincent Florent
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Marc Baroncini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - June Ereño-Orbea
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Maria Mercado-Gómez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Asis Palazon
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Virginie Mattot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Florence Pasquier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Neurology, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France
| | - Sophie Catteau-Jonard
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Gynecology and Obstetrics, Jeanne de Flandres Hospital, F-59000, Lille, France
| | - Maria Martinez-Chantar
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Mercé Jourdain
- Univ. Lille, Inserm, CHU Lille, Service de Médecine Intensive Réanimation, U1190, EGID, F-59000 Lille, France
| | - Dominique Deplanque
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; University Lille, Inserm, CHU Lille, Centre d'investigation Clinique (CIC) 1403, F-59000, Lille, France; LICORNE Study Group, CHU Lille, Lille, France
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Giulia Guarnieri
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Laurent Storme
- CHU Lille, Department of Neonatology, Hôpital Jeanne de Flandre, FHU 1000 Days for Health, F-59000, France
| | - Cyril Robil
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - François Trottein
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Pascal Pigny
- CHU Lille, Service de Biochimie et Hormonologie, Centre de Biologie Pathologie, Lille, France
| | - Julien Poissy
- LICORNE Study Group, CHU Lille, Lille, France; Univ. Lille, Inserm U1285, CHU Lille, Pôle de Réanimation, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Claude-Alain Maurage
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France; LICORNE Study Group, CHU Lille, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Waljit Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - S Rasika
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France.
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France.
| |
Collapse
|
3
|
Krajnak K, Farcas M, McKinney W, Waugh S, Mandler K, Knepp A, Jackson M, Richardson D, Hammer M, Matheson J, Thomas T, Qian Y. Inhalation of polycarbonate emissions generated during 3D printing processes affects neuroendocrine function in male rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2023; 86:575-596. [PMID: 37350301 PMCID: PMC10527863 DOI: 10.1080/15287394.2023.2226198] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Three-dimensional (3D) printing of manufactured goods has increased in the last 10 years. The increased use of this technology has resulted in questions regarding the influence of inhaling emissions generated during printing. The goal of this study was to determine if inhalation of particulate and/or toxic chemicals generated during printing with polycarbonate (PC) plastic affected the neuroendocrine system. Male rats were exposed to 3D-printer emissions (592 µg particulate/m3 air) or filtered air for 4 h/day (d), 4 days/week and total exposures lengths were 1, 4, 8, 15 or 30 days. The effects of these exposures on hormone concentrations, and markers of function and/or injury in the olfactory bulb, hypothalamus and testes were measured after 1, 8 and 30 days exposure. Thirty days of exposure to 3D printer emissions resulted in reductions in thyroid stimulating hormone, follicle stimulating hormone and prolactin. These changes were accompanied by (1) elevation in markers of cell injury; (2) reductions in active mitochondria in the olfactory bulb, diminished gonadotropin releasing hormone cells and fibers as well as less tyrosine hydroxylase immunolabeled fibers in the arcuate nucleus; and (3) decrease in spermatogonium. Polycarbonate plastics may contain bisphenol A, and the effects of exposure to these 3D printer-generated emissions on neuroendocrine function are similar to those noted following exposure to bisphenol A.
Collapse
Affiliation(s)
- Kristine Krajnak
- Physical Effects Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Mariana Farcas
- Physiology and Pathology Research BranchHealth Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Walter McKinney
- Physical Effects Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Stacey Waugh
- Physical Effects Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Kyle Mandler
- Physiology and Pathology Research BranchHealth Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Alycia Knepp
- Physiology and Pathology Research BranchHealth Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Mark Jackson
- Physical Effects Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Diana Richardson
- Histopathology Core, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - MaryAnne Hammer
- Histopathology Core, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Joanna Matheson
- Office of Hazard Identification and Reduction, U.S. Consumer Product Safety Commission, Bethesda, MD, USA
| | - Treye Thomas
- Office of Hazard Identification and Reduction, U.S. Consumer Product Safety Commission, Bethesda, MD, USA
| | - Yong Qian
- Physiology and Pathology Research BranchHealth Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|
4
|
Plakkot B, Di Agostino A, Subramanian M. Implications of Hypothalamic Neural Stem Cells on Aging and Obesity-Associated Cardiovascular Diseases. Cells 2023; 12:cells12050769. [PMID: 36899905 PMCID: PMC10000584 DOI: 10.3390/cells12050769] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The hypothalamus, one of the major regulatory centers in the brain, controls various homeostatic processes, and hypothalamic neural stem cells (htNSCs) have been observed to interfere with hypothalamic mechanisms regulating aging. NSCs play a pivotal role in the repair and regeneration of brain cells during neurodegenerative diseases and rejuvenate the brain tissue microenvironment. The hypothalamus was recently observed to be involved in neuroinflammation mediated by cellular senescence. Cellular senescence, or systemic aging, is characterized by a progressive irreversible state of cell cycle arrest that causes physiological dysregulation in the body and it is evident in many neuroinflammatory conditions, including obesity. Upregulation of neuroinflammation and oxidative stress due to senescence has the potential to alter the functioning of NSCs. Various studies have substantiated the chances of obesity inducing accelerated aging. Therefore, it is essential to explore the potential effects of htNSC dysregulation in obesity and underlying pathways to develop strategies to address obesity-induced comorbidities associated with brain aging. This review will summarize hypothalamic neurogenesis associated with obesity and prospective NSC-based regenerative therapy for the treatment of obesity-induced cardiovascular conditions.
Collapse
|
5
|
Desroziers E. Unusual suspects: Glial cells in fertility regulation and their suspected role in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13136. [PMID: 35445462 PMCID: PMC9489003 DOI: 10.1111/jne.13136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
Gonadotropin-releasing-hormone (GnRH) neurons sitting within the hypothalamus control the production of gametes and sex steroids by the gonads, therefore ensuring survival of species. As orchestrators of reproductive function, GnRH neurons integrate information from external and internal cues. This occurs through an extensively studied neuronal network known as the "GnRH neuronal network." However, the brain is not simply composed of neurons. Evidence suggests a role for glial cells in controlling GnRH neuron activity, secretion and fertility outcomes, although numerous questions remain. Glial cells have historically been seen as support cells for neurons. This idea has been challenged by the discovery that some neurological diseases originate from glial dysfunction. The prevalence of infertility disorders is increasing worldwide, with one in four couples being affected; therefore, it remains essential to understand the mechanisms by which the brain controls fertility. The "GnRH glial network" could be a major player in infertility disorders and represent a potential therapeutic target. In polycystic ovary syndrome (PCOS), the most common infertility disorder of reproductive aged women worldwide, the brain is considered a prime suspect. Recent studies have demonstrated pathological neuronal wiring of the "GnRH neuronal network" in PCOS-like animal models. However, the role of the "GnRH glial network" remains to be elucidated. In this review, I aim to propose glial cells as unusual suspects in infertility disorders such as PCOS. In the first part, I state our current knowledge about the role of glia in the regulation of GnRH neurons and fertility. In the second part, based on our recent findings, I discuss how glial cells could be implicated in PCOS pathology.
Collapse
Affiliation(s)
- Elodie Desroziers
- Department of Physiology, Centre for NeuroendocrinologyUniversity of OtagoDunedinNew Zealand
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Neuroplasticity of Reproductive Behaviours TeamParisFrance
| |
Collapse
|
6
|
Prevot V, Sharif A. The polygamous GnRH neuron: Astrocytic and tanycytic communication with a neuroendocrine neuronal population. J Neuroendocrinol 2022; 34:e13104. [PMID: 35233849 DOI: 10.1111/jne.13104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
To ensure the survival of the species, hypothalamic neuroendocrine circuits controlling fertility, which converge onto neurons producing gonadotropin-releasing hormone (GnRH), must respond to fluctuating physiological conditions by undergoing rapid and reversible structural and functional changes. However, GnRH neurons do not act alone, but through reciprocal interactions with multiple hypothalamic cell populations, including several glial and endothelial cell types. For instance, it has long been known that in the hypothalamic median eminence, where GnRH axons terminate and release their neurohormone into the pituitary portal blood circulation, morphological plasticity displayed by distal processes of tanycytes modifies their relationship with adjacent neurons as well as the spatial properties of the neurohemal junction. These alterations not only regulate the capacity of GnRH neurons to release their neurohormone, but also the activation of discrete non-neuronal pathways that mediate feedback by peripheral hormones onto the hypothalamus. Additionally, a recent breakthrough has demonstrated that GnRH neurons themselves orchestrate the establishment of their neuroendocrine circuitry during postnatal development by recruiting an entourage of newborn astrocytes that escort them into adulthood and, via signalling through gliotransmitters such as prostaglandin E2, modulate their activity and GnRH release. Intriguingly, several environmental and behavioural toxins perturb these neuron-glia interactions and consequently, reproductive maturation and fertility. Deciphering the communication between GnRH neurons and other neural cell types constituting hypothalamic neuroendocrine circuits is thus critical both to understanding physiological processes such as puberty, oestrous cyclicity and aging, and to developing novel therapeutic strategies for dysfunctions of these processes, including the effects of endocrine disruptors.
Collapse
Affiliation(s)
- Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| |
Collapse
|
7
|
Singular Adult Neural Stem Cells Do Not Exist. Cells 2022; 11:cells11040722. [PMID: 35203370 PMCID: PMC8870225 DOI: 10.3390/cells11040722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/04/2022] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Adult neural stem cells (aNSCs) are the source for the continuous production of new neurons throughout life. This so-called adult neurogenesis has been extensively studied; the intermediate cellular stages are well documented. Recent discoveries have raised new controversies in the field, such as the notion that progenitor cells hold similar self-renewal potential as stem cells, or whether different types of aNSCs exist. Here, we discuss evidence for heterogeneity of aNSCs, including short-term and long-term self-renewing aNSCs, regional and temporal differences in aNSC function, and single cell transcriptomics. Reviewing various genetic mouse models used for targeting aNSCs and lineage tracing, we consider potential lineage relationships between Ascl1-, Gli1-, and Nestin-targeted aNSCs. We present a multidimensional model of adult neurogenesis that incorporates recent findings and conclude that stemness is a phenotype, a state of properties that can change with time, rather than a cell property, which is static and immutable. We argue that singular aNSCs do not exist.
