1
|
Farkas I, Skrapits K, Sárvári M, Göcz B, Takács S, Rumpler É, Hrabovszky E. Functional GnRH receptor signaling regulates striatal cholinergic neurons in neonatal but not in adult mice. Front Endocrinol (Lausanne) 2024; 15:1353151. [PMID: 38348415 PMCID: PMC10859511 DOI: 10.3389/fendo.2024.1353151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
Reproduction in mammals is controlled by hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Recent studies from our laboratory established that the basal ganglia of the human brain contain additional large populations of GnRH synthesizing neurons which are absent in adult mice. Such extrahypothalamic GnRH neurons mostly occur in the putamen where they correspond to subsets of the striatal cholinergic interneurons (ChINs) and express GnRHR autoreceptors. In an effort to establish a mouse model for functional studies of striatal GnRH/GnRHR signaling, we carried out electrophysiological experiments on acute brain slices from male transgenic mice. Using PN4-7 neonatal mice, half of striatal ChINs responded with transient hyperpolarization and decreased firing rate to 1.2 µM GnRH, whereas medium spiny projection neurons remained unaffected. GnRH acted on its specific receptor because no response was observed in the presence of the GnRHR antagonist Antide. Addition of the membrane-impermeable G protein-coupled receptor inhibitor GDP-β-S to the internal electrode solution eliminated the effect of GnRH. Further, GnRH was able to inhibit ChINs in presence of tetrodotoxin which blocked action potential mediated events. Collectively, these data indicated that the receptor underlying the effects of GnRH in neonatal mice is localized within ChINs. GnRH responsiveness of ChINs was transient and entirely disappeared in adult mice. These results raise the possibility to use neonatal transgenic mice as a functional model to investigate the role of GnRH/GnRHR signaling discovered earlier in adult human ChINs.
Collapse
Affiliation(s)
- Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine (Hungarian Research Network), Budapest, Hungary
| | | | | | | | | | | | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine (Hungarian Research Network), Budapest, Hungary
| |
Collapse
|
2
|
von Schalburg KR, Gowen BE, Christensen KA, Ignatz EH, Hall JR, Rise ML. The late-evolving salmon and trout join the GnRH1 club. Histochem Cell Biol 2023; 160:517-539. [PMID: 37566258 DOI: 10.1007/s00418-023-02227-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/12/2023]
Abstract
Although it is known that the whitefish, an ancient salmonid, expresses three distinct gonadotropin-releasing hormone (GnRH) forms in the brain, it has been thought that the later-evolving salmonids (salmon and trout) had only two types of GnRH: GnRH2 and GnRH3. We now provide evidence for the expression of GnRH1 in the gonads of Atlantic salmon by rapid amplification of cDNA ends, real-time quantitative PCR and immunohistochemistry. We examined six different salmonid genomes and found that each assembly has one gene that likely encodes a viable GnRH1 prepropeptide. In contrast to both functional GnRH2 and GnRH3 paralogs, the GnRH1 homeolog can no longer express the hormone. Furthermore, the viable salmonid GnRH1 mRNA is composed of only three exons, rather than the four exons that build the GnRH2 and GnRH3 mRNAs. Transcribed gnrh1 is broadly expressed (in 17/18 tissues examined), with relative abundance highest in the ovaries. Expression of the gnrh2 and gnrh3 mRNAs is more restricted, primarily to the brain, and not in the gonads. The GnRH1 proximal promoter presents composite binding elements that predict interactions with complexes that contain diverse cell fate and differentiation transcription factors. We provide immunological evidence for GnRH1 peptide in the nucleus of 1-year-old type A spermatogonia and cortical alveoli oocytes. GnRH1 peptide was not detected during other germ cell or reproductive stages. GnRH1 activity in the salmonid gonad may occur only during early stages of development and play a key role in a regulatory network that controls mitotic and/or meiotic processes within the germ cell.
Collapse
Affiliation(s)
- Kristian R von Schalburg
- Department of Biology, Electron Microscopy Laboratory, University of Victoria, Victoria, BC, V8W 3N5, Canada.
| | - Brent E Gowen
- Department of Biology, Electron Microscopy Laboratory, University of Victoria, Victoria, BC, V8W 3N5, Canada
| | - Kris A Christensen
- Department of Biology, University of Victoria, Victoria, BC, V8W 3N5, Canada
| | - Eric H Ignatz
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, A1C 5S7, Canada
| | - Jennifer R Hall
- Aquatic Research Cluster, CREAIT Network, Ocean Sciences Centre, Memorial University of Newfoundland, St. John's, NL, A1C 5S7, Canada
| | - Matthew L Rise
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, A1C 5S7, Canada
| |
Collapse
|
3
|
Chung WCJ, Tsai PS. The initiation and maintenance of gonadotropin-releasing hormone neuron identity in congenital hypogonadotropic hypogonadism. Front Endocrinol (Lausanne) 2023; 14:1166132. [PMID: 37181038 PMCID: PMC10173152 DOI: 10.3389/fendo.2023.1166132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023] Open
Abstract
Neurons that secrete gonadotropin-releasing hormone (GnRH) drive vertebrate reproduction. Genetic lesions that disrupt these neurons in humans lead to congenital hypogonadotropic hypogonadism (CHH) and reproductive failure. Studies on CHH have largely focused on the disruption of prenatal GnRH neuronal migration and postnatal GnRH secretory activity. However, recent evidence suggests a need to also focus on how GnRH neurons initiate and maintain their identity during prenatal and postnatal periods. This review will provide a brief overview of what is known about these processes and several gaps in our knowledge, with an emphasis on how disruption of GnRH neuronal identity can lead to CHH phenotypes.
Collapse
Affiliation(s)
- Wilson CJ Chung
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Pei-San Tsai
- Department of Integrative Physiology, University of Colorado, Boulder, CO, United States
| |
Collapse
|
4
|
Miyakawa M, Murakmai S, Uchiyama Y. Wide-ranging migration and destination of early olfactory placode-derived neurons in chick embryos. Anat Rec (Hoboken) 2023; 306:298-310. [PMID: 36104941 PMCID: PMC10087695 DOI: 10.1002/ar.25080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/15/2022] [Accepted: 09/11/2022] [Indexed: 01/27/2023]
Abstract
Cell migration from the olfactory placode (OP) is a well-known phenomenon wherein various cell types, such as gonadotropin-releasing hormone (GnRH)-producing neurons, migrate toward the telencephalon (TEL) during early embryonic development. However, the spatial relationship between early migratory cells and the forebrain is unclear. We examined the early development of whole-mount chick embryos to observe the three-dimensional spatial relationship among OP-derived migratory neurons, olfactory nerve (ON), and TEL. Migratory neurons that express highly polysialylated neural cell adhesion molecule (PSA-NCAM) emerge from the OP and spread over a relatively wide TEL area at the Hamburger and Hamilton (HH) stage 17. Most migratory neurons form a cellular cord between the olfactory pit and rostral TEL within HH18-20. The earliest axons from the olfactory epithelium (OE) were detected along this neuronal cord using DiI-labeling at HH21. Furthermore, a few PSA-NCAM-positive neurons were dispersed around the cellular cord and over the lateral TEL at HH18. A long cellular cord branch extending to the lateral TEL was transiently observed within HH18-24. These results suggest a novel migratory route of OP-derived neurons during the early developmental stages. Following GFP vector introduction into the OP of HH13-15 embryos, labeled neurons were detected around and within the lateral TEL at HH23 and HH27. At HH36, labeled cells were observed in the rostral-lateral TEL, including the olfactory bulb (OB) region. GFP-labeled and calretinin-positive neurons were detected in the OB, suggesting that early OP-derived neurons enter the forebrain and function as interneurons in the OB.
Collapse
Affiliation(s)
- Momoko Miyakawa
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Cell Biology and Neuroscience, Juntendo University School of Medicine, Tokyo, Japan
| | - Shizuko Murakmai
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Duittoz AH, Tillet Y, Geller S. The great migration: how glial cells could regulate GnRH neuron development and shape adult reproductive life. J Chem Neuroanat 2022; 125:102149. [PMID: 36058434 DOI: 10.1016/j.jchemneu.2022.102149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 10/31/2022]
Abstract
In mammals, reproductive function is under the control of hypothalamic neurons named Gonadotropin-Releasing Hormone (GnRH) neurons. These neurons migrate from the olfactory placode to the brain, during embryonic development. For the past 40 years, these neurons have been considered an example of tangential migration, i.e., dependent on the olfactory/vomeronasal/terminal nerves. Numerous studies have highlighted the factors involved in the migration of these neurons but thus far overlooked the cellular microenvironment that produces them. Many of these factors are dysregulated in hypogonadotropic hypogonadism, resulting in subfertility/infertility. Nevertheless, over the past ten years, several papers have reported the influence of glial cells (named olfactory ensheathing cells [OECs]) in the migration and differentiation of GnRH neurons. This review will describe the atypical origins, migration, and differentiation of these neurons, focusing on the latest discoveries. There will be a more specific discussion on the involvement of OECs in the development of GnRH neurons, during embryonic and perinatal life; as well as on their potential implication in the development of congenital or idiopathic hypogonadotropic hypogonadism (such as Kallmann syndrome).
Collapse
Affiliation(s)
- Anne H Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Yves Tillet
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Sarah Geller
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
6
|
Campbell RE, Coolen LM, Hoffman GE, Hrabovszky E. Highlights of neuroanatomical discoveries of the mammalian gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13115. [PMID: 35502534 PMCID: PMC9232911 DOI: 10.1111/jne.13115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.
