1
|
Kacimi L, Prevot V. GnRH and Cognition. Endocrinology 2025; 166:bqaf033. [PMID: 39996304 DOI: 10.1210/endocr/bqaf033] [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: 12/18/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 02/26/2025]
Abstract
GnRH is traditionally recognized as the central regulator of reproduction through its pulsatile secretion, which governs the hypothalamic-pituitary-gonadal axis. However, recent evidence has highlighted its broader role in brain development and function, including in cognitive and higher intellectual processes. GnRH production follows distinct phases, from its early activation during minipuberty-the first postnatal activation of GnRH neurons during the infantile period-, its reactivation and stabilization starting at puberty, and its eventual decline with age and the loss of gonadal steroid feedback. This evolution depends on the establishment, maturation and activation of GnRH neurons, a complex process regulated by the cellular and molecular environment of these neurons, including multiple neuronal and glial types as well as a minipubertal "switch" in gene expression, the perturbation of which may have long-term or delayed consequences for both reproductive and cognitive function. The cognitive role of GnRH may be related to its recently revealed involvement in maintaining myelination and synaptic plasticity, whereas disruptions in its finely tuned rhythmic secretion, either age-related or pathological, are associated with cognitive decline and neurodegenerative disorders. Restoring physiological GnRH levels and pulsatility can reverse age-related cognitive decline and improve sensory functions even in adulthood, suggesting a mobilization of the "cognitive reserve" in both animal models and human patients. This review highlights recent advances in our understanding of the GnRH system and the therapeutic potential of pulsatile GnRH therapy to mitigate age-related cognitive decline and neurodegenerative processes.
Collapse
Affiliation(s)
- Loïc Kacimi
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, FHU 1000 days for health, EGID, DistALZ, UMR_S112, Lille 59000, France
| |
Collapse
|
2
|
Chachlaki K, Duc KL, Storme L, Prévot V. Novel insights into minipuberty and GnRH: Implications on neurodevelopment, cognition, and COVID-19 therapeutics. J Neuroendocrinol 2024; 36:e13387. [PMID: 38565500 PMCID: PMC7616535 DOI: 10.1111/jne.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
In humans, the first 1000 days of life are pivotal for brain and organism development. Shortly after birth, gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are activated, a phenomenon known as minipuberty. This phenomenon, observed in all mammals studied, influences the postnatal development of the hypothalamic-pituitary-gonadal (HPG) axis and reproductive function. This review will put into perspective the results of recent studies showing that the impact of minipuberty extends beyond reproductive function, influencing sensory and cognitive maturation. Studies in mice have revealed the role of nitric oxide (NO) in regulating minipuberty amplitude, with NO deficiency linked to cognitive and olfactory deficits. Additionally, findings indicate that cognitive and sensory defects in adulthood in a mouse model of Down syndrome are associated with an age-dependent decline of GnRH production, whose origin can be traced back to minipuberty, and point to the potential therapeutic role of pulsatile GnRH administration in cognitive disorders. Furthermore, this review delves into the repercussions of COVID-19 on GnRH production, emphasizing potential consequences for neurodevelopment and cognitive function in infected individuals. Notably, GnRH neurons appear susceptible to SARS-CoV-2 infection, raising concerns about potential long-term effects on brain development and function. In conclusion, the intricate interplay between GnRH neurons, GnRH release, and the activity of various extrahypothalamic brain circuits reveals an unexpected role for these neuroendocrine neurons in the development and maintenance of sensory and cognitive functions, supplementing their established function in reproduction. Therapeutic interventions targeting the HPG axis, such as inhaled NO therapy in infancy and pulsatile GnRH administration in adults, emerge as promising approaches for addressing neurodevelopmental cognitive disorders and pathological aging.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| | - Kevin Le Duc
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Laurent Storme
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| |
Collapse
|
3
|
Terrinoni A, Micheloni G, Moretti V, Caporali S, Bernardini S, Minieri M, Pieri M, Giaroni C, Acquati F, Costantino L, Ferrara F, Valli R, Porta G. OTX Genes in Adult Tissues. Int J Mol Sci 2023; 24:16962. [PMID: 38069286 PMCID: PMC10707059 DOI: 10.3390/ijms242316962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
OTX homeobox genes have been extensively studied for their role in development, especially in neuroectoderm formation. Recently, their expression has also been reported in adult physiological and pathological tissues, including retina, mammary and pituitary glands, sinonasal mucosa, in several types of cancer, and in response to inflammatory, ischemic, and hypoxic stimuli. Reactivation of OTX genes in adult tissues supports the notion of the evolutionary amplification of functions of genes by varying their temporal expression, with the selection of homeobox genes from the "toolbox" to drive or contribute to different processes at different stages of life. OTX involvement in pathologies points toward these genes as potential diagnostic and/or prognostic markers as well as possible therapeutic targets.