Collapse
|
8
|
Vanacker C, Defazio RA, Sykes CM, Moenter SM. A role for glial fibrillary acidic protein (GFAP)-expressing cells in the regulation of gonadotropin-releasing hormone (GnRH) but not arcuate kisspeptin neuron output in male mice. eLife 2021; 10:68205. [PMID: 34292152 PMCID: PMC8337074 DOI: 10.7554/elife.68205] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/21/2021] [Indexed: 01/05/2023] Open
Abstract
GnRH neurons are the final central neural output regulating fertility. Kisspeptin neurons in the hypothalamic arcuate nucleus (KNDy neurons) are considered the main regulator of GnRH output. GnRH and KNDy neurons are surrounded by astrocytes, which can modulate neuronal activity and communicate over distances. Prostaglandin E2 (PGE2), synthesized primarily by astrocytes, increases GnRH neuron activity and downstream pituitary release of luteinizing hormone (LH). We hypothesized that glial fibrillary acidic protein (GFAP)-expressing astrocytes play a role in regulating GnRH and/or KNDy neuron activity and LH release. We used adeno-associated viruses to target designer receptors exclusively activated by designer drugs (DREADDs) to GFAP-expressing cells to activate Gq- or Gi-mediated signaling. Activating Gq signaling in the preoptic area, near GnRH neurons, but not in the arcuate, increases LH release in vivo and GnRH firing in vitro via a mechanism in part dependent upon PGE2. These data suggest that astrocytes can activate GnRH/LH release in a manner independent of KNDy neurons.
Collapse
Affiliation(s)
- Charlotte Vanacker
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - R Anthony Defazio
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Charlene M Sykes
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, United States.,Internal Medicine, University of Michigan, Ann Arbor, United States.,Obstetrics & Gynecology, University of Michigan, Ann Arbor, United States.,Reproductive Sciences Program, University of Michigan, Ann Arbor, United States
| |
Collapse
|
9
|
Kostin A, Alam MA, McGinty D, Alam MN. Adult hypothalamic neurogenesis and sleep-wake dysfunction in aging. Sleep 2021; 44:5986548. [PMID: 33202015 DOI: 10.1093/sleep/zsaa173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
In the mammalian brain, adult neurogenesis has been extensively studied in the hippocampal sub-granular zone and the sub-ventricular zone of the anterolateral ventricles. However, growing evidence suggests that new cells are not only "born" constitutively in the adult hypothalamus, but many of these cells also differentiate into neurons and glia and serve specific functions. The preoptic-hypothalamic area plays a central role in the regulation of many critical functions, including sleep-wakefulness and circadian rhythms. While a role for adult hippocampal neurogenesis in regulating hippocampus-dependent functions, including cognition, has been extensively studied, adult hypothalamic neurogenic process and its contributions to various hypothalamic functions, including sleep-wake regulation are just beginning to unravel. This review is aimed at providing the current understanding of the hypothalamic adult neurogenic processes and the extent to which it affects hypothalamic functions, including sleep-wake regulation. We propose that hypothalamic neurogenic processes are vital for maintaining the proper functioning of the hypothalamic sleep-wake and circadian systems in the face of regulatory challenges. Sleep-wake disturbance is a frequent and challenging problem of aging and age-related neurodegenerative diseases. Aging is also associated with a decline in the neurogenic process. We discuss a hypothesis that a decrease in the hypothalamic neurogenic process underlies the aging of its sleep-wake and circadian systems and associated sleep-wake disturbance. We further discuss whether neuro-regenerative approaches, including pharmacological and non-pharmacological stimulation of endogenous neural stem and progenitor cells in hypothalamic neurogenic niches, can be used for mitigating sleep-wake and other hypothalamic dysfunctions in aging.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Md Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychiatry, University of California, Los Angeles, CA
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychology, University of California, Los Angeles, CA
| | - Md Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
10
|
Evans MC, Hill JW, Anderson GM. Role of insulin in the neuroendocrine control of reproduction. J Neuroendocrinol 2021; 33:e12930. [PMID: 33523515 DOI: 10.1111/jne.12930] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/18/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Infertility associated with insulin resistance is characterised by abnormal hormone secretion by the hypothalamus, pituitary gland and gonads. These endocrine tissues can maintain insulin sensitivity even when tissues such as the muscle and liver become insulin-resistant, resulting in excessive insulin stimulation as hyperinsulinaemia develops. Experiments conducted to determine the role of neuronal insulin signalling in fertility were unable to recapitulate early findings of hypogonadotrophic hypogonadism in mice lacking insulin receptors throughout the brain. Rather, it was eventually shown that astrocytes critically mediate the effects of insulin on puberty timing and adult reproductive function. However, specific roles for neurones and gonadotrophs have been revealed under conditions of hyperinsulinaemia and by ablation of insulin and leptin receptors. The collective picture is one of multiple insulin-responsive inputs to gonadotrophin releasing hormone neurones, with astrocytes being the most important player.
Collapse
Affiliation(s)
- Maggie C Evans
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, USA
| | - Greg M Anderson
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
11
|
Tanycytes in the infundibular nucleus and median eminence and their role in the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:253-273. [PMID: 34225934 DOI: 10.1016/b978-0-12-820107-7.00016-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier is generally attributed to endothelial cells. However, in circumventricular organs, such as the median eminence, tanycytes take over the barrier function. These ependymoglial cells form the wall of the third ventricle and send long extensions into the parenchyma to contact blood vessels and hypothalamic neurons. The shape and location of tanycytes put them in an ideal position to connect the periphery with central nervous compartments. In line with this, tanycytes control the transport of hormones and key metabolites in and out of the hypothalamus. They function as sensors of peripheral homeostasis for central regulatory networks. This chapter discusses current evidence that tanycytes play a key role in regulating glucose balance, food intake, endocrine axes, seasonal changes, reproductive function, and aging. The understanding of how tanycytes perform these diverse tasks is only just beginning to emerge and will probably lead to a more differentiated view of how the brain and the periphery interact.
Collapse
|
12
|
Smedlund KB, Hill JW. The role of non-neuronal cells in hypogonadotropic hypogonadism. Mol Cell Endocrinol 2020; 518:110996. [PMID: 32860862 DOI: 10.1016/j.mce.2020.110996] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/01/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
13
|
Prezotto LD, Thorson JF, Prevot V, Redmer DA, Grazul-Bilska AT. Nutritionally induced tanycytic plasticity in the hypothalamus of adult ewes. Domest Anim Endocrinol 2020; 72:106438. [PMID: 32388344 DOI: 10.1016/j.domaniend.2020.106438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier regulates the transport of molecules that convey global energetic status to the feeding circuitry within the hypothalamus. Capillaries within the median eminence (ME) and tight junctions between tanycytes lining the third ventricle (3V) are critical components of this barrier. Herein, we tested the hypothesis that altering the plane of nutrition results in the structural reorganization of tanycytes, tight junctions, and capillary structure within the medial basal hypothalamus. Proopiomelanocortin (POMC) neuronal content within the arcuate nucleus of the hypothalamus (ARC) was also assessed to test whether reduced nutritional status improved access of nutrients to the ARC, while decreasing the access of nutrients of overfed animals. Multiparous, nongestating ewes were stratified by weight and randomly assigned to dietary treatments offered for 75 d: 200% of dietary recommendations (overfed), 100% of dietary recommendations (control), or 60% of dietary recommendations (underfed). The number of POMC-expressing neurons within the ARC was increased (P ≤ 0.002) in underfed ewes. Overfeeding increased (P ≤ 0.01) tanycyte cellular process penetration and density compared with control and underfeeding as assessed using vimentin immunostaining. Immunostaining of tight junctions along the wall of the 3V did not differ (P = 0.32) between treatments. No differences were observed in capillary density (P = 0.21) or classification (P ≥ 0.47) within the ME. These results implicate that changes within the satiety center and morphology of tanycytes within the ARC occur as an adaptation to nutrient availability.
Collapse
Affiliation(s)
- L D Prezotto
- Nutritional & Reproductive Physiology Laboratory, Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre, MT 59501, USA.
| | - J F Thorson
- Nutritional & Reproductive Physiology Laboratory, Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre, MT 59501, USA
| | - V Prevot
- INSERM, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France
| | - D A Redmer
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - A T Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
14
|
Hypothalamic tanycytes generate acute hyperphagia through activation of the arcuate neuronal network. Proc Natl Acad Sci U S A 2020; 117:14473-14481. [PMID: 32513737 PMCID: PMC7322081 DOI: 10.1073/pnas.1919887117] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Tanycytes are nutrient-sensing cells that line the third ventricle within the hypothalamus. The role of tanycytes in the regulation of food intake has not been documented. Indeed, the mechanistic link between nutrient concentrations in the CSF and activation of neurons responsible for the regulation of food intake, such as orexigenic (NPY/AgRP) or anorexigenic (POMC) cells, is not yet clear. Here, we demonstrate that tanycytes, engineered to express channelrhodopsin, can activate arcuate neurons to induce acute hyperphagia when activated by light. These data provide further evidence that tanycytes are an integral link between CSF nutrients and the hypothalamic neuronal networks that regulate appetite and energy balance. Hypothalamic tanycytes are chemosensitive glial cells that contact the cerebrospinal fluid in the third ventricle and send processes into the hypothalamic parenchyma. To test whether they can activate neurons of the arcuate nucleus, we targeted expression of a Ca2+-permeable channelrhodopsin (CatCh) specifically to tanycytes. Activation of tanycytes ex vivo depolarized orexigenic (neuropeptide Y/agouti-related protein; NPY/AgRP) and anorexigenic (proopiomelanocortin; POMC) neurons via an ATP-dependent mechanism. In vivo, activation of tanycytes triggered acute hyperphagia only in the fed state during the inactive phase of the light–dark cycle.