Collapse
Affiliation(s)
- Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Lique M. Coolen
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | | | - Erik Hrabovszky
- Laboratory of Reproductive NeurobiologyInstitute of Experimental MedicineBudapestHungary
| |
Collapse
|
7
|
Terasawa E. The mechanism underlying the pubertal increase in pulsatile GnRH release in primates. J Neuroendocrinol 2022; 34:e13119. [PMID: 35491543 PMCID: PMC9232993 DOI: 10.1111/jne.13119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/17/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
In primates, the gonatotropin-releasing hormone (GnRH) neurosecretory system, consisting of GnRH, kisspeptin, and neurokinin B neurons, is active during the neonatal/early infantile period. During the late infantile period, however, activity of the GnRH neurosecretory system becomes minimal as a result of gonadal steroid independent central inhibition, and this suppressed GnRH neurosecretory state continues throughout the prepubertal period. At the initiation of puberty, the GnRH neurosecretory system becomes active again because of the decrease in central inhibition. During the progress of puberty, kisspeptin and neurokinin B signaling to GnRH neurons further increases, resulting in the release of gonadotropins and subsequent gonadal maturation, and hence puberty. This review further discusses potential substrates of central inhibition and subsequent pubertal modification of the GnRH neurosecretory system by the pubertal increase in steroid hormones, which ensures the regulation of adult reproductive function.
Collapse
Affiliation(s)
- Ei Terasawa
- Department of Pediatrics and Wisconsin National Primate Research CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| |
Collapse
|
8
|
Duittoz AH, Forni PE, Giacobini P, Golan M, Mollard P, Negrón AL, Radovick S, Wray S. Development of the gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13087. [PMID: 35067985 PMCID: PMC9286803 DOI: 10.1111/jne.13087] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/02/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022]
Abstract
This review summarizes the current understanding of the development of the neuroendocrine gonadotropin-releasing hormone (GnRH) system, including discussion on open questions regarding (1) transcriptional regulation of the Gnrh1 gene; (2) prenatal development of the GnRH1 system in rodents and humans; and (3) paracrine and synaptic communication during migration of the GnRH cells.
Collapse
Affiliation(s)
| | - Paolo E. Forni
- Department of Biological SciencesUniversity at AlbanyAlbanyNYUSA
- The RNA InstituteUniversity at AlbanyAlbanyNYUSA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Postnatal BrainLille Neuroscience & Cognition, UMR‐S1172, Inserm, CHU LilleUniversity of LilleLilleFrance
| | - Matan Golan
- Institute of Animal SciencesAgricultural Research Organization – Volcani CenterRishon LetziyonIsrael
| | - Patrice Mollard
- Institute of Functional GenomicsCNRS, InsermMontpellier UniversityMontpellierFrance
| | - Ariel L. Negrón
- Clinical and Translational ResearchRutgers Robert Wood Johnson Medical SchoolNew BrunswickNJUSA
| | - Sally Radovick
- Clinical and Translational ResearchRutgers Robert Wood Johnson Medical SchoolNew BrunswickNJUSA
| | - Susan Wray
- Cellular and Developmental Neurobiology SectionNational Institute of Neurological Disorders and Stroke/National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
9
|
Chandra K, Banerjee A, Das M. Epigenetic and transcriptional regulation of GnRH gene under altered metabolism and ageing. THE NUCLEUS 2021. [DOI: 10.1007/s13237-021-00374-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
10
|
Skrapits K, Sárvári M, Farkas I, Göcz B, Takács S, Rumpler É, Váczi V, Vastagh C, Rácz G, Matolcsy A, Solymosi N, Póliska S, Tóth B, Erdélyi F, Szabó G, Culler MD, Allet C, Cotellessa L, Prévot V, Giacobini P, Hrabovszky E. The cryptic gonadotropin-releasing hormone neuronal system of human basal ganglia. eLife 2021; 10:67714. [PMID: 34128468 PMCID: PMC8245125 DOI: 10.7554/elife.67714] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 01/20/2023] Open
Abstract
Human reproduction is controlled by ~2000 hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Here, we report the discovery and characterization of additional ~150,000–200,000 GnRH-synthesizing cells in the human basal ganglia and basal forebrain. Nearly all extrahypothalamic GnRH neurons expressed the cholinergic marker enzyme choline acetyltransferase. Similarly, hypothalamic GnRH neurons were also cholinergic both in embryonic and adult human brains. Whole-transcriptome analysis of cholinergic interneurons and medium spiny projection neurons laser-microdissected from the human putamen showed selective expression of GNRH1 and GNRHR1 autoreceptors in the cholinergic cell population and uncovered the detailed transcriptome profile and molecular connectome of these two cell types. Higher-order non-reproductive functions regulated by GnRH under physiological conditions in the human basal ganglia and basal forebrain require clarification. The role and changes of GnRH/GnRHR1 signaling in neurodegenerative disorders affecting cholinergic neurocircuitries, including Parkinson’s and Alzheimer’s diseases, need to be explored.
Collapse
Affiliation(s)
- Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Viktória Váczi
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Vastagh
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Matolcsy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Budapest, Hungary
| | - Ferenc Erdélyi
- Department of Gene Technology and Developmental Biology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Szabó
- Department of Gene Technology and Developmental Biology, Institute of Experimental Medicine, Budapest, Hungary
| | | | - Cecile Allet
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, Lille, France
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
11
|
Abstract
An increase in pulsatile release of gonadotropin releasing hormone (GnRH) initiates puberty in mammalian species. While mutations in KISS1 and TAC3 and their receptors, KISS1R and NK3R, respectively, result in the absence or abnormal timing of puberty, the neurocircuitry and precise role of kisspeptin and neurokinin B (NKB) in regulation of the GnRH neurosecretory system in primate puberty remain elusive. This review discusses how kisspeptin and NKB signaling contributes to the pubertal increase in GnRH release in non-human primates and how remodeling of the NKB and kisspeptin signaling circuitry controlling GnRH neurons takes place during the progress of puberty. Importantly, the pubertal remodeling of kisspeptin and NKB signaling ensures efficient functions of the GnRH neurosecretory system that regulates sex-specific reproduction in primates.
Collapse
Affiliation(s)
- Ei Terasawa
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| | - James P Garcia
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA
| |
Collapse
|
12
|
Linscott ML, Chung WCJ. Epigenomic control of gonadotrophin-releasing hormone neurone development and hypogonadotrophic hypogonadism. J Neuroendocrinol 2020; 32:e12860. [PMID: 32452569 DOI: 10.1111/jne.12860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 11/30/2022]
Abstract
Mammalian reproductive success depends on gonadotrophin-releasing hormone (GnRH) neurones to stimulate gonadotrophin secretion from the anterior pituitary and activate gonadal steroidogenesis and gametogenesis. Genetic screening studies in patients diagnosed with Kallmann syndrome (KS), a congenital form of hypogonadotrophic hypogonadism (CHH), identified several causal mutations, including those in the fibroblast growth factor (FGF) system. This signalling pathway regulates neuroendocrine progenitor cell proliferation, fate specification and cell survival. Indeed, the GnRH neurone system was absent or abrogated in transgenic mice with reduced (ie, hypomorphic) Fgf8 and/or Fgf receptor (Fgfr) 1 expression, respectively. Moreover, we found that GnRH neurones were absent in the embryonic olfactory placode of Fgf8 hypomorphic mice, the putative birthplace of GnRH neurones. These observations, together with those made in human KS/CHH patients, indicate that the FGF8/FGFR1 signalling system is a requirement for the ontogenesis of the GnRH neuronal system and function. In this review, we discuss how epigenetic factors control the expression of genes such as Fgf8 that are known to be critical for GnRH neurone ontogenesis, fate specification, and the pathogenesis of KS/CHH.
Collapse
Affiliation(s)
- Megan L Linscott
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Wilson C J Chung
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
13
|
Whitlock KE, Postlethwait J, Ewer J. Neuroendocrinology of reproduction: Is gonadotropin-releasing hormone (GnRH) dispensable? Front Neuroendocrinol 2019; 53:100738. [PMID: 30797802 PMCID: PMC7216701 DOI: 10.1016/j.yfrne.2019.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Gonadotropin releasing hormone (GnRH) is a highly conserved neuroendocrine decapeptide that is essential for the onset of puberty and the maintenance of the reproductive state. First identified in mammals, the GnRH signaling pathway is found in all classes of vertebrates; homologues of GnRH have also been identified in invertebrates. In addition to its role as a hypothalamic releasing hormone, GnRH has multiple functions including modulating neural activity within specific regions of the brain. These various functions are mediated by multiple isoforms, which are expressed at diverse locations within the central nervous system. Here we discuss the GnRH signaling pathways in light of new reports that reveal that some vertebrate genomes lack GnRH1. Not only do other isoforms of GnRH not compensate for this gene loss, but elements upstream of GnRH1, including kisspeptins, appear to also be dispensable. We discuss routes that may compensate for the loss of the GnRH1 pathway.
Collapse
Affiliation(s)
- Kathleen E Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaiso (CINV), Instituto de Neurociencia, Universidad de Valparaiso, Avenida Gran Bretaña 1111, Valparaiso, Chile.
| | - John Postlethwait
- Institute of Neuroscience, 324 Huestis Hall, 1254 University of Oregon, Eugene, OR 97403-1254, USA
| | - John Ewer
- Centro Interdisciplinario de Neurociencia de Valparaiso (CINV), Instituto de Neurociencia, Universidad de Valparaiso, Avenida Gran Bretaña 1111, Valparaiso, Chile
| |
Collapse
|
14
|
Whitlock KE. The loss of scents: do defects in olfactory sensory neuron development underlie human disease? ACTA ACUST UNITED AC 2015; 105:114-25. [PMID: 26111003 DOI: 10.1002/bdrc.21094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/12/2015] [Indexed: 12/20/2022]
Abstract
The olfactory system is a fascinating and beguiling sensory system: olfactory sensory neurons detect odors underlying behaviors essential for mate choice, food selection, and escape from predators, among others. These sensory neurons are unique in that they have dendrites contacting the outside world, yet their first synapse lies in the central nervous system. The information entering the central nervous system is used to create odor memories that play a profound role in recognition of individuals, places, and appropriate foods. Here, the structure of the olfactory epithelium is given as an overview to discuss the origin of the olfactory placode, the plasticity of the olfactory sensory neurons, and finally the origins of the gonadotropin-releasing hormone neuroendocrine cells. For the purposes of this review, the development of the peripheral sensory system will be analyzed, incorporating recently published studies highlighting the potential novelties in development mechanisms. Specifically, an emerging model where the olfactory epithelium and olfactory bulb develop simultaneously from a continuous neurectoderm patterned at the end of gastrulation, and the multiple origins of the gonadotropin-releasing hormone neuroendocrine cells associated with the olfactory sensory system development will be presented. Advances in the understanding of the basic mechanisms underlying olfactory sensory system development allows for a more thorough understanding of the potential causes of human disease.