Collapse
Affiliation(s)
- Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giovanni Micheloni
- Genomic Medicine Research Center, Department of Medicine and Surgery, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
| | - Vittoria Moretti
- Genomic Medicine Research Center, Department of Medicine and Surgery, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
| | - Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, Via del Politecnico 1, 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Marilena Minieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Cristina Giaroni
- Department of Medicina e Innovazione Tecnologica, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
| | - Francesco Acquati
- Genomic Medicine Research Center, Department of Medicine and Surgery, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
- Department of Biotechnology and Life Science, University of Insubria, Via JH Dunant 3, 21100 Varese, Italy
| | - Lucy Costantino
- Department of Molecular Genetics, Centro Diagnostico Italiano, Via Saint Bon 20, 20147 Milano, Italy
| | - Fulvio Ferrara
- Department of Molecular Genetics, Centro Diagnostico Italiano, Via Saint Bon 20, 20147 Milano, Italy
| | - Roberto Valli
- Genomic Medicine Research Center, Department of Medicine and Surgery, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
| | - Giovanni Porta
- Genomic Medicine Research Center, Department of Medicine and Surgery, University of Insubria, Via JH Dunant 5, 21100 Varese, Italy
| |
Collapse
|
4
|
Manfredi-Lozano M, Leysen V, Adamo M, Paiva I, Rovera R, Pignat JM, Timzoura FE, Candlish M, Eddarkaoui S, Malone SA, Silva MSB, Trova S, Imbernon M, Decoster L, Cotellessa L, Tena-Sempere M, Claret M, Paoloni-Giacobino A, Plassard D, Paccou E, Vionnet N, Acierno J, Maceski AM, Lutti A, Pfrieger F, Rasika S, Santoni F, Boehm U, Ciofi P, Buée L, Haddjeri N, Boutillier AL, Kuhle J, Messina A, Draganski B, Giacobini P, Pitteloud N, Prevot V. GnRH replacement rescues cognition in Down syndrome. Science 2022; 377:eabq4515. [PMID: 36048943 PMCID: PMC7613827 DOI: 10.1126/science.abq4515] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At the present time, no viable treatment exists for cognitive and olfactory deficits in Down syndrome (DS). We show in a DS model (Ts65Dn mice) that these progressive nonreproductive neurological symptoms closely parallel a postpubertal decrease in hypothalamic as well as extrahypothalamic expression of a master molecule that controls reproduction-gonadotropin-releasing hormone (GnRH)-and appear related to an imbalance in a microRNA-gene network known to regulate GnRH neuron maturation together with altered hippocampal synaptic transmission. Epigenetic, cellular, chemogenetic, and pharmacological interventions that restore physiological GnRH levels abolish olfactory and cognitive defects in Ts65Dn mice, whereas pulsatile GnRH therapy improves cognition and brain connectivity in adult DS patients. GnRH thus plays a crucial role in olfaction and cognition, and pulsatile GnRH therapy holds promise to improve cognitive deficits in DS.