Collapse
|
15
|
Dai X, Hong L, Shen H, Du Q, Ye Q, Chen X, Zhang J. Estradiol-induced senescence of hypothalamic astrocytes contributes to aging-related reproductive function declines in female mice. Aging (Albany NY) 2020; 12:6089-6108. [PMID: 32259796 PMCID: PMC7185128 DOI: 10.18632/aging.103008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022]
Abstract
Hypothalamic astrocytes are important contributors that activate gonadotropin-releasing hormone (GnRH) neurons and promote GnRH/LH (luteinizing hormone) surge. However, the potential roles and mechanisms of astrocytes during the early reproductive decline remain obscure. The current study reported that, in intact middle-aged female mice, astrocytes within the hypothalamic RP3V accumulated senescence-related markers with increasing age. It employed an ovariectomized animal model and a cell model receiving estrogen intervention to confirm the estrogen-induced senescence of hypothalamic astrocytes. It found that estrogen metabolites may be an important factor for the estrogen-induced astrocyte senescence. In vitro molecular analysis revealed that ovarian estradiol activated PKA and up-regulated CYPs expression, metabolizing estradiol into 2-OHE2 and 4-OHE2. Of note, in middle-aged mice, the progesterone synthesis and the ability to promote GnRH release were significantly reduced. Besides, the expression of growth factors decreased and the mRNA levels of proinflammatory cytokines significantly increased in the aging astrocytes. The findings confirm that ovarian estradiol induces the senescence of hypothalamic astrocytes and that the senescent astrocytes compromise the regulation of progesterone synthesis and GnRH secretion, which may contribute to the aging-related declines in female reproductive function.
Collapse
Affiliation(s)
- Xiaoman Dai
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Luyan Hong
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China.,Department of Biochemistry and Molecular Biology, Gannan Medical University, Ganzhou 341000, Jiangxi. China
| | - Hui Shen
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Qiang Du
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, FujianChina
| | - Qinyong Ye
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Xiaochun Chen
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Jing Zhang
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350001, Fujian, China
| |
Collapse
|
16
|
MacDonald AJ, Robb JL, Morrissey NA, Beall C, Ellacott KLJ. Astrocytes in neuroendocrine systems: An overview. J Neuroendocrinol 2019; 31:e12726. [PMID: 31050045 DOI: 10.1111/jne.12726] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/11/2022]
Abstract
A class of glial cell, astrocytes, is highly abundant in the central nervous system (CNS). In addition to maintaining tissue homeostasis, astrocytes regulate neuronal communication and synaptic plasticity. There is an ever-increasing appreciation that astrocytes are involved in the regulation of physiology and behaviour in normal and pathological states, including within neuroendocrine systems. Indeed, astrocytes are direct targets of hormone action in the CNS, via receptors expressed on their surface, and are also a source of regulatory neuropeptides, neurotransmitters and gliotransmitters. Furthermore, as part of the neurovascular unit, astrocytes can regulate hormone entry into the CNS. This review is intended to provide an overview of how astrocytes are impacted by and contribute to the regulation of a diverse range of neuroendocrine systems: energy homeostasis and metabolism, reproduction, fluid homeostasis, the stress response and circadian rhythms.
Collapse
Affiliation(s)
- Alastair J MacDonald
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Josephine L Robb
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicole A Morrissey
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Neuroendocrine Research Group, Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
17
|
Denis J, Dangouloff-Ros V, Pinto G, Flechtner I, Piketty M, Samara D, Levy R, Grévent D, Millischer AE, Brunelle F, Prevot V, Polak M, Boddaert N. Arterial Spin Labeling and Central Precocious Puberty. Clin Neuroradiol 2018; 30:137-144. [DOI: 10.1007/s00062-018-0738-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/19/2018] [Indexed: 12/23/2022]
|
18
|
Abstract
Astrocytes have historically been considered structural supporting cells for neurons. Thanks to new molecular tools, allowing specific cell ablation or over-expression of genes, new unexpected astrocytic functions have recently been unveiled. This review focus on emerging groundbreaking findings showing that hypothalamic astrocytes are pivotal for the regulation of whole body energy homeostasis. Hypothalamic astrocytes sense glucose and fatty acids, and express receptors for several peripheral hormones such as leptin and insulin. Furthermore, they display striking sexual dimorphism which may account, at least partially, for gender specific differences in energy homeostasis. Metabolic alterations have been shown to influence the initiation and progression of many neurodegenerative disorders. A better understanding of the roles and interplay between the different brain cells in regulating energy homeostasis could help develop new therapeutic strategies to prevent or cure neurodegenerative disorders.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
19
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
20
|
Pellegrino G, Trubert C, Terrien J, Pifferi F, Leroy D, Loyens A, Migaud M, Baroncini M, Maurage CA, Fontaine C, Prévot V, Sharif A. A comparative study of the neural stem cell niche in the adult hypothalamus of human, mouse, rat and gray mouse lemur (Microcebus murinus). J Comp Neurol 2018; 526:1419-1443. [PMID: 29230807 DOI: 10.1002/cne.24376] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
The adult brain contains niches of neural stem cells that continuously add new neurons to selected circuits throughout life. Two niches have been extensively studied in various mammalian species including humans, the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus. Recently, studies conducted mainly in rodents have identified a third neurogenic niche in the adult hypothalamus. In order to evaluate whether a neural stem cell niche also exists in the adult hypothalamus in humans, we performed multiple immunofluorescence labeling to assess the expression of a panel of neural stem/progenitor cell (NPC) markers (Sox2, nestin, vimentin, GLAST, GFAP) in the human hypothalamus and compared them with the mouse, rat and a non-human primate species, the gray mouse lemur (Microcebus murinus). Our results show that the adult human hypothalamus contains four distinct populations of cells that express the five NPC markers: (a) a ribbon of small stellate cells that lines the third ventricular wall behind a hypocellular gap, similar to that found along the lateral ventricles, (b) ependymal cells, (c) tanycytes, which line the floor of the third ventricle in the tuberal region, and (d) a population of small stellate cells in the suprachiasmatic nucleus. In the mouse, rat and mouse lemur hypothalamus, co-expression of NPC markers is primarily restricted to tanycytes, and these species lack a ventricular ribbon. Our work thus identifies four cell populations with the antigenic profile of NPCs in the adult human hypothalamus, of which three appear specific to humans.
Collapse
Affiliation(s)
- Giuliana Pellegrino
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France
| | - Claire Trubert
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France
| | - Jérémy Terrien
- MECADEV UMR 7179, Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Fabien Pifferi
- MECADEV UMR 7179, Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Danièle Leroy
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France
| | - Anne Loyens
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France
| | - Martine Migaud
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France.,CNRS, UMR7247, Nouzilly, France; Université de Tours, Tours, France.,Institut Français du Cheval et de l'Equitation (IFCE), Nouzilly, France
| | - Marc Baroncini
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France.,Department of Neurosurgery, Lille University Hospital, Lille, France
| | - Claude-Alain Maurage
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France.,Department of Neuropathology, Lille University Hospital, Lille, France
| | - Christian Fontaine
- University of Lille, School of Medicine, Lille Cedex, France.,Laboratory of Anatomy, Lille University Hospital, Lille, France
| | - Vincent Prévot
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France
| | - Ariane Sharif
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Lille Cedex, France.,University of Lille, School of Medicine, Lille Cedex, France
| |
Collapse
|
21
|
Clasadonte J, Prevot V. The special relationship: glia-neuron interactions in the neuroendocrine hypothalamus. Nat Rev Endocrinol 2018; 14:25-44. [PMID: 29076504 DOI: 10.1038/nrendo.2017.124] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Natural fluctuations in physiological conditions require adaptive responses involving rapid and reversible structural and functional changes in the hypothalamic neuroendocrine circuits that control homeostasis. Here, we discuss the data that implicate hypothalamic glia in the control of hypothalamic neuroendocrine circuits, specifically neuron-glia interactions in the regulation of neurosecretion as well as neuronal excitability. Mechanistically, the morphological plasticity displayed by distal processes of astrocytes, pituicytes and tanycytes modifies the geometry and diffusion properties of the extracellular space. These changes alter the relationship between glial cells of the hypothalamus and adjacent neuronal elements, especially at specialized intersections such as synapses and neurohaemal junctions. The structural alterations in turn lead to functional plasticity that alters the release and spread of neurotransmitters, neuromodulators and gliotransmitters, as well as the activity of discrete glial signalling pathways that mediate feedback by peripheral signals to the hypothalamus. An understanding of the contributions of these and other non-neuronal cell types to hypothalamic neuroendocrine function is thus critical both to understand physiological processes such as puberty, the maintenance of bodily homeostasis and ageing and to develop novel therapeutic strategies for dysfunctions of these processes, such as infertility and metabolic disorders.