Collapse
Affiliation(s)
- Kathleen E Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaiso, Valparaiso, Chile
| |
Collapse
|
15
|
Zhu J, Xu XH, Knight GE, He C, Burnstock G, Xiang Z. A subpopulation of gonadotropin-releasing hormone neurons in the adult mouse forebrain is γ-Aminobutyric acidergic. J Neurosci Res 2015; 93:1611-21. [DOI: 10.1002/jnr.23610] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/30/2015] [Accepted: 06/01/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Jiao Zhu
- Department of Neurobiology; Key Laboratory of Molecular Neurobiology; Ministry of Education; Second Military Medical University; Shanghai People's Republic of China
| | - Xiao-hui Xu
- School of Life Science; Shanghai University; Shanghai People's Republic of China
| | - Gillian E. Knight
- Autonomic Neuroscience Centre; University College Medical School; London United Kingdom
| | - Cheng He
- Department of Neurobiology; Key Laboratory of Molecular Neurobiology; Ministry of Education; Second Military Medical University; Shanghai People's Republic of China
| | - Geoffrey Burnstock
- Autonomic Neuroscience Centre; University College Medical School; London United Kingdom
- Department of Pharmacology and Therapeutics; The University of Melbourne; Melbourne Australia
| | - Zhenghua Xiang
- Department of Neurobiology; Key Laboratory of Molecular Neurobiology; Ministry of Education; Second Military Medical University; Shanghai People's Republic of China
| |
Collapse
|
16
|
Pillon D, Cadiou V, Angulo L, Duittoz AH. Maternal exposure to 17-alpha-ethinylestradiol alters embryonic development of GnRH-1 neurons in mouse. Brain Res 2012; 1433:29-37. [DOI: 10.1016/j.brainres.2011.11.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 11/07/2011] [Accepted: 11/11/2011] [Indexed: 11/16/2022]
|
17
|
Fuenzalida LC, Keen KL, Terasawa E. Colocalization of FM1-43, Bassoon, and GnRH-1: GnRH-1 release from cell bodies and their neuroprocesses. Endocrinology 2011; 152:4310-21. [PMID: 21896672 PMCID: PMC3199012 DOI: 10.1210/en.2011-1416] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pulsatile release of GnRH-1 is critical for reproductive function. However, the cellular mechanism of GnRH-1 neurosecretion is still elusive. In this study, we examined the neurosecretory process of GnRH-1 neurons using time-lapse image acquisition followed by immunocytochemistry with confocal microscopy. To monitor exocytotic processes, cultured GnRH-1 neurons derived from monkey embryos were labeled with the lipophilic dye, FM1-43, or its fixable form FM1-43Fx, in the presence or absence of depolarization signals, and changes in vesicles labeled with FM1-43 were analyzed. The results show FM1-43 was taken up into the cell and labeled puncta in the soma and neuroprocesses in the absence of depolarization signals, indicating that GnRH-1 neurons were spontaneously active. Depolarization of GnRH-1 neurons with high K+ or veratridine challenge increased the intensity and size of puncta in both soma and neuroprocesses, and the veratridine-induced changes in puncta were blocked by tetrodotoxin, indicating that changes in the puncta intensity and size reflect neurosecretory activity. Subsequent double immunocytochemistry for GnRH-1 and the synaptic vesicle marker, vesicle-associated membrane protein, demonstrated that the FM1-43Fx-labeled puncta were synaptic vesicles with the GnRH-1 peptide. Additional double immunocytochemistry for GnRH-1 and the marker of the neurosecretory active zone, Bassoon, indicated that the FM1-43Fx-labeled puncta were located at the sites of neurosecretory active zones in GnRH-1 neurons. These results suggest that GnRH-1 neurons have the capacity to release the peptide from the soma and dendrites. Collectively, we hypothesize that soma-dendritic release of the peptide may be a mechanism of synchronized activity among GnRH-1 neurons.
Collapse
Affiliation(s)
- Lidia C Fuenzalida
- Department of Pediatrics and Wisconsin National Primate Research Center, 1223 Capitol Court, Madison, Wisconsin 53715-1299, USA
| | | | | |
Collapse
|
18
|
Teixeira L, Guimiot F, Dodé C, Fallet-Bianco C, Millar RP, Delezoide AL, Hardelin JP. Defective migration of neuroendocrine GnRH cells in human arrhinencephalic conditions. J Clin Invest 2010; 120:3668-72. [PMID: 20940512 DOI: 10.1172/jci43699] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 07/28/2010] [Indexed: 12/16/2022] Open
Abstract
Patients with Kallmann syndrome (KS) have hypogonadotropic hypogonadism caused by a deficiency of gonadotropin-releasing hormone (GnRH) and a defective sense of smell related to olfactory bulb aplasia. Based on the findings in a fetus affected by the X chromosome–linked form of the disease, it has been suggested that hypogonadism in KS results from the failed embryonic migration of neuroendocrine GnRH1 cells from the nasal epithelium to the forebrain. We asked whether this singular observation might extend to other developmental disorders that also include arrhinencephaly. We therefore studied the location of GnRH1 cells in fetuses affected by different arrhinencephalic disorders, specifically X-linked KS, CHARGE syndrome, trisomy 13, and trisomy 18, using immunohistochemistry. Few or no neuroendocrine GnRH1 cells were detected in the preoptic and hypothalamic regions of all arrhinencephalic fetuses, whereas large numbers of these cells were present in control fetuses. In all arrhinencephalic fetuses, many GnRH1 cells were present in the frontonasal region, the first part of their migratory path, as were interrupted olfactory nerve fibers that formed bilateral neuromas. Our findings define a pathological sequence whereby a lack of migration of neuroendocrine GnRH cells stems from the primary embryonic failure of peripheral olfactory structures. This can occur either alone, as in isolated KS, or as part of a pleiotropic disease, such as CHARGE syndrome, trisomy 13, and trisomy 18.
Collapse
Affiliation(s)
- Luis Teixeira
- INSERM U1016, Département de Génétique et Développement, Institut Cochin, Université Paris-Descartes, Paris, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Cellular and molecular mechanisms underlying pulsatile GnRH release are not well understood. In the present study, we examined the developmental changes in intracellular calcium dynamics, peptide release, gene expression, and DNA methylation in cultured GnRH neurons derived from the nasal placode of rhesus monkeys. We found that GnRH neurons were functionally immature, exhibiting little fluctuation in intracellular calcium ([Ca(2+)](i)) and sparse pulses of GnRH peptide release in the first 12 d in vitro (div). By 14-18 div, GnRH neurons exhibited periodic [Ca(2+)](i) oscillations, synchronizing at approximately 60-min intervals and GnRH pulses occurred at approximately 60-min intervals. Interestingly, the total GnRH peptide release further increased after 18 div. Measurement of GnRH mRNA and gene CpG methylation status at 0, 14, and 20 div indicated that mRNA levels significantly (P < 0.05) increased between 14 and 20 div, just as maximal decapeptide release was observed. By bisulfite sequencing across a 5' CpG island of the GnRH gene, we further found that methylation at eight of 14 CpG sites significantly (P < 0.05) decreased between 0 and 20 div. These data indicate that epigenetic differentiation occurs during GnRH neuronal development and suggest that increased GnRH gene expression and decreased CpG methylation status are molecular phenotypes of mature GnRH neurons. To our knowledge, this is the first report that developmental DNA demethylation occurs in postmitotic neurons toward a stable neuronal phenotype.
Collapse
Affiliation(s)
- Joseph R Kurian
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin 53715, USA
| | | | | |
Collapse
|
20
|
Booth KK, Webb EC. Effect of Blockage of the Ducts of the Vomeronasal Organ on LH Plasma Levels during the "Whitten Effect" in Does. Vet Med Int 2010; 2010. [PMID: 20871868 PMCID: PMC2943108 DOI: 10.4061/2011/305468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 07/13/2010] [Indexed: 11/25/2022] Open
Abstract
Eighteen mature, nonpregnant, and indigenous South African does were randomly divided into two groups to test if their vomeronasal organs exert an influence on LH plasma levels during a Whitten effect experimental trial. Does in the treatment (VNO ablated) group had their vomeronasal organs rendered nonfunctional by cauterization of the nasoincisive duct under surgical anesthesia. Does in the control group had their nasal civities irrigated with physiological saline under surgical anesthesia. All does were synchronized into oestrus and introduced to bucks one day prior to their expected second oestrus cycle. Successful matings were recorded. Timely blood samples were collected during each of the five days before and five days after buck introduction. Blood plasma concentrations of estradiol and LH were determined by radioimmunoassay. Analysis of variance between groups demonstrated that the does in the VNO ablated group did not demonstrate any interest in mating, did not become pregnant, and did not demonstrate the primary increase in tonic plasma levels of LH that is necessary for ovulation to occur. By contrast, all of the does in the control group demonstrated successful matings, became pregnant, and demonstrated typical primary tonic level increases and preovulation surges in LH. Thus, it was concluded that the vomeronasal organ modulates the primary increase in tonic levels of LH and thus influences ovulation that occurs during the Whitten effect in South African indigenous does.