Collapse
Affiliation(s)
- Maria Manfredi-Lozano
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michela Adamo
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Isabel Paiva
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Renaud Rovera
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Jean-Michel Pignat
- Department of Clinical Neurosciences, Neurorehabilitation Unit, University Hospital CHUV, Lausanne, Switzerland
| | - Fatima Ezzahra Timzoura
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Michael Candlish
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Samuel A. Malone
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Mauro S. B. Silva
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Sara Trova
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Monica Imbernon
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Laurine Decoster
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Manuel Tena-Sempere
- Univ. Cordoba, IMIBC/HURS, CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| | - Ariane Paoloni-Giacobino
- Department of Genetic Medicine, University Hospitals of Geneva, 4 rue Gabrielle-Perret-Gentil, 1211, Genève 14, Switzerland
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Emmanuelle Paccou
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Vionnet
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - James Acierno
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Aleksandra Maleska Maceski
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Frank Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 67000 Strasbourg, France
| | - S. Rasika
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Federico Santoni
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421, Homburg, Germany
| | - Philippe Ciofi
- Univ. Bordeaux, Inserm, U1215, Neurocentre Magendie, Bordeaux, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
| | - Nasser Haddjeri
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron 69500, France
| | - Anne-Laurence Boutillier
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg-CNRS, Strasbourg, France
| | - Jens Kuhle
- Neurologic Clinic and Polyclinic, MS Centre and Research Centre for Clinical Neuroimmunology and Neuroscience Basel; University Hospital Basel, University of Basel, Basel Switzerland
| | - Andrea Messina
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, LabexDistAlz, Lille, France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 days for health, EGID, Lille, France
| |
Collapse
|
5
|
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
|
6
|
Pandolfi EC, Tonsfeldt KJ, Hoffmann HM, Mellon PL. Deletion of the Homeodomain Protein Six6 From GnRH Neurons Decreases GnRH Gene Expression, Resulting in Infertility. Endocrinology 2019; 160:2151-2164. [PMID: 31211355 PMCID: PMC6821215 DOI: 10.1210/en.2019-00113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
Hypothalamic GnRH (luteinizing hormone-releasing hormone) neurons are crucial for the hypothalamic-pituitary-gonadal (HPG) axis, which regulates mammalian fertility. Insufficient GnRH disrupts the HPG axis and is often associated with the genetic condition idiopathic hypogonadotropic hypogonadism (IHH). The homeodomain protein sine oculis-related homeobox 6 (Six6) is required for the development of GnRH neurons. Although it is known that Six6 is specifically expressed within a more mature GnRH neuronal cell line and that overexpression of Six6 induces GnRH transcription in these cells, the direct role of Six6 within the GnRH neuron in vivo is unknown. Here we find that global Six6 knockout (KO) embryos show apoptosis of GnRH neurons beginning at embryonic day 14.5 with 90% loss of GnRH neurons by postnatal day 1. We sought to determine whether the hypogonadism and infertility reported in the Six6KO mice are generated via actions within the GnRH neuron in vivo by creating a Six6-flox mouse and crossing it with the LHRHcre mouse. Loss of Six6 specifically within the GnRH neuron abolished GnRH expression in ∼0% of GnRH neurons. We further demonstrated that deletion of Six6 only within the GnRH neuron leads to infertility, hypogonadism, hypogonadotropism, and delayed puberty. We conclude that Six6 plays distinct roles in maintaining fertility in the GnRH neuron vs in the migratory environment of the GnRH neuron by maintaining expression of GnRH and survival of GnRH neurons, respectively. These results increase knowledge of the role of Six6 in the brain and may offer insight into the mechanism of IHH.
Collapse
Affiliation(s)
- Erica C Pandolfi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| | - Hanne M Hoffmann
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
7
|
Li Z, Guo R, Gu Z, Wang X, Wang Y, Xu H, Wang C, Liu X. Identification of a promoter element mediating kisspeptin-induced increases in GnRH gene expression in sheep. Gene 2019; 699:1-7. [PMID: 30853631 DOI: 10.1016/j.gene.2019.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 01/04/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) plays an important role in regulating the activities of other components downstream of the hypothalamic-pituitary-gonadal (HPG) axis and maintaining the normal reproductive cycle of animals. However, the molecular mechanisms by which GnRH synthesis and secretion are regulated in sheep remains unclear. In this study, a series of eight recombinant vectors with deletion fragments were constructed and cotransfected with pGL3-Basic and pRL-SV40 into sheep hypothalamic neuronal cells. After treatment with 1 nM kisspeptin, the core promoter of the sheep GnRH gene was identified to be in the region of -1912 bp to -1461 bp by dual-luciferase reporter assay. Bioinformatics analysis showed that there was a binding site for the transcription factor Otx-2 in the core promoter region (-1786 to -1770 bp) that was highly conserved among different species. The expression patterns of Kiss-1, Otx-2 and GnRH in the sheep hypothalamus were the same, and the expression of Kiss-1, Otx-2 and GnRH was significantly higher in the breeding season than in nonbreeding season (P < 0.01). In addition, when hypothalamic neurons were cultured in vitro with kisspeptin, kisspeptin induced the expression of GnRH and Otx-2. In conclusion, these results provide evidence that the core promoter region (-1786 to -1770 bp) of the GnRH gene is involved in the regulation of hypothalamic activity by kisspeptin and that binding of the transcription factor Otx-2 mediates this activation.