Collapse
Affiliation(s)
- Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Bâtiment Biserte, 1 Place de Verdun, 59045, Lille, Cedex, France
- University of Lille, FHU 1000 days for Health, School of Medicine, Lille 59000, France
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Bâtiment Biserte, 1 Place de Verdun, 59045, Lille, Cedex, France
- University of Lille, FHU 1000 days for Health, School of Medicine, Lille 59000, France
| |
Collapse
|
22
|
Lévy F, Batailler M, Meurisse M, Migaud M. Adult Neurogenesis in Sheep: Characterization and Contribution to Reproduction and Behavior. Front Neurosci 2017; 11:570. [PMID: 29109674 PMCID: PMC5660097 DOI: 10.3389/fnins.2017.00570] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/28/2017] [Indexed: 01/18/2023] Open
Abstract
Sheep have many advantages to study neurogenesis in comparison to the well-known rodent models. Their development and life expectancy are relatively long and they possess a gyrencephalic brain. Sheep are also seasonal breeders, a characteristic that allows studying the involvement of hypothalamic neurogenesis in the control of seasonal reproduction. Sheep are also able to individually recognize their conspecifics and develop selective and lasting bonds. Adult olfactory neurogenesis could be adapted to social behavior by supporting recognition of conspecifics. The present review reveals the distinctive features of the hippocampal, olfactory, and hypothalamic neurogenesis in sheep. In particular, the organization of the subventricular zone and the dynamic of neuronal maturation differs from that of rodents. In addition, we show that various physiological conditions, such as seasonal reproduction, gestation, and lactation differently modulate these three neurogenic niches. Last, we discuss recent evidence indicating that hypothalamic neurogenesis acts as an important regulator of the seasonal control of reproduction and that olfactory neurogenesis could be involved in odor processing in the context of maternal behavior.
Collapse
Affiliation(s)
- Frederic Lévy
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Martine Batailler
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Maryse Meurisse
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Martine Migaud
- Institut National de la Recherche Agronomique, UMR85, Centre National de la Recherche Scientifique, UMR7247, Université F. Rabelais, IFCE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| |
Collapse
|
23
|
Pouchain Ribeiro Neto R, Clarke IJ, Conductier G. Alteration in the relationship between tanycytes and gonadotrophin-releasing hormone neurosecretory terminals following long-term metabolic manipulation in the sheep. J Neuroendocrinol 2017; 29. [PMID: 28722251 DOI: 10.1111/jne.12509] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/30/2022]
Abstract
The activity of the hypothalamic-pituitary gonadal axis is influenced by energy reserves, such that an increase or a decrease in adiposity may perturb the secretion and action of gonadotrophin-releasing hormone (GnRH). This is considered to be a result of the signalling of hormones such as leptin, which act upon neuronal systems controlling GnRH secretion. Other work shows plasticity in the relationship between tanycytes and GnRH neurosecretory terminals in the median eminence across the oestrous cycle and we hypothesised that a similar plasticity may occur with altered metabolic status. We studied Lean, Normal and Fat ovariectomised ewes, which displayed differences in gonadotrophin status, and investigated the relationship between tanycytes and GnRH neuroterminals. Under both Lean and Fat conditions, an altered anatomical arrangement between these two elements was observed in the vicinity of the blood vessels of the primary plexus of the hypophysial portal blood system. These data suggest that such plasticity is an important determinant of the rate of secretion of GnRH in animals of differing metabolic status and that this also contributes to the relative hypogonadotrophic condition prevailing with metabolic extremes.
Collapse
Affiliation(s)
- R Pouchain Ribeiro Neto
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
- Neuroscience Program, Monash Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - I J Clarke
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
- Neuroscience Program, Monash Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - G Conductier
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
- Neuroscience Program, Monash Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Koopman ACM, Taziaux M, Bakker J. Age-related changes in the morphology of tanycytes in the human female infundibular nucleus/median eminence. J Neuroendocrinol 2017; 29. [PMID: 28295754 DOI: 10.1111/jne.12467] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 01/21/2023]
Abstract
Tanycytes are emerging as key players in the neuroendocrine control of gonadotrophin-releasing hormone (GnRH) release. Rodent studies have demonstrated that the structural relationship between tanycytes and GnRH terminals in the median eminence is highly dynamic, regulated by gonadal steroids and undergoes age-related changes. The present study aimed to determine whether the number and organisation of tanycytes changes throughout life in the female infundibular nucleus/median eminence (INF/ME) region. Post-mortem hypothalamic tissues were collected at the Netherlands Brain Bank and were stained for vimentin by immunohistochemistry. Hypothalami of 22 control female subjects were categorised into three periods: infant/prepubertal, adult and elderly. We measured the fractional area covered by vimentin immunoreactivity in the INF. Qualitative analysis demonstrated a remarkable parallel organisation of vimentin-immunoreactive processes during the infant/prepubertal and adult periods. During the elderly period, this organisation was largely lost. Semi-quantitatively, the fractional area covered in vimentin immunoreactivity was significantly higher at the infant/prepubertal compared to the adult period and almost reached statistical significance compared to the elderly period. By contrast, the number of tanycyte cell bodies did not appear to change throughout life. The results of the present study thus demonstrate that the number and structure of tanycytic processes are altered during ageing, suggesting that tanycytes might be involved in the age-related changes observed in GnRH release.
Collapse
Affiliation(s)
- A C M Koopman
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | - M Taziaux
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | - J Bakker
- GIGA Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
25
|
Evans MC, Anderson GM. Neuroendocrine integration of nutritional signals on reproduction. J Mol Endocrinol 2017; 58:R107-R128. [PMID: 28057770 DOI: 10.1530/jme-16-0212] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Reproductive function in mammals is energetically costly and therefore tightly regulated by nutritional status. To enable this integration of metabolic and reproductive function, information regarding peripheral nutritional status must be relayed centrally to the gonadotropin-releasing hormone (GNRH) neurons that drive reproductive function. The metabolically relevant hormones leptin, insulin and ghrelin have been identified as key mediators of this 'metabolic control of fertility'. However, the neural circuitry through which they act to exert their control over GNRH drive remains incompletely understood. With the advent of Cre-LoxP technology, it has become possible to perform targeted gene-deletion and gene-rescue experiments and thus test the functional requirement and sufficiency, respectively, of discrete hormone-neuron signaling pathways in the metabolic control of reproductive function. This review discusses the findings from these investigations, and attempts to put them in context with what is known from clinical situations and wild-type animal models. What emerges from this discussion is clear evidence that the integration of nutritional signals on reproduction is complex and highly redundant, and therefore, surprisingly difficult to perturb. Consequently, the deletion of individual hormone-neuron signaling pathways often fails to cause reproductive phenotypes, despite strong evidence that the targeted pathway plays a role under normal physiological conditions. Although transgenic studies rarely reveal a critical role for discrete signaling pathways, they nevertheless prove to be a good strategy for identifying whether a targeted pathway is absolutely required, critically involved, sufficient or dispensable in the metabolic control of fertility.
Collapse
Affiliation(s)
- Maggie C Evans
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| |
Collapse
|
26
|
Chen W, Balland E, Cowley MA. Hypothalamic Insulin Resistance in Obesity: Effects on Glucose Homeostasis. Neuroendocrinology 2017; 104:364-381. [PMID: 28122381 DOI: 10.1159/000455865] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
The central link between obesity and type 2 diabetes is the development of insulin resistance. To date, it is still not clear whether hyperinsulinemia causes insulin resistance, which underlies the pathogenesis of obesity-associated type 2 diabetes, owing to the sophisticated regulatory mechanisms that exist in the periphery and in the brain. In recent years, accumulating evidence has demonstrated the existence of insulin resistance within the hypothalamus. In this review, we have integrated the recent discoveries surrounding both central and peripheral insulin resistance to provide a comprehensive overview of insulin resistance in obesity and the regulation of systemic glucose homeostasis. In particular, this review will discuss how hyperinsulinemia and hyperleptinemia in obesity impair insulin sensitivity in tissues such as the liver, skeletal muscle, adipose tissue, and the brain. In addition, this review highlights insulin transport into the brain, signaling pathways associated with hypothalamic insulin receptor expression in the regulation of hepatic glucose production, and finally the perturbation of systemic glucose homeostasis as a consequence of central insulin resistance. We also suggest future approaches to overcome both central and peripheral insulin resistance to treat obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Physiology/Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
27
|
Miyata S. Advances in Understanding of Structural Reorganization in the Hypothalamic Neurosecretory System. Front Endocrinol (Lausanne) 2017; 8:275. [PMID: 29089925 PMCID: PMC5650978 DOI: 10.3389/fendo.2017.00275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic neurosecretory system synthesizes neuropeptides in hypothalamic nuclei and releases them from axonal terminals into the circulation in the neurohypophysis (NH) and median eminence (ME). This system plays a crucial role in regulating body fluid homeostasis and social behaviors as well as reproduction, growth, metabolism, and stress responses, and activity-dependent structural reorganization has been reported. Current knowledge on dynamic structural reorganization in the NH and ME, in which the axonal terminals of neurosecretory neurons directly contact the basement membrane (BM) of a fenestrated vasculature, is discussed herein. Glial cells, pituicytes in the NH and tanycytes in the ME, engulf axonal terminals and interpose their cellular processes between axonal terminals and the BM when hormonal demands are low. Increasing demands for neurosecretion result in the retraction of the cellular processes of glial cells from axonal terminals and the BM, permitting increased neurovascular contact. The shape conversion of pituicytes and tanycytes is mediated by neurotransmitters and sex steroid hormones, respectively. The NH and ME have a rough vascular BM profile of wide perivascular spaces and specialized extension structures called "perivascular protrusions." Perivascular protrusions, the insides of which are occupied by the cellular processes of vascular mural cells pericytes, contribute to increasing neurovascular contact and, thus, the efficient diffusion of hypothalamic neuropeptides. A chronic physiological stimulation has been shown to increase perivascular protrusions via the shape conversion of pericytes and the profile of the vascular surface. Continuous angiogenesis occurs in the NH and ME of healthy normal adult rodents depending on the signaling of vascular endothelial growth factor (VEGF). The inhibition of VEGF signaling suppresses the proliferation of endothelial cells (ECs) and promotes their apoptosis, which results in decreases in the population of ECs and axonal terminals. Pituicytes and tanycytes are continuously replaced by the proliferation and differentiation of stem/progenitor cells, which may be regulated by matching those of ECs and axonal terminals. In conclusion, structural reorganization in the NH and ME is caused by the activity-dependent shape conversion of glial cells and vascular mural cells as well as the proliferation of endothelial and glial cells by angiogenesis and gliogenesis, respectively.