Collapse
Affiliation(s)
- Kenneth Kurt Booth
- Department of Anatomy and Physiology, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa
| | | |
Collapse
|
21
|
Kim W, Jessen HM, Auger AP, Terasawa E. Postmenopausal increase in KiSS-1, GPR54, and luteinizing hormone releasing hormone (LHRH-1) mRNA in the basal hypothalamus of female rhesus monkeys. Peptides 2009; 30:103-10. [PMID: 18619506 PMCID: PMC2612733 DOI: 10.1016/j.peptides.2008.06.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 06/10/2008] [Accepted: 06/12/2008] [Indexed: 10/21/2022]
Abstract
The G-protein coupled receptor, GPR54, and its ligand, kisspeptin-54 (a KiSS-1 derived peptide) have been reported to be important players in control of LHRH-1 release. However, the role of the GPR54 signaling in primate reproductive senescence is still unclear. In the present study we investigated whether KiSS-1, GPR54, and LHRH-1 mRNA in the brain change after menopause in female rhesus monkeys using quantitative real-time PCR. Results indicate that KiSS-1, GPR54, and LHRH-1 mRNA levels in the medial basal hypothalamus (MBH) in postmenopausal females (28.3+/-1.1 years of age, n=5) were all significantly higher than that in eugonadal adult females (14.7+/-2.1 years of age, n=9), whereas KiSS-1, GPR54, and LHRH-1 mRNA levels in the preoptic area (POA) did not have any significant changes between the two age groups. To further determine the potential contribution by the absence of ovarian steroids, we compared the changes in KiSS-1, GPR54, and LHRH-1 mRNA levels in young adult ovarian intact vs. young ovariectomized females. Results indicate that KiSS-1 and LHRH-1 mRNA levels in the MBH, not POA, in ovariectomized females were significantly higher than those in ovarian intact females, whereas GPR54 mRNA levels in ovariectomized females had a tendency to be elevated in the MBH, although the values were not quite statistically significant. Collectively, in the primate the reduction in the negative feedback control by ovarian steroids appears to be responsible for the aging changes in kisspeptin-GPR54 signaling and the elevated state of the LHRH-1 neuronal system.
Collapse
Affiliation(s)
- Wooram Kim
- Wisconsin National Primate Research Center, University of Wisconsin, 1223 Capitol Court, Madison, WI 53715-1299, USA
| | | | | | | |
Collapse
|
22
|
Schneider JS, Rissman EF. Gonadotropin-releasing hormone II: a multi-purpose neuropeptide. Integr Comp Biol 2008; 48:588-95. [PMID: 21669818 DOI: 10.1093/icb/icn018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Close to 30 forms of gonadotropin releasing hormone (GnRH) and at least five GnRH receptors have been identified in a wide variety of vertebrates and some invertebrates. One form, now called GnRH II, has the broadest distribution and the most ancient and conserved phylogeny. The distribution of the neurons that produce this peptide are completely nonoverlapping with any other GnRH forms. Fibers that project from these neurons overlap with GnRH I cells and/or fibers in a few regions, but are primarily divergent. The musk shrew (Suncus murinus) continues to be the most tractable mammalian species to use for studies of the function of GnRH II. The brain of the musk shrew has two GnRH genes (I and II), two GnRH receptors (types-1 and -2), and two different behaviors can be influenced by central infusion of GnRH II, but not by GnRH I; receptivity and feeding. Here, we summarize research on the musk shrew relative to the behavioral functions of GnRH II. First, female musk shrews are continually sexually receptive by virtue of their lack of an ovarian and/or behavioral estrus cycle. This feature of their reproductive ecology may be related to their semi-tropical distribution and their breeding season is highly dependent on changes in the availability of food. When food is not abundant, females stop mating, but brief bouts of feeding reinstate reproductive behavior. Likewise, intake of food is related to GnRH II mRNA and peptide content in the brain; after mild food restriction both decline. When GnRH II is infused centrally, at times when its content is low, it can both enhance receptivity and inhibit food intake. Simultaneous administration of a type-1 antagonist does not change the effect of GnRH II and use of an analog (135-18) that is a specific GnRH II agonist as well as a type-1 antagonist has the same effect as the endogenous GnRH II peptide. We propose that GnRH II plays a critical role by orchestrating the coordination of reproduction with the availability of nutritional support for these activities. Humans are bombarded with copious nutritional opportunities and at present obesity is a larger threat to health in many parts of the world than is under nutrition. It is our hope that understanding neuropeptides such as GnRH II that regulate food intake can ultimately lead to products that may curb appetite and thus decrease obesity and related risks to health.
Collapse
Affiliation(s)
- Johanna S Schneider
- Department of Biochemistry and Molecular Genetics and Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | |
Collapse
|
23
|
Millar RP, Pawson AJ, Morgan K, Rissman EF, Lu ZL. Diversity of actions of GnRHs mediated by ligand-induced selective signaling. Front Neuroendocrinol 2008; 29:17-35. [PMID: 17976709 PMCID: PMC2667102 DOI: 10.1016/j.yfrne.2007.06.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 06/22/2007] [Accepted: 06/26/2007] [Indexed: 12/27/2022]
Abstract
Geoffrey Wingfield Harris' demonstration of hypothalamic hormones regulating pituitary function led to their structural identification and therapeutic utilization in a wide spectrum of diseases. Amongst these, Gonadotropin Releasing Hormone (GnRH) and its analogs are widely employed in modulating gonadotropin and sex steroid secretion to treat infertility, precocious puberty and many hormone-dependent diseases including endometriosis, uterine fibroids and prostatic cancer. While these effects are all mediated via modulation of the pituitary gonadotrope GnRH receptor and the G(q) signaling pathway, it has become increasingly apparent that GnRH regulates many extrapituitary cells in the nervous system and periphery. This review focuses on two such examples, namely GnRH analog effects on reproductive behaviors and GnRH analog effects on the inhibition of cancer cell growth. For both effects the relative activities of a range of GnRH analogs is distinctly different from their effects on the pituitary gonadotrope and different signaling pathways are utilized. As there is only a single functional GnRH receptor type in man we have proposed that the GnRH receptor can assume different conformations which have different selectivity for GnRH analogs and intracellular signaling proteins complexes. This ligand-induced selective-signaling recruits certain pathways while by-passing others and has implications in developing more selective GnRH analogs for highly specific therapeutic intervention.
Collapse
Affiliation(s)
- Robert P Millar
- MRC Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | | | | | | | |
Collapse
|
24
|
Tannenbaum PL, Schultz-Darken NJ, Saltzman W, Terasawa E, Woller MJ, Abbott DH. Gonadotrophin-releasing hormone (GnRH) release in marmosets I: in vivo measurement in ovary-intact and ovariectomised females. J Neuroendocrinol 2007; 19:342-53. [PMID: 17425609 DOI: 10.1111/j.1365-2826.2007.01534.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vivo hypothalamic gonadotrophin-releasing hormone (GnRH) release was characterised for the first time in a New World primate. A nonterminal and repeatable push-pull perfusion (PPP) technique reliably measured GnRH in conscious common marmoset monkeys. Nineteen adult females (n = 8 ovary-intact in the mid-follicular phase; n = 11 ovariectomised) were fitted with long-term cranial pedestals, and a push-pull cannula was temporarily placed in unique locations within the pituitary stalk-median eminence (S-ME) 2 days prior to each PPP session. Marmosets underwent 1-3 PPPs (32 PPPs in total) lasting up to 12 h. Plasma cortisol levels were not elevated in these habituated marmosets during PPP, and PPP did not disrupt ovulatory cyclicity or subsequent fertility in ovary-intact females. GnRH displayed an organised pattern of release, with pulses occurring every 50.0 +/- 2.6 min and lasting 25.4 +/- 1.3 min. GnRH pulse frequency was consistent within individual marmosets across multiple PPPs. GnRH mean concentration, baseline concentration and pulse amplitude varied predictably with anatomical location of the cannula tip within the S-ME. GnRH release increased characteristically in response to a norepinephrine infusion and decreased abruptly during the evening transition to lights off. Ovary-intact (mid-follicular phase) and ovariectomised marmosets did not differ significantly on any parameter of GnRH release. Overall, these results indicate that PPP can be used to reliably assess in vivo GnRH release in marmosets and will be a useful tool for future studies of reproductive neuroendocrinology in this small primate.
Collapse
Affiliation(s)
- P L Tannenbaum
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | | | |
Collapse
|
25
|
Windsor-Engnell BM, Kasuya E, Mizuno M, Keen KL, Terasawa E. An increase in in vivo release of LHRH and precocious puberty by posterior hypothalamic lesions in female rhesus monkeys (Macaca mulatta). Am J Physiol Endocrinol Metab 2007; 292:E1000-9. [PMID: 17148755 PMCID: PMC2203965 DOI: 10.1152/ajpendo.00493.2006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that a decrease in gamma-aminobutyric acid (GABA) tone and a subsequent increase in glutamatergic tone occur in association with the pubertal increase in luteinizing hormone releasing hormone (LHRH) release in primates. To further determine the causal relationship between developmental changes in GABA and glutamate levels and the pubertal increase in LHRH release, we examined monkeys with precocious puberty induced by lesions in the posterior hypothalamus (PH). Six prepubertal female rhesus monkeys (17.4 +/- 0.1 mo of age) received lesions in the PH, three prepubertal females (17.5 +/- 0.1 mo) received sham lesions, and two females received no treatments. LHRH, GABA, and glutamate levels in the stalk-median eminence before and after lesions were assessed over two 6-h periods (0600-1200 and 1800-2400) using push-pull perfusion. Monkeys with PH lesions exhibited external signs of precocious puberty, including significantly earlier menarche in PH lesion animals (18.8 +/- 0.2 mo) than in sham/controls (25.5 +/- 0.9 mo, P<0.001). Moreover, PH lesion animals had elevated LHRH levels and higher evening glutamate levels after lesions, whereas LHRH changes did not occur in sham/controls until later. Changes in GABA release were not discernible, since evening GABA levels already deceased at 18-20 mo of age in both groups and morning levels remained at the prepubertal levels. The age of first ovulation in both groups did not differ. Collectively, PH lesions may not be a good tool to investigate the mechanism of puberty, and, taking into account the recent findings on the role of kisspeptins, the mechanism of the puberty onset in primates is more complex than we initially anticipated.