Collapse
Affiliation(s)
- Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Ruoting Guo
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, China; Center for Animal Disease Control and Prevention, Changji 831100, Xinjiang, China
| | - Zhenzhen Gu
- Key Laboratory of Genetics, Breeding & Reproduction of Grass-Feeding Livestock, Ministry of Agriculture, Xinjiang Academy of Animal Science, Urumqi 830026, Xinjiang, China
| | - Xiangnan Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yongcai Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Huifen Xu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Chunxiu Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Xiaojun Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; College of Animal Science and Technology, Shihezi University, Shihezi 832003, Xinjiang, China.
| |
Collapse
|
8
|
Yellapragada V, Liu X, Lund C, Känsäkoski J, Pulli K, Vuoristo S, Lundin K, Tuuri T, Varjosalo M, Raivio T. MKRN3 Interacts With Several Proteins Implicated in Puberty Timing but Does Not Influence GNRH1 Expression. Front Endocrinol (Lausanne) 2019; 10:48. [PMID: 30800097 PMCID: PMC6375840 DOI: 10.3389/fendo.2019.00048] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Paternally-inherited loss-of-function mutations in makorin ring finger protein 3 gene (MKRN3) underlie central precocious puberty. To investigate the puberty-related mechanism(s) of MKRN3 in humans, we generated two distinct bi-allelic MKRN3 knock-out human pluripotent stem cell lines, Del 1 and Del 2, and differentiated them into GNRH1-expressing neurons. Both Del 1 and Del 2 clones could be differentiated into neuronal progenitors and GNRH1-expressing neurons, however, the relative expression of GNRH1 did not differ from wild type cells (P = NS). Subsequently, we investigated stable and dynamic protein-protein interaction (PPI) partners of MKRN3 by stably expressing it in HEK cells followed by mass spectrometry analyses. We found 81 high-confidence novel protein interaction partners, which are implicated in cellular processes such as insulin signaling, RNA metabolism and cell-cell adhesion. Of the identified interactors, 20 have been previously implicated in puberty timing. In conclusion, our stem cell model for generation of GNRH1-expressing neurons did not offer mechanistic insight for the role of MKRN3 in puberty initiation. The PPI data, however, indicate that MKRN3 may regulate puberty by interacting with other puberty-related proteins. Further studies are required to elucidate the possible mechanisms and outcomes of these interactions.
Collapse
Affiliation(s)
- Venkatram Yellapragada
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Xiaonan Liu
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Carina Lund
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Känsäkoski
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sanna Vuoristo
- Department of Obstetrics and Gynecology, Helsinki University Hospital, HUH, Helsinki, Finland
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, Helsinki University Hospital, HUH, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, Helsinki University Hospital, HUH, Helsinki, Finland
| | - Markku Varjosalo
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- New Children's Hospital, Pediatric Research Center, Helsinki University Hospital, HUH, Helsinki, Finland
- *Correspondence: Taneli Raivio
| |
Collapse
|
9
|
Ding J, Wang J, Jin H, Xia T, Cheng Y, Wu J, Han X. Microcystin-LR reduces the synthesis of gonadotropin-releasing hormone by activating multiple signaling pathways resulting in decrease of testosterone in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 643:496-506. [PMID: 29945085 DOI: 10.1016/j.scitotenv.2018.06.123] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/29/2018] [Accepted: 06/10/2018] [Indexed: 06/08/2023]
Abstract
We previously reported Microcystin-LR (MC-LR) could enter the hypothalamus, reduce the expression of gonadotropin-releasing hormone (GnRH), and induce male reproductive barriers. However, the molecular mechanisms underlying in the hypothalamus have not been elucidated in detail. In this study, we further showed that MC-LR inhibited the synthesis of GnRH in GnRH neurons via activating protein kinase a (PKA), cAMP-response element binding protein (Creb), protein kinase c (PKC), nuclear factor kappa B (NF-κB), extracellular regulated protein kinases (Erk) and P38 protein, and thus resulted in the change of activity of transcriptional enhancers or suppressors such as Oct-1, Otx-2, Pbx1a, Dlx-2, c-Jun and c-Fos. Following exposure, MC-LR-treated mice exhibited decreased GnRH level. Our data demonstrated that MC-LR can stimulate intracellular Ca2+ and cAMP to activate PKC, PKA and MAPK signaling pathways in GnRH neurons, and then inhibit Pbx1a, Oct-1, Dlx-2, Otx-2 and upregulate c-Jun and c-Fos to initiate the transcription of GnRH, which provides novel insights to explore the mechanism associated with MC-LR-induced male reproductive barriers.