Collapse
Affiliation(s)
- Seiji Miyata
- Department of Applied Biology, The Center for Advanced Insect Research Promotion (CAIRP), Kyoto Institute of Technology, Kyoto, Japan
- *Correspondence: Seiji Miyata,
| |
Collapse
|
28
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
29
|
Chowen JA, Argente-Arizón P, Freire-Regatillo A, Frago LM, Horvath TL, Argente J. The role of astrocytes in the hypothalamic response and adaptation to metabolic signals. Prog Neurobiol 2016; 144:68-87. [PMID: 27000556 DOI: 10.1016/j.pneurobio.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
The hypothalamus is crucial in the regulation of homeostatic functions in mammals, with the disruption of hypothalamic circuits contributing to chronic conditions such as obesity, diabetes mellitus, hypertension, and infertility. Metabolic signals and hormonal inputs drive functional and morphological changes in the hypothalamus in attempt to maintain metabolic homeostasis. However, the dramatic increase in the incidence of obesity and its secondary complications, such as type 2 diabetes, have evidenced the need to better understand how this system functions and how it can go awry. Growing evidence points to a critical role of astrocytes in orchestrating the hypothalamic response to metabolic cues by participating in processes of synaptic transmission, synaptic plasticity and nutrient sensing. These glial cells express receptors for important metabolic signals, such as the anorexigenic hormone leptin, and determine the type and quantity of nutrients reaching their neighboring neurons. Understanding the mechanisms by which astrocytes participate in hypothalamic adaptations to changes in dietary and metabolic signals is fundamental for understanding the neuroendocrine control of metabolism and key in the search for adequate treatments of metabolic diseases.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain.
| | - Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
30
|
Naugle MM, Lozano SA, Guarraci FA, Lindsey LF, Kim JE, Morrison JH, Janssen WG, Yin W, Gore AC. Age and Long-Term Hormone Treatment Effects on the Ultrastructural Morphology of the Median Eminence of Female Rhesus Macaques. Neuroendocrinology 2016; 103:650-64. [PMID: 26536204 PMCID: PMC4860175 DOI: 10.1159/000442015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022]
Abstract
The median eminence (ME) of the hypothalamus comprises the hypothalamic nerve terminals, glia (especially tanycytes) and the portal capillary vasculature that transports hypothalamic neurohormones to the anterior pituitary gland. The ultrastructure of the ME is dynamically regulated by hormones and undergoes organizational changes during development and reproductive cycles in adult females, but relatively little is known about the ME during aging, especially in nonhuman primates. Therefore, we used a novel transmission scanning electron microscopy technique to examine the cytoarchitecture of the ME of young and aged female rhesus macaques in a preclinical monkey model of menopausal hormone treatments. Rhesus macaques were ovariectomized and treated for 2 years with vehicle, estradiol (E2), or estradiol + progesterone (E2 + P4). While the overall cytoarchitecture of the ME underwent relatively few changes with age and hormones, changes to some features of neural and glial components near the portal capillaries were observed. Specifically, large neuroterminal size was greater in aged compared to young adult animals, an effect that was mitigated or reversed by E2 alone but not by E2 + P4 treatment. Overall glial size and the density and tissue fraction of the largest subset of glia were greater in aged monkeys, and in some cases reversed by E2 treatment. Mitochondrial size was decreased by E2, but not E2 + P4, only in aged macaques. These results contrast substantially with work in rodents, suggesting that the ME of aging macaques is less vulnerable to age-related disorganization, and that the effects of E2 on monkeys' ME are age specific.
Collapse
Affiliation(s)
| | - Sateria A. Lozano
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - Fay A. Guarraci
- Department of Psychology, Southwestern University, Georgetown, TX
| | - Larry F. Lindsey
- Center for Learning and Memory, University of Texas at Austin, Austin, TX
| | - Ji E. Kim
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - John H. Morrison
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - William G.M. Janssen
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Weiling Yin
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
| | - Andrea C. Gore
- Institute for Neuroscience, University of Texas at Austin, Austin, TX
- Division of Pharmacology & Toxicology, University of Texas at Austin, Austin, TX
- Institute for Cellular & Molecular Biology, University of Texas at Austin, Austin, TX
- Correspondence: Andrea C Gore, PhD, The University of Texas at Austin, 107 West Dean Keeton, C0875, Austin, TX, 78712, USA, ; Tel: +1-512-471-3669; Fax: +1-512-471-5002
| |
Collapse
|
31
|
Evans MC, Rizwan MZ, Anderson GM. Insulin Does Not Target CamkIIα Neurones to Critically Regulate the Neuroendocrine Reproductive Axis in Mice. J Neuroendocrinol 2015; 27:899-910. [PMID: 26485112 DOI: 10.1111/jne.12330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 10/05/2015] [Accepted: 10/14/2015] [Indexed: 11/28/2022]
Abstract
Insulin signalling in the brain plays an important role in the central regulation of energy homeostasis and fertility, such that mice exhibiting widespread deletion of insulin receptors (InsR) throughout the brain and peripheral nervous system display diet sensitive obesity and hypothalamic hypogonadism. However, the specific cell types mediating the central effects of insulin on fertility remain largely unidentified. To date, the targeted deletion of InsR from individual neuronal populations implicated in the metabolic control of fertility has failed to recapitulate the hypogonadic and subfertile phenotype observed in brain-specific InsR knockout mice. Because insulin and leptin share similar roles as centrally-acting metabolic regulators of fertility, we used the Cre-loxP system to generate mice with a selective inactivation of the Insr gene from the same widespread neuronal population previously shown to mediate the central effects of leptin on fertility by crossing Insr-flox mice with calcium/calmodulin-dependent protein kinase type IIα (CamkIIα)-Cre mice. Multiple reproductive and metabolic parameters were then compared between male and female Insr-flox/Cre-positive (CamK-IRKO) and Insr-flox/Cre-negative control mice. Consistent with brain-specific InsR knockout mice, CamK-IRKO mice exhibited a mild but significant obesogenic phenotype. Unexpectedly, CamK-IRKO mice exhibited normal reproductive maturation and function compared to controls. No differences in the age of puberty onset, oestrous cyclicity or fecundity were observed between CamK-IRKO and control mice. We conclude that the central effects of insulin on the neuroendocrine reproductive axis are not critically mediated via the same neuronal populations targeted by leptin.
Collapse
Affiliation(s)
- M C Evans
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - M Z Rizwan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - G M Anderson
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Medical Sciences, Dunedin, New Zealand
| |
Collapse
|
32
|
Sensitivity to the photoperiod and potential migratory features of neuroblasts in the adult sheep hypothalamus. Brain Struct Funct 2015; 221:3301-14. [DOI: 10.1007/s00429-015-1101-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/27/2015] [Indexed: 12/14/2022]
|
33
|
Migaud M, Butrille L, Batailler M. Seasonal regulation of structural plasticity and neurogenesis in the adult mammalian brain: focus on the sheep hypothalamus. Front Neuroendocrinol 2015; 37:146-57. [PMID: 25462590 DOI: 10.1016/j.yfrne.2014.11.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 01/19/2023]
Abstract
To cope with variations in the environment, most mammalian species exhibit seasonal cycles in physiology and behaviour. Seasonal plasticity during the lifetime contributes to seasonal physiology. Over the years, our ideas regarding adult brain plasticity and, more specifically, hypothalamic plasticity have greatly evolved. Along with the two main neurogenic regions, namely the hippocampal subgranular and lateral ventricle subventricular zones, the hypothalamus, which is the central homeostatic regulator of numerous physiological functions that comprise sexual behaviours, feeding and metabolism, also hosts neurogenic niches. Both endogenous and exogenous factors, including the photoperiod, modulate the hypothalamic neurogenic capacities. The present review describes the effects of season on adult morphological plasticity and neurogenesis in seasonal species, for which the photoperiod is a master environmental cue for the successful programming of seasonal functions. In addition, the potential functional significance of adult neurogenesis in the mediation of the seasonal control of reproduction and feeding is discussed.
Collapse
Affiliation(s)
- Martine Migaud
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université de Tours, F-37041 Tours, France; Haras Nationaux, F-37380 Nouzilly, France.
| | - Lucile Butrille
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université de Tours, F-37041 Tours, France; Haras Nationaux, F-37380 Nouzilly, France
| | - Martine Batailler
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université de Tours, F-37041 Tours, France; Haras Nationaux, F-37380 Nouzilly, France
| |
Collapse
|
34
|
Batailler M, Droguerre M, Baroncini M, Fontaine C, Prevot V, Migaud M. DCX-expressing cells in the vicinity of the hypothalamic neurogenic niche: a comparative study between mouse, sheep, and human tissues. J Comp Neurol 2014; 522:1966-85. [PMID: 24288185 DOI: 10.1002/cne.23514] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 12/31/2022]
Abstract
Neural stem and precursor cells persist postnatally throughout adulthood and are capable of responding to numerous endogenous and exogenous signals by modifying their proliferation and differentiation. Whereas adult neurogenesis has been extensively studied in the dentate gyrus of the hippocampal formation and in the subventricular zone adjacent to the wall of the lateral ventricles, we and others have recently reported constitutive adult neurogenesis in other brain structures, including the hypothalamus. In this study, we used immunohistochemistry to study the expression of the neuroblast marker doublecortin (DCX), and compared its expression pattern in adult ovine, mouse, and human hypothalamic tissues. Our results indicate that DCX-positive cells resembling immature and developing neurons occur in a wide range of hypothalamic nuclei in all three species, although with different distribution patterns. In addition, the morphology of DCX-positive cells varied depending on their location. DCX-positive cells near the third ventricle had the morphology of very immature neuroblasts, a round shape with no processes, whereas those located deeper in the parenchyma such as in the ventromedial nucleus were fusiform and showed a bipolar morphology. Extending this observation, we showed that among the cohort of immature neurons entering the ventromedial nucleus, some appeared to undergo maturation, as revealed by the partial colocalization of DCX with markers of more mature neurons, e.g., human neuronal protein C and D (HuC/D). This study provides further confirmation of the existence of an adult hypothalamic neurogenic niche and argues for the potential existence of a migratory path within the hypothalamus.