Collapse
Affiliation(s)
| | - Etsuko Kasuya
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715-1261
| | - Masaharu Mizuno
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715-1261
| | - Kim L. Keen
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715-1261
| | - Ei Terasawa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715-1261
- Department of Pediatrics, University of Wisconsin, Madison, WI 53715-1261
| |
Collapse
|
26
|
Repérant J, Médina M, Ward R, Miceli D, Kenigfest N, Rio J, Vesselkin N. The evolution of the centrifugal visual system of vertebrates. A cladistic analysis and new hypotheses. ACTA ACUST UNITED AC 2007; 53:161-97. [DOI: 10.1016/j.brainresrev.2006.08.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Revised: 08/10/2006] [Accepted: 08/21/2006] [Indexed: 12/23/2022]
|
27
|
Jastrow H, Oelschläger HHA. Terminal nerve in the mouse-eared bat (Myotis myotis): ontogenetic aspects. ACTA ACUST UNITED AC 2006; 288:1201-15. [PMID: 17031808 DOI: 10.1002/ar.a.20390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As in other mammals, ontogenesis of the terminal nerve (TN) in the mouse-eared bat (Myotis myotis) starts shortly after the formation of the olfactory placode, a derivative of the ectoderm. During development of the olfactory pit, proliferating neuroblasts thicken the placodal epithelium and one cell population migrates toward the rostroventral tip of the telencephalon. Here they accumulate in a primordial terminal ganglion, which successively divides into smaller units. Initial fibers of the TN can be distinguished from olfactory fibers in the mid-embryonic period. The main TN fiber bundle (mfb) originates from the anteriormost ganglion in the nasal roof, whereas one or more inconstant smaller fiber bundles (sfb) originate from one or more smaller ganglia in the basal part of the rostral nasal septum. The fibers of the mfb and sfbs join in the posterior quarter of the nasal roof before reaching the central ganglion (M) located in the meninges medial to the olfactory bulb. From the mid-fetal period onward, a thin TN fiber bundle with some intermingled perikarya connects M to the brain by penetrating its wall rostral to the olfactory tubercle. Additional smaller ganglia may occur in this region. The TN and its ganglia persist in postnatal and adult bats but the number of perikarya is reduced here. Moreover, the different potential functions of the TN are discussed briefly.
Collapse
Affiliation(s)
- Holger Jastrow
- Department of Anatomy and Cell Biology, Histology, Johannes Gutenberg University, Mainz, Germany.
| | | |
Collapse
|
28
|
Wu S, Page L, Sherwood NM. A role for GnRH in early brain regionalization and eye development in zebrafish. Mol Cell Endocrinol 2006; 257-258:47-64. [PMID: 16934393 DOI: 10.1016/j.mce.2006.06.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 06/12/2006] [Accepted: 06/23/2006] [Indexed: 11/22/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a highly conserved peptide that is expressed early in brain development in vertebrates. In zebrafish, we detected GnRH mRNA within 2h post fertilization by RT-PCR. To determine if GnRH is involved in development, we used gene knockdown techniques to block translation of gnrh2 or gnrh3 mRNA after which the expression patterns for gene markers were examined at 24h post fertilization with in situ hybridization. First, loss of either GnRH2 or GnRH3 affected regionalization of the brain as shown by a change in expression of fgf8 or pax2.1 genes in the midbrain-hindbrain boundary or diencephalon-midbrain boundary. Second, lack of GnRH2 and/or GnRH3 altered gene markers expressed in the formation of the eye cup (pax2.1, pax6.1, mab21l2 and meis1.1) or eye stalk (fgf8 and pax2.1). Third, knockdown of GnRH2 affected the size and shape of the midbrain and expression of gene markers therein. Results from assays with the TUNEL method and caspase-3 and -9 activity showed the brain and eye changes were unlikely to result from secondary apoptotic cell death before 24h post fertilization. These experiments suggest that GnRH loss-of-function affects early brain and eye formation during development.
Collapse
Affiliation(s)
- Sheng Wu
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | | |
Collapse
|
29
|
Terasawa E. Postnatal remodeling of gonadotropin-releasing hormone I neurons: toward understanding the mechanism of the onset of puberty. Endocrinology 2006; 147:3650-1. [PMID: 16847090 PMCID: PMC1780282 DOI: 10.1210/en.2006-0588] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Ei Terasawa
- Department of Pediatrics and Wisconsin National Primate Research Center, University of Wisconsin-Madison, Wisconsin 53715-1299, USA.
| |
Collapse
|
30
|
Tsai PS. Gonadotropin-releasing hormone in invertebrates: structure, function, and evolution. Gen Comp Endocrinol 2006; 148:48-53. [PMID: 16256989 DOI: 10.1016/j.ygcen.2005.09.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 09/01/2005] [Accepted: 09/12/2005] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is central to the initiation and maintenance of reproduction in vertebrates. GnRH is found in all major groups of Phylum Chordata, including the protochordates. Studies on functional and structural evolution of GnRH have, in the past, focused exclusively on chordates. However, the recent structural elucidation of an octopus GnRH-like molecule and increasing evidence that GnRH-like substances are present in multiple invertebrate phyla suggest GnRH is an ancient peptide that arose prior to the divergence of protostomes and deuterostomes. The extraordinary conservation of GnRH structure and function raises interesting questions regarding the functional role assumed by GnRH over the course of evolution. This review will focus on the current understanding of GnRH structure and function in non-chordate invertebrates. Special emphasis will be placed upon the possible and speculated functions of GnRH in mollusks.
Collapse
Affiliation(s)
- Pei-San Tsai
- Department of Integrative Physiology and the Center for Neuroscience, University of Colorado, Boulder, CO 80309-0354, USA.
| |
Collapse
|
31
|
Whitlock KE, Illing N, Brideau NJ, Smith KM, Twomey S. Development of GnRH cells: Setting the stage for puberty. Mol Cell Endocrinol 2006; 254-255:39-50. [PMID: 16777316 DOI: 10.1016/j.mce.2006.04.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Cells containing gonadotropin-releasing hormone (GnRH) are essential not only for reproduction but also for neuromodulatory functions in the adult animal. A variety of studies have hinted at multiple origins for GnRH-containing cells in the developing embryo. We have shown, using zebrafish as a model system, that GnRH cells originate from precursors lying outside the olfactory placode: the region of the anterior pituitary gives rise to hypothalamic GnRH cells and the cranial neural crest gives rise to the GnRH cells of the terminal nerve and midbrain. Cells of both the forming anterior pituitary and cranial neural crest are closely apposed to the precursors of the olfactory epithelium during early development. Disruption of kallmann gene function results in loss of the hypothalamic but not the terminal nerve GnRH cells during early development. The GnRH proteins are expressed early in development and this expression is mirrored by the onset of GnRH receptor (GnRH-R) expression during early development. Thus the signaling of the GnRH neuronal circuitry is set up early in development laying the foundation for the GnRH network that is activated at puberty leading to reproductive function in the mature animal.
Collapse
Affiliation(s)
- K E Whitlock
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY 14853, United States.
| | | | | | | | | |
Collapse
|
32
|
Guilgur LG, Moncaut NP, Canário AVM, Somoza GM. Evolution of GnRH ligands and receptors in gnathostomata. Comp Biochem Physiol A Mol Integr Physiol 2006; 144:272-83. [PMID: 16716622 DOI: 10.1016/j.cbpa.2006.02.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Revised: 01/19/2006] [Accepted: 02/14/2006] [Indexed: 10/24/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is the final common signaling molecule used by the brain to regulate reproduction in all vertebrates. Until now, a total of 24 GnRH structural variants have been characterized from vertebrate, protochordate and invertebrate nervous tissue. Almost all vertebrates already investigated have at least two GnRH forms coexisting in the central nervous system. Furthermore, it is now well accepted that three GnRH forms are present both in early and late evolved teleostean fishes. The number and taxonomic distribution of the different GnRH variants also raise questions about the phylogenetic relationships between them. Most of the GnRH phylogenetic analyses are in agreement with the widely accepted idea that the GnRH family can be divided into three main groups. However, the examination of the gnathostome GnRH phylogenetic relationships clearly shows the existence of two main paralogous GnRH lineages: the ''midbrain GnRH" group and the "forebrain GnRH" group. The first one, represented by chicken GnRH-II forms, and the second one composed of two paralogous lineages, the salmon GnRH cluster (only represented in teleostean fish species) and the hypophysotropic GnRH cluster, also present in tetrapods. This analysis suggests that the two forebrain clades share a common precursor and reinforces the idea that the salmon GnRH branch has originated from a duplication of the hypophysotropic lineage. GnRH ligands exert their activity through G protein-coupled receptors of the rhodopsin-like family. As with the ligands, multiple GnRHRs are expressed in individual vertebrate species and phylogenetic analyses have revealed that all vertebrate GnRHRs cluster into three main receptor types. However, new data and a new phylogenetic analysis propose a two GnRHR type model, in which different rounds of gene duplications may have occurred in different groups within each lineage.