Collapse
Affiliation(s)
- Jie Ding
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Jing Wang
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Haibo Jin
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Tian Xia
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Yi Cheng
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
| | - Jiang Wu
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
10
|
Hoffmann HM, Pandolfi EC, Larder R, Mellon PL. Haploinsufficiency of Homeodomain Proteins Six3, Vax1, and Otx2 Causes Subfertility in Mice via Distinct Mechanisms. Neuroendocrinology 2018; 109:200-207. [PMID: 30261489 PMCID: PMC6437011 DOI: 10.1159/000494086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 09/26/2018] [Indexed: 12/17/2022]
Abstract
Haploinsufficiency occurs when loss of one copy of a diploid gene (hemizygosity) causes a phenotype. It is relatively rare, in that most genes can produce sufficient mRNA and protein from a single copy to prevent any loss of normal activity and function. Reproduction is a complex process relying on migration of GnRH neurons from the olfactory placode to the hypothalamus during development. We have studied 3 different homeodomain genes Otx2, Vax1, and Six3 and found that the deletion of one allele for any of these genes in mice produces subfertility or infertility in one or both sexes, despite the presence of one intact allele. All 3 heterozygous mice have reduced numbers of GnRH neurons, but the mechanisms of subfertility differ significantly. This review compares the subfertility phenotypes and their mechanisms.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Erica C Pandolfi
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
| | - Rachel Larder
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California, USA,
| |
Collapse
|
11
|
Hoffmann HM, Gong P, Tamrazian A, Mellon PL. Transcriptional interaction between cFOS and the homeodomain-binding transcription factor VAX1 on the GnRH promoter controls Gnrh1 expression levels in a GnRH neuron maturation specific manner. Mol Cell Endocrinol 2018; 461:143-154. [PMID: 28890143 PMCID: PMC5756504 DOI: 10.1016/j.mce.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/23/2017] [Accepted: 09/05/2017] [Indexed: 12/18/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is required for pubertal onset and reproduction, thus the control of GnRH transcription is tightly regulated during development and adulthood. GnRH neuron development depends on transcription factors of the homeodomain family. For example, Ventral anterior homeobox 1 (Vax1) is necessary to maintain GnRH expression after embryonic day 13 in the mouse. To further our understanding of the mechanisms by which VAX1 regulates GnRH gene expression, we asked whether VAX1 interacts with other transcription factors to modify GnRH expression levels. Using the GnRH cell lines, GN11 and GT1-7, we found that activation of PKC enhances expression of the immediate early gene cFos in both GN11, and GT1-7, and represses expression of Vax1 in GT1-7. Further, VAX1 interacts with cFOS while bound to the GnRH promoter. In immature GN11 cells, VAX1 and cFOS enhance GnRH expression, whereas VAX1 and cFOS have a repressive role in the mature GT1-7 cells.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Ping Gong
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Anika Tamrazian
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Pamela L Mellon
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Nesan D, Kurrasch DM. Genetic programs of the developing tuberal hypothalamus and potential mechanisms of their disruption by environmental factors. Mol Cell Endocrinol 2016; 438:3-17. [PMID: 27720896 DOI: 10.1016/j.mce.2016.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/22/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a critical regulator of body homeostasis, influencing the autonomic nervous system and releasing trophic hormones to modulate the endocrine system. The developmental mechanisms that govern formation of the mature hypothalamus are becoming increasingly understood as research in this area grows, leading us to gain appreciation for how these developmental programs are susceptible to disruption by maternal exposure to endocrine disrupting chemicals or other environmental factors in utero. These vulnerabilities, combined with the prominent roles of the various hypothalamic nuclei in regulating appetite, reproductive behaviour, mood, and other physiologies, create a window whereby early developmental disruption can have potent long-term effects. Here we broadly outline our current understanding of hypothalamic development, with a particular focus on the tuberal hypothalamus, including what is know about nuclear coalescing and maturation. We finish by discussing how exposure to environmental or maternally-derived factors can perhaps disrupt these hypothalamic developmental programs, and potentially lead to neuroendocrine disease states.