Collapse
Affiliation(s)
- Martine Batailler
- INRA, Unité Mixte de Recherche (UMR) 85 Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France; Institut Français du Cheval et de l'équitation (IFCE), F-37380, Nouzilly, France
| | | | | | | | | | | |
Collapse
|
35
|
Sharif A, Ojeda SR, Prevot V. Neurogenesis and Gliogenesis in the Postnatal Hypothalamus: A New Level of Plasticity for the Regulation of Hypothalamic Function? ENDOGENOUS STEM CELL-BASED BRAIN REMODELING IN MAMMALS 2014. [DOI: 10.1007/978-1-4899-7399-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Steinman MQ, Valenzuela AE, Siopes TD, Millam JR. Tuberal hypothalamic expression of the glial intermediate filaments, glial fibrillary acidic protein and vimentin across the turkey hen (Meleagris gallopavo) reproductive cycle: Further evidence for a role of glial structural plasticity in seasonal reproduction. Gen Comp Endocrinol 2013; 193:141-8. [PMID: 23948371 PMCID: PMC3812377 DOI: 10.1016/j.ygcen.2013.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 08/01/2013] [Accepted: 08/04/2013] [Indexed: 11/25/2022]
Abstract
Glia regulate the hypothalamic-pituitary-gonadal (HPG) axis in birds and mammals. This is accomplished mechanically by ensheathing gonadotrophin-releasing hormone I (GnRH) nerve terminals thereby blocking access to the pituitary blood supply, or chemically in a paracrine manner. Such regulation requires appropriate spatial associations between glia and nerve terminals. Female turkeys (Meleagris gallopavo) use day length as a primary breeding cue. Long days activate the HPG-axis until the hen enters a photorefractory state when previously stimulatory day lengths no longer support HPG-axis activity. Hens must then be exposed to short days before reactivation of the reproductive axis occurs. As adult hens have discrete inactive reproductive states in addition to a fertile state, they are useful for examining the glial contribution to reproductive function. We immunostained tuberal hypothalami from short and long-day photosensitive hens, plus long-day photorefractory hens to examine expression of two intermediate filaments that affect glial morphology: glial fibrillary acidic protein (GFAP) and vimentin. GFAP expression was drastically reduced in the central median eminence of long day photosensitive hens, especially within the internal zone. Vimentin expression was similar among groups. However, vimentin-immunoreactive fibers abutting the portal vasculature were significantly negatively correlated with GFAP expression in the median eminence, which is consistent with our hypothesis for a reciprocal relationship between GFAP and vimentin expression. It appears that up-regulation of GFAP expression in the central median eminence of turkey hens is associated with periods of reproductive quiescence and that photofractoriness is associated with the lack of a glial cytoskeletal response to long days.
Collapse
Affiliation(s)
- Michael Q Steinman
- Molecular, Cellular and Integrative Physiology Graduate Group, University of California, Davis, CA 95616, USA; Department of Psychology, University of California, Davis, CA 95616, USA.
| | | | | | | |
Collapse
|
37
|
Brauksiepe B, Baumgarten L, Reuss S, Schmidt ER. Co-localization of serine/threonine kinase 33 (Stk33) and vimentin in the hypothalamus. Cell Tissue Res 2013; 355:189-99. [PMID: 24057876 DOI: 10.1007/s00441-013-1721-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 08/16/2013] [Indexed: 01/04/2023]
Abstract
We investigate the immunoreactivity of serine/threonine kinase 33 (Stk33) and of vimentin in the brain of mouse, rat and hamster. Using a Stk33-specific polyclonal antibody, we show by immunofluorescence staining that Stk33 is present in a variety of brain regions. We found a strong staining in the ependymal lining of all cerebral ventricles and the central canal of the spinal cord as well as in hypothalamic tanycytes. Stk33 immunoreactivity was also found in circumventricular organs such as the area postrema, subfornical organ and pituitary and pineal glands. Double-immunostaining experiments with antibodies against Stk33 and vimentin showed a striking colocalization of Stk33 and vimentin. As shown previously, Stk33 phosphorylates recombinant vimentin in vitro. Co-immunoprecipitation experiments and co-sedimentation assays indicate that Stk33 and vimentin are associated in vivo and that this association does not depend on further interacting partners (Brauksiepe et al. in BMC Biochem 9:25, 2008). This indicates that Stk33 is involved in the dynamics of vimentin polymerization/depolymerization. Since in tanycytes the vimentin expression is regulated by the photoperiod (Kameda et al. in Cell Tissue Res 314:251-262, 2003), we determine whether this also holds true for Stk33. We study hypothalamic sections from adult Djungarian hamsters (Phodopus sungorus) held under either long photoperiods (L:D 16:8 h) or short photoperiods (L:D 8:16 h) for 2 months. In addition, we examine whether age-dependent changes in Stk33 protein content exist. Our results show that Stk33 in tanycytes is regulated by the photoperiod as is the case for vimentin. Stk33 may participate in photoperiodic regulation of the endocrine system.
Collapse
Affiliation(s)
- Bastienne Brauksiepe
- Institute of Molecular Genetics, Johannes Gutenberg-University Mainz, Johann-Joachim Becherweg 32, 55128, Mainz, Germany
| | | | | | | |
Collapse
|
38
|
Bolborea M, Dale N. Hypothalamic tanycytes: potential roles in the control of feeding and energy balance. Trends Neurosci 2013; 36:91-100. [PMID: 23332797 PMCID: PMC3605593 DOI: 10.1016/j.tins.2012.12.008] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/11/2012] [Accepted: 12/18/2012] [Indexed: 02/08/2023]
Abstract
Tanycytes, glial-like cells that line the third ventricle, are emerging as components of the hypothalamic networks that control body weight and energy balance. They contact the cerebrospinal fluid (CSF) and send processes that come into close contact with neurons in the arcuate and ventromedial hypothalamic nuclei. Tanycytes are glucosensitive and are able to respond to transmitters associated with arousal and the drive to feed. At least some tanycytes are stem cells and, in the median eminence, may be stimulated by diet to generate new neurons. The quest is on to understand how tanycytes detect and respond to changes in energy balance and how they communicate with the rest of the nervous system to effect their functions.
Collapse
Affiliation(s)
- Matei Bolborea
- School of Life Sciences, University of Warwick, Coventry, UK
| | | |
Collapse
|
39
|
Sharif A, Baroncini M, Prevot V. Role of glia in the regulation of gonadotropin-releasing hormone neuronal activity and secretion. Neuroendocrinology 2013; 98:1-15. [PMID: 23735672 DOI: 10.1159/000351867] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/08/2013] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the final common pathway for the central control of reproduction. The coordinated and timely activation of these hypothalamic neurons, which determines sexual development and adult reproductive function, lies under the tight control of a complex array of excitatory and inhibitory transsynaptic inputs. In addition, research conducted over the past 20 years has unveiled the major contribution of glial cells to the control of GnRH neurons. Glia use a variety of molecular and cellular strategies to modulate GnRH neuronal function both at the level of their cell bodies and at their nerve terminals. These mechanisms include the secretion of bioactive molecules that exert paracrine effects on GnRH neurons, juxtacrine interactions between glial cells and GnRH neurons via adhesive molecules and the morphological plasticity of the glial coverage of GnRH neurons. It now appears that glial cells are integral components, along with upstream neuronal networks, of the central control of GnRH neuronal function. This review attempts to summarize our current knowledge of the mechanisms used by glial cells to control GnRH neuronal activity and secretion.
Collapse
Affiliation(s)
- Ariane Sharif
- INSERM, Jean-Pierre Aubert Research Center, Development and Plasticity of the Postnatal Brain, Unit 837, and UDSL, School of Medicine, Lille, France.
| | | | | |
Collapse
|
40
|
Kumar S, Parkash J, Kataria H, Kaur G. Enzymatic removal of polysialic acid from neural cell adhesion molecule interrupts gonadotropin releasing hormone (GnRH) neuron-glial remodeling. Mol Cell Endocrinol 2012; 348:95-103. [PMID: 21846489 DOI: 10.1016/j.mce.2011.07.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 07/20/2011] [Accepted: 07/22/2011] [Indexed: 11/22/2022]
Abstract
There is abundant evidence to prove that the astrocytes are highly dynamic cell type in CNS and under physiological conditions such as reproduction, these cells display a remarkable structural plasticity especially at the level of their distal processes ensheathing the gonadotropin releasing hormone (GnRH) axon terminals. The morphology of GnRH axon terminals and astrocytes in the median eminence region of hypothalamus show activity dependent structural plasticity during different phases of estrous cycle. In the current study, we have assessed the functional contribution of ∞-2,8-linked polysialic acid (PSA) on neural cell adhesion molecule (PSA-NCAM) in this neuronal-glial plasticity using both in vitro and in vivo model systems. In vivo experiments were carried out after stereotaxic injection of endoneuraminidase enzyme (endo-N) near median eminence region of hypothalamus to specifically remove PSA residues on NCAM followed by localization of GnRH, PSA-NCAM and glial fibrillary acidic protein (GFAP) by immunostaining. Using in vitro model, structural remodeling of GnV-3 cells, (a conditionally immortalized GnRH cell line) co-cultured with primary astrocytes was studied after treating the cells with endo-N. Marked morphological changes were observed in GnRH axon terminals in proestrous phase rats and control GnV-3 cells as compared to endo-N treatment i.e. after removal of PSA. The specificity of endo-N treatment was also confirmed by studying the expression of PSA-NCAM by Western blotting in cultures treated with and without endo-N. Removal of PSA from surfaces with endo-N prevented stimulation associated remodeling of GnRH axon terminals as well as their associated glial cells under both in vivo and in vitro conditions. The current data confirms the permissive role of PSA to promote dynamic remodeling of GnRH axon terminals and their associated glia during reproductive cycle in rats.