Collapse
Affiliation(s)
- Leonardo G Guilgur
- Laboratorio de Ictiofisiología y Acuicultura, IIB-INTECH, CONICET-Universidad Nacional de General San Martín, IIB-INTECH, Camino de Circunvalación Laguna Km. 6, CC 164, B7130IWA, Chascomús, Provincia de Buenos Aires, Argentina
| | | | | | | |
Collapse
|
33
|
Whitlock KE, Smith KM, Kim H, Harden MV. A role forfoxd3andsox10in the differentiation of gonadotropin-releasing hormone (GnRH) cells in the zebrafishDanio rerio. Development 2005; 132:5491-502. [PMID: 16291787 DOI: 10.1242/dev.02158] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is found in a wide range of vertebrate tissues, including the nervous system. In general, GnRH has two functions: endocrine, acting as a releasing hormone; and neuromodulatory,affecting neural activity in the peripheral and central nervous system. The best understood population of GnRH cells is that of the hypothalamus, which is essential for reproduction. Less well understood are the populations of GnRH cells found in the terminal nerve and midbrain, which appear to be neuromodulatory in function. The GnRH-containing cells of the midbrain are proposed to arise from the mesencephalic region of the neural tube. Previously, we showed that neuromodulatory GnRH cells of the terminal nerve arise from cranial neural crest. To test the hypothesis that neuromodulatory GnRH cells of the midbrain also arise from neural crest, we used gene knockdown experiments in zebrafish to disrupt neural crest development. We demonstrate that decrement of the function of foxd3 and/or sox10, two genes important for the development and specification of neural crest, resulted in a reduction and/or loss of GnRH cells of the midbrain, as well as a reduction in the number of terminal nerve GnRH cells. Therefore, our data support a neural crest origin for midbrain GnRH cells. Additionally, we demonstrate that knockdown of kallmann gene function resulted in the loss of endocrine GnRH cells of the hypothalamus, but not of neuromodulatory GnRH cells of the midbrain and terminal nerve, thus providing additional evidence for separate pathways controlling the development of neuromodulatory and endocrine GnRH cells.
Collapse
Affiliation(s)
- Kathleen E Whitlock
- Department of Molecular Biology and Genetics, 445 Biotechnology Building, Cornell University, Ithaca, NY 14853, USA.
| | | | | | | |
Collapse
|
34
|
Abbott DH, Barnett DK, Bruns CM, Dumesic DA. Androgen excess fetal programming of female reproduction: a developmental aetiology for polycystic ovary syndrome? Hum Reprod Update 2005; 11:357-74. [PMID: 15941725 DOI: 10.1093/humupd/dmi013] [Citation(s) in RCA: 354] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The aetiology of polycystic ovary syndrome (PCOS) remains unknown. This familial syndrome is prevalent among reproductive-aged women and its inheritance indicates a dominant regulatory gene with incomplete penetrance. However, promising candidate genes have proven unreliable as markers for the PCOS phenotype. This lack of genetic linkage may represent both extreme heterogeneity of PCOS and difficulty in establishing a universally accepted PCOS diagnosis. Nevertheless, hyperandrogenism is one of the most consistently expressed PCOS traits. Animal models that mimic fetal androgen excess may thus provide unique insight into the origins of the PCOS syndrome. Many female mammals exposed to androgen excess in utero or during early post-natal life typically show masculinized and defeminized behaviour, ovulatory dysfunction and virilized genitalia, although behavioural and ovulatory dysfunction can coexist without virilized genitalia based upon the timing of androgen excess. One animal model shows particular relevance to PCOS: the prenatally androgenized female rhesus monkey. Females exposed to androgen excess early in gestation exhibit hyperandrogenism, oligomenorrhoea and enlarged, polyfollicular ovaries, in addition to LH hypersecretion, impaired embryo development, insulin resistance accompanying abdominal obesity, impaired insulin response to glucose and hyperlipidaemia. Female monkeys exposed to androgen excess late in gestation mimic these programmed changes, except for LH and insulin secretion defects. In utero androgen excess may thus variably perturb multiple organ system programming and thereby provide a single, fetal origin for a heterogeneous adult syndrome.
Collapse
|
35
|
Abstract
Gonadotropin-releasing hormone (GnRH) is an essential decapeptide, with both endocrine and neuromodulatory functions in vertebrates. GnRH-containing cells of the forebrain were thought to originate in the olfactory placode and migrate to their central nervous system destinations, and those of the midbrain to arise locally from the neural tube. Here, the embryonic origins of GnRH cells are re-examined in light of recent data suggesting that forebrain GnRH cells arise from the anterior pituitary placode and cranial neural crest, from where they migrate to their final destinations. The emerging picture suggests that GnRH cells do not originate from the olfactory placodes, but arise from multiple embryonic origins, and transiently associate with the developing olfactory system as they migrate to ventral forebrain locations.
Collapse
Affiliation(s)
- Kathleen E Whitlock
- Department of Molecular Biology and Genetics, 445/449 Biotechnology Building, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
36
|
Müller F, O'Rahilly R. Olfactory structures in staged human embryos. Cells Tissues Organs 2005; 178:93-116. [PMID: 15604533 DOI: 10.1159/000081720] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2004] [Indexed: 12/24/2022] Open
Abstract
The olfactory region was investigated in 303 serially sectioned human embryos, 23 of which were controlled by precise graphic reconstructions. The following findings in the embryonic period are new for the human. (1) The nasal plates arise at the neurosomatic junction, as do also the otic placodes. (2) Crest comes from the nasal plates later (stage 13) than the maximum production in the neural folds (stage 10). (3) The crest arises and migrates during a much longer time (at least until the end of the embryonic period) than the neural crest of the head, where origin and migration end at stage 12. (4) Olfactory nerve fibres enter the brain at stage 17, the vomeronasal fibres and those of the nervus terminalis at stages 17 and 18. (5) Fibre connections between the olfactory tubercle and the olfactory bulb, as well as those to the amygdaloid nuclei, forebrain septum, and hippocampus, develop during and after stage 17. (6) Mitral cells appear late in the embryonic period. (7) Localized, although incomplete, lamination of the olfactory bulb is detectable at the embryonic/fetal transition. (8) Tangential migratory streams of neurons, from stage 22 to the early fetal period, proceed from the subventricular zone of the olfactory bulb towards the future claustrum; they remain within the insular region but are separated from the cortical plate. (9) In future cebocephaly morphological indications may be visible as early as stage 13. The various findings are integrated by means of staging, and current information for the fetal period is tabulated from the literature.
Collapse
Affiliation(s)
- F Müller
- School of Medicine, University of California, Davis, CA, USA
| | | |
Collapse
|
37
|
Giampietro C, Luzzati F, Gambarotta G, Giacobini P, Boda E, Fasolo A, Perroteau I. Stathmin expression modulates migratory properties of GN-11 neurons in vitro. Endocrinology 2005; 146:1825-34. [PMID: 15625246 DOI: 10.1210/en.2004-0972] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Expression of stathmin, a microtubule-associated cytoplasmic protein, prominently localized in neuroproliferative zones and neuronal migration pathways in brain, was investigated in the GnRH neuroendocrine system in vivo and the function was analyzed using an in vitro approach. Here we present novel data demonstrating that GnRH migrating neurons in nasal regions and basal forebrain areas of mouse embryos express stathmin protein. In addition, this expression pattern is dependent on location, as GnRH neurons reaching the hypothalamus are stathmin negative. Immortalized GN-11 cells, which retain many characteristics of migrating GnRH neurons, strongly express stathmin mRNA and protein. The role of stathmin in GnRH migratory properties was evaluated using GN-11 cell line. We up-regulated [stathmin-transfected clones (STMN)+] and down-regulated (STMN-) the expression of stathmin in GN-11 cells, and we investigated changes in cell morphology and motility in vitro. Cells overexpressing stathmin assume a spindle-shaped morphology and their proliferation, as well as their motility, is higher with respect to parental cells. Furthermore, they do not aggregate and express low levels of cadherins compared with control cells. STMN- GN-11 cells are endowed with multipolar processes, and they show a decreased motility and express high levels of cadherin protein. Our findings suggest that stathmin plays a permissive role in GnRH cell motility, possibly via modulation of cadherins expression.
Collapse
Affiliation(s)
- Costanza Giampietro
- Department of Human and Animal Biology, University of Torino, Via Accademia Albertina 13, 10123 Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Gautron JP, Gras C, Enjalbert A. Molecular polymorphism of native gonadotropin-releasing hormone (GnRH) is restricted to mammalian GnRH and [hydroxyproline9] GnRH in the developing rat brain. Neuroendocrinology 2005; 81:69-86. [PMID: 15809516 DOI: 10.1159/000084896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Accepted: 02/01/2005] [Indexed: 11/19/2022]
Abstract
Although chicken gonadotropin-releasing hormone (GnRH)-II is thought to occur in most animal species, its presence and that of two other variants (lamprey GnRH-III, salmon GnRH) is questionable in rodents. Here we report on the GnRH peptides present in the hypothalamus and the remaining brain of rat of both sexes during development. No immunoreactivity was detected in the elution zone of either native or hydroxylated forms of the above three variants in any of brain extracts chromatographed. The main peptides detected were mammalian GnRH (mGnRH) and m[hydroxyproline9]GnRH (mHypGnRH). In the hypothalamus, these peptides were associated with their free acid and precursor forms. N-terminal fragments from both native decapeptides (GnRH) and mGnRH (GnRH) were observed only in the hypothalamus. C-terminal fragments were detected in both tissues. The relative proportions of mGnRH and mHypGnRH showed no developmental changes in the remaining brain. The hypothalamic proportions of mHypGnRH were high on day 5, and decreased from day 15 onwards. The [Gly11]-precursor to mHypGnRH molar ratio was twofold lower than with the non-hydroxylated peptides. The mGnRH to GnRH molar ratio increased in males but decreased in females during development. No sex-related differences were observed in the native decapeptide to GnRH molar ratio. It was concluded that (1) chicken GnRH-II is not present in all mammals, (2) mGnRH and mHypGnRH are the main GnRH isoforms present in the rat brain, (3) the processing of [Gly11]-precursor into mHypGnRH occurs at a higher rate than that of mGnRH, and (4) the catabolism does not interfere with the developmental changes undergone by the mGnRH and mHypGnRH brain contents.