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
13
|
Wang X, Ding J, Xiang Z, Jiang P, Du J, Han X. Microcystin-LR causes sexual hormone disturbance in male rat by targeting gonadotropin-releasing hormone neurons. Toxicon 2016; 123:45-55. [DOI: 10.1016/j.toxicon.2016.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/14/2016] [Accepted: 10/19/2016] [Indexed: 11/30/2022]
|
14
|
Lonero A, Delvecchio M, Primignani P, Caputo R, Bargiacchi S, Penco S, Mauri L, Andreucci E, Faienza MF, Cavallo L. A novel OTX2 gene frameshift mutation in a child with microphthalmia, ectopic pituitary and growth hormone deficiency. J Pediatr Endocrinol Metab 2016; 29:603-5. [PMID: 26974134 DOI: 10.1515/jpem-2015-0425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/22/2016] [Indexed: 11/15/2022]
Abstract
OTX2 mutations are reported in patients with eye maldevelopment and in some cases with brain or pituitary abnormalities. We describe a child carrying a novel OTX2 heterozygous mutation. She presented microphthalmia, absence of retinal vascularization, vitreal spots and optic nerve hypoplasia in the right eye and mild macular dystrophy in the left eye. Midline brain structures and cerebral parenchyma were normal, except for the ectopic posterior pituitary gland. OTX2 sequencing showed a heterozygous c.402del mutation. Most of OTX2 mutations are nonsense or frameshift introducing a premature termination codon and resulting in a truncated protein. More rarely missense mutations occur. Our novel OTX2 mutation (c.402del) is a frameshift mutation (p.S135Lfs*43), never reported before, causing a premature codon stop 43 amino-acids downstream, which is predicted to generate a premature truncation. The mutation was associated with microphthalmia and ectopic posterior pituitary.
Collapse
|
15
|
|
16
|
Aberrantly Expressed OTX Homeobox Genes Deregulate B-Cell Differentiation in Hodgkin Lymphoma. PLoS One 2015; 10:e0138416. [PMID: 26406991 PMCID: PMC4583255 DOI: 10.1371/journal.pone.0138416] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/29/2015] [Indexed: 12/20/2022] Open
Abstract
In Hodgkin lymphoma (HL) we recently reported that deregulated homeobox gene MSX1 mediates repression of the B-cell specific transcription factor ZHX2. In this study we investigated regulation of MSX1 in this B-cell malignancy. Accordingly, we analyzed expression and function of OTX homeobox genes which activate MSX1 transcription during embryonal development in the neural plate border region. Our data demonstrate that OTX1 and OTX2 are aberrantly expressed in both HL patients and cell lines. Moreover, both OTX loci are targeted by genomic gains in overexpressing cell lines. Comparative expression profiling and subsequent pathway modulations in HL cell lines indicated that aberrantly enhanced FGF2-signalling activates the expression of OTX2. Downstream analyses of OTX2 demonstrated transcriptional activation of genes encoding transcription factors MSX1, FOXC1 and ZHX1. Interestingly, examination of the physiological expression profile of ZHX1 in normal hematopoietic cells revealed elevated levels in T-cells and reduced expression in B-cells, indicating a discriminatory role in lymphopoiesis. Furthermore, two OTX-negative HL cell lines overexpressed ZHX1 in correlation with genomic amplification of its locus at chromosomal band 8q24, supporting the oncogenic potential of this gene in HL. Taken together, our data demonstrate that deregulated homeobox genes MSX1 and OTX2 respectively impact transcriptional inhibition of (B-cell specific) ZHX2 and activation of (T-cell specific) ZHX1. Thus, we show how reactivation of a specific embryonal gene regulatory network promotes disturbed B-cell differentiation in HL.