Collapse
Affiliation(s)
- Sushil Kumar
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, India
| | | | | | | |
Collapse
|
41
|
Glanowska KM, Moenter SM. Endocannabinoids and prostaglandins both contribute to GnRH neuron-GABAergic afferent local feedback circuits. J Neurophysiol 2011; 106:3073-81. [PMID: 21917995 DOI: 10.1152/jn.00046.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons form the final common pathway for central control of fertility. Regulation of GnRH neurons by long-loop gonadal steroid feedback through steroid receptor-expressing afferents such as GABAergic neurons is well studied. Recently, local central feedback circuits regulating GnRH neurons were identified. GnRH neuronal depolarization induces short-term inhibition of their GABAergic afferents via a mechanism dependent on metabotropic glutamate receptor (mGluR) activation. GnRH neurons are enveloped in astrocytes, which express mGluRs. GnRH neurons also produce endocannabinoids, which can be induced by mGluR activation. We hypothesized the local GnRH-GABA circuit utilizes glia-derived and/or cannabinoid mechanisms and is altered by steroid milieu. Whole cell voltage-clamp was used to record GABAergic postsynaptic currents (PSCs) from GnRH neurons before and after action potential-like depolarizations were mimicked. In GnRH neurons from ovariectomized (OVX) mice, this depolarization reduced PSC frequency. This suppression was blocked by inhibition of prostaglandin synthesis with indomethacin, by a prostaglandin receptor antagonist, or by a specific glial metabolic poison, together suggesting the postulate that prostaglandins, potentially glia-derived, play a role in this circuit. This circuit was also inhibited by a CB1 receptor antagonist or by blockade of endocannabinoid synthesis in GnRH neurons, suggesting an endocannabinoid element, as well. In females, local circuit inhibition persisted in androgen-treated mice but not in estradiol-treated mice or young ovary-intact mice. In contrast, local circuit inhibition was present in gonad-intact males. These data suggest GnRH neurons interact with their afferent neurons using multiple mechanisms and that these local circuits can be modified by both sex and steroid feedback.
Collapse
Affiliation(s)
- Katarzyna M Glanowska
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | | |
Collapse
|
42
|
Abstract
Non-technical summary The hypothalamus contains key neural circuits involved in the control of feeding and energy balance. Stimulated by the inexorable rise of obesity, there has been intense study of these neural circuits. However, the possible role of non-neuronal cells in the brain has not been extensively considered. We now demonstrate that hypothalamic tanycytes, cells that lie at the interface between the ventricular cerebrospinal fluid and the brain parenchyma, respond to both neuron-derived and circulating agents that signal energy status and arousal. Our study therefore suggests that tanycytes should now be considered as active signalling cells in the brain capable of responding to several types of input and having the potential to participate in the control of energy balance and feeding. Abstract The brain plays a vital role in the regulation of food intake, appetite and ultimately bodyweight. Neurons in the hypothalamic arcuate nucleus, the ventromedial hypothalamic nuclei (VMH) and the lateral hypothalamus are sensitive to a number of circulating signals such as leptin, grehlin, insulin and glucose. These neurons are part of a network that integrates this information to regulate feeding and appetite. Hypothalamic tanycytes contact the cerebral spinal fluid of the third ventricle and send processes into the parenchyma. A subset of tanycytes are located close to, and send processes towards, the hypothalamic nuclei that contain neurons that are glucosensitive and are involved in the regulation of feeding. Nevertheless the signalling properties of tanycytes remain largely unstudied. We now demonstrate that tanycytes signal via waves of intracellular Ca2+; they respond strongly to ATP, histamine and acetylcholine – transmitters associated with the drive to feed. Selective stimulation by glucose of tanycyte cell bodies evokes robust ATP-mediated Ca2+ responses. Tanycytes release ATP in response to glucose. Furthermore tanycytes also respond to non-metabolisable analogues of glucose. Although tanycytes have been proposed as glucosensors, our study provides the first direct demonstration of this hypothesis. Tanycytes must therefore now be considered as active signalling cells within the brain that can respond to a number of neuronally derived and circulating transmitters and metabolites.
Collapse
|
43
|
Mansuy V, Geller S, Rey JP, Campagne C, Boccard J, Poulain P, Prevot V, Pralong FP. Phenotypic and molecular characterization of proliferating and differentiated GnRH-expressing GnV-3 cells. Mol Cell Endocrinol 2011; 332:97-105. [PMID: 20937356 DOI: 10.1016/j.mce.2010.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 09/14/2010] [Accepted: 10/02/2010] [Indexed: 12/27/2022]
Abstract
GnRH neurons provide the primary driving force upon the neuroendocrine reproductive axis. Here we used GnV-3 cells, a model of conditionally immortalized GnRH-expressing neurons, to perform an analysis of cell cycle and compare the gene expression profile of proliferating cells with differentiated cells. In the proliferation medium, 45 ± 1.5% of GnV-3 cells are in S-phase by FACS analysis. In the differentiation medium, only 9 ± 0.9% of them are in S-phase, and they acquire the characteristic bipolar shape displayed by preoptic GnRH neurons in vivo. In addition, GnV-3 cells in the differentiated state exhibit electrophysiological properties characteristic of neurons. Transcriptomic analysis identified up-regulation of 1931 genes and down-regulation of 1270 genes in cells grown in the differentiation medium compared to cells in the proliferation medium. Subsequent gene ontology study indicated that genes over-expressed in proliferating GnV-3 cells were mainly involved in cell cycle regulations, whereas genes over-expressed in differentiated cells were mainly involved in processes of differentiation, neurogenesis and neuronal morphogenesis. Taken together, these data demonstrate the occurrence of morphological and physiological changes in GnV-3 cells between the proliferating and the differentiated state. Moreover, the genes differentially regulated between these two different states are providing novel pathways potentially important for a better understanding of the physiology of mature GnRH neurons.
Collapse
Affiliation(s)
- Virginie Mansuy
- Service of Endocrinology, Diabetology and Metabolism, University Hospital and Faculty of Biology and Medicine, 1011 Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Clasadonte J, Sharif A, Baroncini M, Prevot V. Gliotransmission by prostaglandin e(2): a prerequisite for GnRH neuronal function? Front Endocrinol (Lausanne) 2011; 2:91. [PMID: 22649391 PMCID: PMC3355930 DOI: 10.3389/fendo.2011.00091] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 11/17/2011] [Indexed: 02/06/2023] Open
Abstract
Over the past four decades it has become clear that prostaglandin E(2) (PGE(2)), a phospholipid-derived signaling molecule, plays a fundamental role in modulating the gonadotropin-releasing hormone (GnRH) neuroendocrine system and in shaping the hypothalamus. In this review, after a brief historical overview, we highlight studies revealing that PGE(2) released by glial cells such as astrocytes and tanycytes is intimately involved in the active control of GnRH neuronal activity and neurosecretion. Recent evidence suggests that hypothalamic astrocytes surrounding GnRH neuronal cell bodies may respond to neuronal activity with an activation of the erbB receptor tyrosine kinase signaling, triggering the release of PGE(2) as a chemical transmitter from the glia themselves, and, in turn, leading to the feedback regulation of GnRH neuronal activity. At the GnRH neurohemal junction, in the median eminence of the hypothalamus, PGE(2) is released by tanycytes in response to cell-cell signaling initiated by glial cells and vascular endothelial cells. Upon its release, PGE(2) causes the retraction of the tanycyte end-feet enwrapping the GnRH nerve terminals, enabling them to approach the adjacent pericapillary space and thus likely facilitating neurohormone diffusion from these nerve terminals into the pituitary portal blood. In view of these new insights, we suggest that synaptically associated astrocytes and perijunctional tanycytes are integral modulatory elements of GnRH neuronal function at the cell soma/dendrite and nerve terminal levels, respectively.