Collapse
Affiliation(s)
- Jean-Pierre Gautron
- Interactions Cellulaires Neuroendocriniennes (UMR 6544) CNRS, Université de la Méditerranée, IFR Jean Roche, Faculté de Médecine Nord, Marseille, France.
| | | | | |
Collapse
|
39
|
Belsham DD, Lovejoy DA. Gonadotropin‐Releasing Hormone: Gene Evolution, Expression, and Regulation. VITAMINS & HORMONES 2005; 71:59-94. [PMID: 16112265 DOI: 10.1016/s0083-6729(05)71003-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The gonadotropin-releasing hormone (GnRH) gene is a superb example of the diverse regulation that is required to maintain the function of an evolutionarily conserved and fundamental gene. Because reproductive capacity is critical to the survival of the species, physiological homeostasis dictates optimal conditions for reproductive success, and any perturbation from this balance may affect GnRH expression. These disturbances may include alterations in signals dictated by stress, nutritional imbalance, body weight, and neurological problems; therefore, changes in other neuroendocrine systems may directly influence the hypothalamic-pituitary-gonadal axis through direct regulation of GnRH. Thus, to maintain optimal reproductive capacity, the regulation of the GnRH gene is tightly constrained by a number of diverse signaling pathways and neuromodulators. In this review, we summarize what is currently known of GnRH gene structure, the location and function of the two isoforms of the GnRH gene, some of the many hormones and neuromodulators found to affect GnRH expression, and the molecular mechanisms responsible for the regulation of the GnRH gene. We also discuss the latest models used to study the transcriptional regulation of the GnRH gene, from cell models to evolving in vivo technologies. Although we have come a long way in the last two decades toward uncovering the intricacies behind the control of the GnRH neuron, there remain vast distances to cover before direct therapeutic manipulation of the GnRH gene to control reproductive competence is possible.
Collapse
Affiliation(s)
- Denise D Belsham
- Department of Physiology, University of Toronto, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | | |
Collapse
|
40
|
Wong TT, Gothilf Y, Zmora N, Kight KE, Meiri I, Elizur A, Zohar Y. Developmental Expression of Three Forms of Gonadotropin-Releasing Hormone and Ontogeny of the Hypothalamic-Pituitary-Gonadal Axis in Gilthead Seabream (Sparus aurata)1. Biol Reprod 2004; 71:1026-35. [PMID: 15163612 DOI: 10.1095/biolreprod.104.028019] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
To address the complexity of the origin of the GnRH system in perciforms, we investigated the ontogenic expression of three GnRHs in gilthead seabream. Using in situ hybridization, chicken (c) GnRH-II mRNA-expressing cells were detected in the hindbrain at 1.5 days postfertilization (DPF) and in the midbrain at 2 DPF and thereafter; the hindbrain signals became undetectable after 10 DPF. Salmon (s) GnRH mRNA-expressing cells were first seen in the olfactory placode at 3 DPF, started caudal migration at 14 DPF, and reached the preoptic areas at 59 DPF. Seabream (sb) GnRH mRNA-expressing cells were first detected in the terminal nerve ganglion cells (TNgc), ventral part of the ventral telencephalon, nucleus preopticus parvocellularis, and thalamus at 39 DPF, and extended to the nucleus preopticus magnocellularis at 43 DPF, ventrolateral hypothalamus at 51 DPF, and nucleus lateralis tuberis and posterior tuberculum at 59 DPF. Coexpression of sbGnRH and sGnRH transcripts was found in the TNgc. Using real-time fluorescence-based quantitative polymerase chain reaction, transcript levels of cGnRH-II and sGnRH were first detected at 1 and 1.5 DPF, respectively, and increased and remained high thereafter. Transcript levels of sbGnRH remained low after first detection at 1 DPF. Furthermore, these GnRH expression profiles were correlated with the expression profiles of reproduction-related genes in which at least four concomitant increases of GnRH, GnRH receptor, gonadotropin, gonadotropin receptor, and Vasa transcripts were found at 5, 8, 14, and 28 DPF. Our data provide an expanded view of the ontogeny of the GnRH system and reproductive axis in perciforms.
Collapse
Affiliation(s)
- Ten-Tsao Wong
- Center of Marine Biotechnology, University of Maryland Biotechnology Institute, Baltimore, Maryland 21202, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
González-Martínez D, Zmora N, Saligaut D, Zanuy S, Elizur A, Kah O, Muñoz-Cueto JA. New insights in developmental origins of different GnRH (gonadotrophin-releasing hormone) systems in perciform fish: an immunohistochemical study in the European sea bass (Dicentrarchus labrax). J Chem Neuroanat 2004; 28:1-15. [PMID: 15363486 DOI: 10.1016/j.jchemneu.2004.05.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 04/01/2004] [Accepted: 05/07/2004] [Indexed: 11/16/2022]
Abstract
The knowledge of the roles and origins of different gonadotrophin-releasing hormone (GnRH) systems could greatly contribute to improve the understanding of mechanisms involved in the physiological control of early development, puberty and spawning. Thus, in this study, we have analyzed the distribution of the cells expressing salmon GnRH, seabream GnRH and chicken GnRH-II forms in the brain and pituitary of developing sea bass using specific antibodies to their corresponding GnRH-associated peptides. The first prepro-chicken GnRH-II-immunoreactive cells arose in the germinal zone of the third ventricle at 4 days after hatching, increasing their number from days 10 to 30, in which they adopted their adult position. The prepro-chicken GnRH-II-immunoreactive fibers became conspicuous in the first week and from day 26 they reached almost all brain areas, especially the hindbrain, being never detected in the pituitary. First prepro-salmon GnRH-immunoreactive cells were detected in the olfactory placode at day 7 after hatching and reached the olfactory bulbs at day 10. Migrating prepro-salmon GnRH cells arrived at the ventral telencephalon at day 15, and became apparent in the preoptic area from day 45. The prepro-salmon GnRH innervation was more evident in the forebrain and increased notably between 10 and 30 days, at which fibers already extended from the olfactory bulbs to the medulla. A few prepro-salmon GnRH-immunoreactive fibers were observed in the pituitary from day 30. The prepro-seabream GnRH-immunoreactive cells were first detected at day 26 in the rostral olfactory bulbs. On day 30, prepro-seabream GnRH-immunoreactive cells were also present in the ventral telencephalon, reaching the preoptic area and the hypothalamus at 45 and 60 days, respectively. The prepro-seabream GnRH innervation appeared restricted to the ventral forebrain, increasing notably during the sixth week, when fibers also reached the pituitary. A significant prepro-seabream GnRH innervation was not detected in the pituitary until day 60.
Collapse
Affiliation(s)
- David González-Martínez
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, Polígono Río San Pedro, 11510 Puerto Real, Cádiz, Spain
| | | | | | | | | | | | | |
Collapse
|
42
|
Schwarting GA, Raitcheva D, Bless EP, Ackerman SL, Tobet S. Netrin 1-mediated chemoattraction regulates the migratory pathway of LHRH neurons. Eur J Neurosci 2004; 19:11-20. [PMID: 14750959 DOI: 10.1111/j.1460-9568.2004.03094.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Luteinizing hormone-releasing hormone (LHRH) neurons migrate from the vomeronasal organ (VNO) to the forebrain in all mammals studied. In mice, the direction of LHRH neuron migration is dependent upon axons that originate in the VNO, but bypass the olfactory bulb and project caudally into the basal forebrain. Thus, factors that guide this unique subset of vomeronasal axons that comprise the caudal vomeronasal nerve (cVNN) are candidates for regulating the migration of LHRH neurons. We previously showed that deleted in colorectal cancer (DCC) is expressed by neurons that migrate out of the VNO during development [Schwarting et al. (2001) J. Neurosci., 21, 911-919]. We examined LHRH neuron migration in Dcc-/- mice and found that trajectories of the cVNN and positions of LHRH neurons are abnormal. Here we extend these studies to show that cVNN trajectories and LHRH cell migration in netrin 1 (Ntn1) mutant mice are also abnormal. Substantially reduced numbers of LHRH neurons are found in the basal forebrain and many LHRH neurons migrate into the cerebral cortex of Ntn1 knockout mice. In contrast, migration of LHRH cells is normal in Unc5h3rcm mutant mice. These results are consistent with the idea that the chemoattraction of DCC+ vomeronasal axons by a gradient of netrin 1 protein in the ventral forebrain guides the cVNN, which, in turn, determines the direction of LHRH neuron migration in the forebrain. Loss of function through a genetic deletion in either Dcc or Ntn1 results in the migration of many LHRH neurons to inappropriate destinations.
Collapse
|
43
|
Abstract
A few examples of hypothalamic, peptidergic disorders leading to clinical signs and symptoms are presented in this review. Increased activity of corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus (PVN) and decreased activity of the vasopressin neurons in the biological clock and of the thyroxine-releasing hormone (TRH) neurons in the PVN contribute to the signs and symptoms of depression. In men, the central nucleus of the bed nucleus of the stria terminalis (BSTc) is about twice as large and contains twice as many somatostatin neurons as in women. In transsexuals this sex difference is reversed, pointing to a role of this structure in gender. Luteinizing hormone-releasing hormone (LHRH) neurons are formed in the fetal olfactory placade and migrate along the terminal nerve fibers into the hypothalamus. In Kallmann's syndrome the migration process of the LHRH (gonadotropin-releasing hormone) neurons is aborted, which explains the joint occurrence of hypogonadotropic hypogonadism and anosmia in this syndrome. In postmenopausal women, the neurons of the infundibular nucleus hypertrophy and become hyperactive because of the disappearance of the estrogen feedback and contain hyperactive peptidergic neurons. Climacteric flushes may be caused by hyperactivity of the neurokinin-B or LHRH neurons in this nucleus. The hypocretin (orexin) neurons in the perifornical area are involved in sleep. In narcolepsy with cataplexy, a loss of these neurons, probably due to an autoimmune process, is found. Obese subjects with a mutation in the gene that encodes for leptin, the preproghrelin gene, or the alpha-melanocyte-stimulating hormone (alpha-MSH) gene have been described. Decreased numbers and activity of the oxytocin neurons in the PVN may be responsible for the absence of satiety in Prader-Willi syndrome. Moreover, a glucocorticoid receptor polymorphism is associated with obesitas and dysregulation of the hypothalamus-pituitary-adrenal axis. In contrast, two single nucleotide polymorphisms (SNPs) of the AGRP gene have been associated with anorexia nervosa.