Collapse
Affiliation(s)
- Jerome Clasadonte
- Jean-Pierre Aubert Research Center, Inserm, U837, F-59000Lille, France
- Laboratory of Anatomy, Université Lille Nord de FranceLille, France
- School of Medicine, UDSLLille, France
| | - Ariane Sharif
- Jean-Pierre Aubert Research Center, Inserm, U837, F-59000Lille, France
- Laboratory of Anatomy, Université Lille Nord de FranceLille, France
- School of Medicine, UDSLLille, France
| | - Marc Baroncini
- Jean-Pierre Aubert Research Center, Inserm, U837, F-59000Lille, France
- Laboratory of Anatomy, Université Lille Nord de FranceLille, France
- School of Medicine, UDSLLille, France
- Department of Neurosurgery, CHULilleLille, France
| | - Vincent Prevot
- Jean-Pierre Aubert Research Center, Inserm, U837, F-59000Lille, France
- Laboratory of Anatomy, Université Lille Nord de FranceLille, France
- School of Medicine, UDSLLille, France
- *Correspondence: Vincent Prevot, INSERM U837, Bâtiment Biserte, Place de Verdun, 59045 Lille Cedex, France. e-mail:
| |
Collapse
|
45
|
Abstract
The median eminence at the base of the hypothalamus serves as an interface between the neural and peripheral endocrine systems. It releases hypothalamic-releasing hormones into the portal capillary bed for transport to the anterior pituitary, which provides further signals to target endocrine systems. Of specific relevance to reproduction, a group of about 1000 neurons in mammals release the gonadotropin-releasing hormone (GnRH) peptide from neuroterminals in the median eminence. During the life cycle, there are dramatic changes in reproductive demands, and we focus this review on how GnRH terminals in the median eminence change during reproductive senescence. We discuss morphological and functional properties of the median eminence, and how relationships among GnRH terminals and their microenvironment of nerve terminals, glial cells, and the portal capillary vasculature determine the ability of GnRH peptide to be secreted and to reach its target in the anterior pituitary gland.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, USA
| | | |
Collapse
|
46
|
Prevot V, Bellefontaine N, Baroncini M, Sharif A, Hanchate NK, Parkash J, Campagne C, de Seranno S. Gonadotrophin-releasing hormone nerve terminals, tanycytes and neurohaemal junction remodelling in the adult median eminence: functional consequences for reproduction and dynamic role of vascular endothelial cells. J Neuroendocrinol 2010; 22:639-49. [PMID: 20492366 PMCID: PMC3168864 DOI: 10.1111/j.1365-2826.2010.02033.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although coordinated actions of several areas within the hypothalamus are involved in the secretion of gonadotrophin-releasing hormone (GnRH), the median eminence of the hypothalamus, where the nerve terminals are located, plays a particularly critical role in the release of GnRH. In adult females, prior to the preovulatory surge of GnRH, the retraction of specialised ependymoglial cells lining the floor of the third ventricle named tanycytes allows for the juxtaposition of GnRH nerve terminals with the adjacent pericapillary space of the pituitary portal vasculature, thus forming direct neurohaemal junctions. These morphological changes occur within a few hours and are reversible. Such remodelling may promote physiological conditions to enhance the central release of GnRH and potentiate oestrogen-activated GnRH release. This plasticity involves dynamic cell interactions that bring into play tanycytes, astrocytes, vascular endothelial cells and GnRH neurones themselves. The underlying signalling pathways responsible for these structural changes are comprised of highly diffusible gaseous molecules, such as endothelial nitric oxide, and paracrine communication processes involving receptors of the erbB tyrosine kinase family, transforming growth factor beta 1 and eicosanoids, such as prostaglandin E(2). Some of these molecules, as a result of their ability to diffuse within the median eminence, may also serve as synchronizing cues allowing for the occurrence of functionally meaningful episodes of GnRH secretion by coordinating GnRH release from the GnRH neuroendocrine terminals.
Collapse
Affiliation(s)
- V Prevot
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Prevot V, Hanchate NK, Bellefontaine N, Sharif A, Parkash J, Estrella C, Allet C, de Seranno S, Campagne C, de Tassigny XD, Baroncini M. Function-related structural plasticity of the GnRH system: a role for neuronal-glial-endothelial interactions. Front Neuroendocrinol 2010; 31:241-58. [PMID: 20546773 DOI: 10.1016/j.yfrne.2010.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 05/17/2010] [Accepted: 05/18/2010] [Indexed: 12/18/2022]
Abstract
As the final common pathway for the central control of gonadotropin secretion, GnRH neurons are subjected to numerous regulatory homeostatic and external factors to achieve levels of fertility appropriate to the organism. The GnRH system thus provides an excellent model in which to investigate the complex relationships between neurosecretion, morphological plasticity and the expression of a physiological function. Throughout the reproductive cycle beginning from postnatal sexual development and the onset of puberty to reproductive senescence, and even within the ovarian cycle itself, all levels of the GnRH system undergo morphological plasticity. This structural plasticity within the GnRH system appears crucial to the timely control of reproductive competence within the individual, and as such must have coordinated actions of multiple signals secreted from glial cells, endothelial cells, and GnRH neurons. Thus, the GnRH system must be viewed as a complete neuro-glial-vascular unit that works in concert to maintain the reproductive axis.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, F-59000 Lille, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Differential distribution of erbB receptors in human glioblastoma multiforme: expression of erbB3 in CD133-positive putative cancer stem cells. J Neuropathol Exp Neurol 2010; 69:606-22. [PMID: 20467331 DOI: 10.1097/nen.0b013e3181e00579] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastomas are the most common primary central nervous system tumors in adults, and they remain resistant to current treatments. erbB1 signaling is frequently altered in glioblastomas, suggesting thaterbB receptor family members may represent targets for molecular therapy. We performed a comprehensive analysis of erbB receptor and ligand expression profiles in a panel of 9 glioblastomas andcompared them to nonneoplastic cerebral tissue containing neocortex and adjacent white matter. Quantitative reverse transcription-polymerase chain reaction and Western blot analysis showed that erbB1signaling and erbB2 receptors exhibited highly variable deregulation profiles in the tumors, with patterns ranging from underexpression to overexpression; in contrast, erbB3 and erbB4 were downregulated. We next performed immunohistochemistry to determinethe distribution patterns of erbB receptors among the main neuralcell types in the tumors with special reference to the putative tumor stem cell population. Results revealed intertumoral and intratumoral heterogeneity in all 4 erbB expression profiles, but each receptor exhibited a distinct distribution pattern among glial fibrillary acidic protein-, Olig2-, NeuN-, and CD133-positive populations. Although erbB1 immunoreactivity was detected in only small subsets of CD133-positive putative tumor stem cells, erbB3 immunoreactivity was prominent in this population, suggesting that erbB3 may represent a new potential therapeutic target.
Collapse
|
49
|
Sharif A, Prevot V. ErbB receptor signaling in astrocytes: a mediator of neuron-glia communication in the mature central nervous system. Neurochem Int 2010; 57:344-58. [PMID: 20685225 DOI: 10.1016/j.neuint.2010.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/29/2010] [Accepted: 05/18/2010] [Indexed: 10/19/2022]
Abstract
Astrocytes are now recognized as active players in the developing and mature central nervous system. Each astrocyte contacts vascular structures and thousands of synapses within discrete territories. These cells receive a myriad of inputs and generate appropriate responses to regulate the function of brain microdomains. Emerging evidence has implicated receptors of the ErbB tyrosine kinase family in the integration and processing of neuronal inputs by astrocytes: ErbB receptors can be activated by a wide range of neuronal stimuli; they control critical steps of glutamate-glutamine metabolism; and they regulate the biosynthesis and release of various glial-derived neurotrophic factors, gliomediators and gliotransmitters. These key properties of astrocytic ErbB signaling in neuron-glia interactions have significance for the physiology of the mature central nervous system, as exemplified by the central control of reproduction within the hypothalamus, and are also likely to contribute to pathological situations, since both dysregulation of ErbB signaling and glial dysfunction occur in many neurological disorders.
Collapse
Affiliation(s)
- Ariane Sharif
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the postnatal Brain, Lille, France.
| | | |
Collapse
|
50
|
Sidibe A, Mullier A, Chen P, Baroncini M, Boutin JA, Delagrange P, Prevot V, Jockers R. Expression of the orphan GPR50 protein in rodent and human dorsomedial hypothalamus, tanycytes and median eminence. J Pineal Res 2010; 48:263-269. [PMID: 20210849 DOI: 10.1111/j.1600-079x.2010.00750.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The melatonin receptor family is composed of three members, MT(1) and MT(2) receptors that bind melatonin with high affinity and the orphan GPR50 that does not bind melatonin but shares significant sequence homology with the two other subtypes. In the absence of any known ligand for this orphan receptor, little is still known about its function. We recently reported the development of the first anti-GPR50 antibodies that reliably recognized the recombinant human GPR50. We here used these antibodies to study the expression of GPR50 in mouse, rat and human hypothalamus, a region reported to express GPR50 mRNA. GPR50 immunoreactivity (ir) was observed in dorsomedial hypothalamic (DMH) cells co-stained with the neuronal marker HuC/D. GPR50-ir was also observed in cells of the ependymal layer of the third ventricle that co-stained with vimentin. More specifically, its localization in the lower region of the third ventricle and along the long basal processes contacting portal blood vessels in the median eminence (ME) suggested expression of GPR50 in tanycytes. Consistent staining patterns were observed in all three species with two different antibodies. Taken together, our study validates two GPR50-specific antibodies for the use in rodent and human tissue. Evolutionary conserved expression of GPR50 in DMH neurons and tanycytes, together with previously reported expression of the receptor in the pituitary, support the potentially important role of GPR50 in key hypothalamic functions, including regulation of the hypothalamo-pituitary axes.
Collapse
Affiliation(s)
- Anissa Sidibe
- Institut Cochin, Université Paris Descartes, CNRS, Paris, France
- Inserm, U1016, Paris, France
| | | | - Patty Chen
- Institut Cochin, Université Paris Descartes, CNRS, Paris, France
- Inserm, U1016, Paris, France
| | - Marc Baroncini
- Inserm, Jean-Pierre Aubert Research Center, Lille, France
- Laboratoire d'Anatomie, Faculté de médecine, Université Lille 2, Lille, France
| | | | | | - Vincent Prevot
- Inserm, Jean-Pierre Aubert Research Center, Lille, France
| | - Ralf Jockers
- Institut Cochin, Université Paris Descartes, CNRS, Paris, France
- Inserm, U1016, Paris, France
| |
Collapse
|