Collapse
Affiliation(s)
- Dick F Swaab
- Netherlands Institute for Brain Research, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Pierantoni R, Cobellis G, Meccariello R, Fasano S. Evolutionary aspects of cellular communication in the vertebrate hypothalamo-hypophysio-gonadal axis. INTERNATIONAL REVIEW OF CYTOLOGY 2003; 218:69-141. [PMID: 12199520 DOI: 10.1016/s0074-7696(02)18012-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review emphasizes the comparative approach for developing insight into knowledge related to cellular communications occurring in the hypothalamus-pituitary-gonadal axis. Indeed, research on adaptive phenomena leads to evolutionary tracks. Thus, going through recent results, we suggest that pheromonal communication precedes local communication which, in turn, precedes communication via the blood stream. Furthermore, the use of different routes of communication by a certain mediator leads to a conceptual change related to what hormones are. Nevertheless, endocrine communication should leave out of consideration the source (glandular or not) of mediator. Finally, we point out that the use of lower vertebrate animal models is fundamental to understanding general physiological mechanisms. In fact, different anatomical organization permits access to tissues not readily approachable in mammals.
Collapse
|
45
|
Bruneau G, Izvolskaia M, Ugrumov M, Tillet Y, Duittoz AH. Prolonged neurogenesis during early development of gonadotropin-releasing hormone neurones in sheep (Ovis Aries): in vivo and in vitro studies. Neuroendocrinology 2003; 77:177-86. [PMID: 12673051 DOI: 10.1159/000069505] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2002] [Accepted: 11/19/2002] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons involved in controlling the reproductive function in vertebrates are derived from the olfactory placode. However, in the sheep and the rat species, GnRH-immunoreactive (GnRH-IR) neurons could not be detected in the olfactory region during the earliest phase of GnRH system development. Using in situ hybridization (ISH) and immunohistochemistry (IHC) in sheep embryos ranging from 26 to 53 days' gestational age (G26-G53), the present work confirmed that GnRH expression could not be detected during the earliest steps of migration. The first ISH+ cells were detected in the nasal septum and at the entrance of the telencephalon at G33 stage. [(3)H]-thymidine pulses applied to in vitro olfactory explant cultures showed that GnRH neuron precursor cells have an extended multiplication period corresponding to G26-G36 before entering the neuronal differentiation process. Therefore, the lack of GnRH neuron detection during the early phase of development in the sheep compared to the mouse and other vertebrates represents a major difference in the early development of GnRH neurons. In the mouse, GnRH neuron precursors have a limited multiplication period in the vomeronasal pit and only postmitotic neurons start migration, whereas in the sheep embryo, the multiplication period is extended to about 10 days as demonstrated in olfactory explant cultures.
Collapse
Affiliation(s)
- G Bruneau
- Neuroendocrinologie, UMR 6073 INRA CNRS Université de Tours PRC, INRA, Nouzilly, France
| | | | | | | | | |
Collapse
|
46
|
Terasawa E, Richter TA, Keen KL. A role for non-neuronal cells in synchronization of intracellular calcium oscillations in primate LHRH neurons. PROGRESS IN BRAIN RESEARCH 2003; 141:283-91. [PMID: 12508575 DOI: 10.1016/s0079-6123(02)41099-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Affiliation(s)
- Ei Terasawa
- Wisconsin National Primate Research Center, 1223 Capitol Court, Madison, WI 53715-1299, USA.
| | | | | |
Collapse
|
47
|
Krajewski SJ, Abel TW, Voytko ML, Rance NE. Ovarian steroids differentially modulate the gene expression of gonadotropin-releasing hormone neuronal subtypes in the ovariectomized cynomolgus monkey. J Clin Endocrinol Metab 2003; 88:655-62. [PMID: 12574196 DOI: 10.1210/jc.2002-020887] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
In the present study, we compared the morphology and distribution of neurons expressing GnRH gene transcripts in the hypothalamus and forebrain of the cynomolgus monkey to that of the human. As in the human, three subtypes of GnRH neurons were identified. Type I GnRH neurons were small, oval cells with high levels of gene expression and were located within the basal hypothalamus. Type II GnRH neurons were small and sparsely labeled and were widely scattered in the hypothalamus, midline nuclei of the thalamus, and extended amygdala. Type III neurons displayed magnocellular morphology and intermediate labeling intensity and were located in the nucleus basalis of Meynert, caudate, and amygdala. In a second experiment, we determined the effect of estrogen or estrogen plus progesterone on the gene expression of GnRH neurons in the brains of young, ovariectomized cynomolgus monkeys. We report that hormone treatment resulted in a significant decrease in GnRH mRNA in type I neurons within the basal hypothalamus of ovariectomized monkeys. In contrast, there was no effect of hormone treatment on the gene expression of type III GnRH neurons in the nucleus basalis of Meynert. The present findings provide evidence that the increase in gene expression of type I GnRH neurons in postmenopausal women is secondary to the ovarian failure of menopause. The differential responses of type I and III GnRH neurons to hormone treatment provide additional evidence that distinct subpopulations of neurons expressing GnRH mRNA exist in the primate hypothalamus.
Collapse
Affiliation(s)
- Sally J Krajewski
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona 85724, USA
| | | | | | | |
Collapse
|
48
|
Gray SL, Adams BA, Warby CM, Von Schalburg KR, Sherwood NM. Transcription and translation of the salmon gonadotropin-releasing hormone genes in brain and gonads of sexually maturing rainbow trout (Oncorhynchus mykiss). Biol Reprod 2002; 67:1621-7. [PMID: 12390896 DOI: 10.1095/biolreprod.102.004788] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Rainbow trout sexually mature at the end of Year 3. The form of GnRH that controls gonadotropin release in trout is salmon GnRH (sGnRH). In the tetraploid rainbow trout, two genes encode an identical sGnRH peptide. The sGnRH gene-1 produces one mRNA, whereas sGnRH gene-2 can produce more than one. This study asks whether the transcripts and their protein products are expressed in the brain and gonads and whether the pattern correlates with sexual maturity over the final year leading to first spawning. Brain sGnRH mRNA and protein were continuously present throughout the third year. We show for the first time that the long sGnRH-2 mRNA transcript is expressed in neural tissue and not exclusively in gonadal tissue. Expression of the long sGnRH-2 mRNA in the brain coincides with high levels of sGnRH peptide in the brain during a time of increased gonadal growth. Thus, the long sGnRH-2 mRNA in the brain may act to regulate sGnRH production in a stage-specific rather than a tissue-specific manner. In gonads, local sGnRH is thought to play an autocrine/paracrine role in regulating gonadal maturation and spawning. In the maturing gonads, sGnRH gene-1 and -2 are expressed intermittently. Strikingly, sGnRH peptide was not detected in the gonads at any time during Year 3. These results suggest that either the sGnRH transcripts in the gonads are not translated into protein or, if translated, the protein is rapidly released, resulting in gonadal content below 1 fM per fish.
Collapse
Affiliation(s)
- Sarah L Gray
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada V8W 3N5
| | | | | | | | | |
Collapse
|
49
|
Abstract
Gonadotropin releasing hormone-1 (GnRH-1) neurons, critical for reproduction, are derived from the nasal placode and migrate into the brain during prenatal development. Once within the brain, GnRH-1 cells become integral components of the CNS-pituitary-gonadal axis, essential for reproductive maturation and maintenance of reproductive function in adults. This review focuses on the lineage and development of the GnRH-1 neuroendocrine system. Although the migration of these cells from nose to brain has been well documented in a variety of species, many questions remain concerning the melecules and cues directing GnRH-1 cell differentiation, migration, axon targeting, and establishment and control of GnRH-1 secretion. These process most likely involve multiple and redundant cues because if these mechanisms fail, reproduction dysfunction will ensue and guarantee that this defect does not remain in the gene pool.
Collapse
Affiliation(s)
- S Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 36, Room 5A-21, Bethesda, MD 20892-4156, USA.
| |
Collapse
|
50
|
Montaner AD, Mongiat L, Lux-Lantos VA, Warby C, Chewpoy B, Bianchi MS, Libertun C, Rivier JE, Sherwood NM, Somoza GM. Guinea pig gonadotropin-releasing hormone: expression pattern, characterization and biological activity in rodents. Neuroendocrinology 2002; 75:326-38. [PMID: 12006786 DOI: 10.1159/000057342] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a decapeptide widely known for its role in regulating vertebrate reproduction by serving as a signal from the hypothalamus to pituitary gonadotropes. The first form of GnRH to be identified was isolated from mammals (mGnRH) and the same form has been reported for all mammals studied, which includes marsupials and placental mammals. Later, another variant, chicken GnRH-II (cGnRH-II) was shown to be expressed together with mGnRH in the brains of all jawed vertebrates, including mammals such as rats, monkeys and humans. Our objective was to characterize a third form of GnRH that was isolated previously as mRNA from guinea pigs (gpGnRH), but has not been reported for any other mammal to date. Furthermore, the gonadotropic activity of gpGnRH has not been fully characterized. Our results, using chromatographical and immunological methods, show for the first time that gpGnRH is expressed together with mGnRH in some rodents (wild guinea pig and capybara), but not in others (mouse and hamster). Also, the gonadotropic activity of gpGnRH and mGnRH was tested in two different rat cell culture systems. Although there have been reports that the salmon(s) form of GnRH is present in mammals, we did not detect sGnRH in capybara, wild guinea pigs, hamsters, rats or mice. Taken together with previous reports, the present results support the idea that the expression of multiple GnRH variants in a single species is a common pattern in most vertebrate groups.
Collapse
Affiliation(s)
- Alejandro D Montaner
- Instituto de Investigaciones Biomédicas, Fundación Pablo Cassará, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|