Collapse
|
17
|
Kanasaki H, Mijiddorj T, Sukhbaatar U, Oride A, Ishihara T, Yamagami I, Kyo S. Trichostatin A reduces GnRH mRNA expression with a concomitant increase in retinaldehyde dehydrogenase in GnRH-producing neurons. Mol Cell Endocrinol 2015; 413:113-9. [PMID: 26116234 DOI: 10.1016/j.mce.2015.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 06/12/2015] [Accepted: 06/17/2015] [Indexed: 11/30/2022]
Abstract
Trichostatin A (TSA) is a selective inhibitor of mammalian histone deacetylase and is widely used to modify the ability of DNA transcription factors to bind DNA within chromatin by interfering with histone deacetylation. In the GnRH-producing neuronal cell line GT1-7, TSA significantly reduced expression of GnRH mRNA. Kisspeptin, a known regulator of GnRH release, failed to increase GnRH mRNA expression and did not modify TSA-induced reduction of GnRH expression. TSA, but not kisspeptin, increased histone acetylation in whole-cell lysates and significantly stimulated the expression of retinaldehyde dehydrogenase (RALDH), a retinoic acid (RA)-synthesizing enzyme that is known to be involved in cell differentiation. In addition, treatment of the GT1-7 cells with RA dose-dependently inhibited the expression of GnRH mRNA. Whereas, TSA-induced reduction of GnRH mRNA was not modulated by treatment with the pan-RA receptor inverse agonist BMS493 or the RA metabolism inhibitor liarozole. Our current results suggest that the RALDH and RA might not be directly involved in the reduction of GnRH expression induced by TSA, however these substances could be a novel regulator of GnRH.
Collapse
Affiliation(s)
- Haruhiko Kanasaki
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan.
| | - Tselmeg Mijiddorj
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Unurjargal Sukhbaatar
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Aki Oride
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Tomoko Ishihara
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Ikuko Yamagami
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
18
|
Bedont JL, Newman EA, Blackshaw S. Patterning, specification, and differentiation in the developing hypothalamus. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:445-68. [PMID: 25820448 DOI: 10.1002/wdev.187] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 12/21/2022]
Abstract
Owing to its complex structure and highly diverse cell populations, the study of hypothalamic development has historically lagged behind that of other brain regions. However, in recent years, a greatly expanded understanding of hypothalamic gene expression during development has opened up new avenues of investigation. In this review, we synthesize existing work to present a holistic picture of hypothalamic development from early induction and patterning through nuclear specification and differentiation, with a particular emphasis on determination of cell fate. We will also touch on special topics in the field including the prosomere model, adult neurogenesis, and integration of migratory cells originating outside the hypothalamic neuroepithelium, and how these topics relate to our broader theme.
Collapse
Affiliation(s)
- Joseph L Bedont
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth A Newman
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,High-Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Jin JM, Yang WX. Molecular regulation of hypothalamus-pituitary-gonads axis in males. Gene 2014; 551:15-25. [PMID: 25168889 DOI: 10.1016/j.gene.2014.08.048] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 07/24/2014] [Accepted: 08/24/2014] [Indexed: 10/24/2022]
Abstract
The hypothalamic-pituitary-gonadal axis (HPG) plays vital roles in reproduction and steroid hormone production in both sexes. The focus of this review is upon gene structures, receptor structures and the signaling pathways of gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The hormones' functions in reproduction as well as consequences resulting from mutations are also summarized. Specific characteristics of hormones such as the pulsatile secretions of GnRH are also covered. The different regulators of the HPG axis are introduced including kisspeptin, activin, inhibin, follistatin, androgens and estrogen. This review includes not only their basic information, but also their unique function in the HPG axis. Here we view the HPG axis as a whole, so relations between ligands and receptors are well described crossing different levels of the HPG axis. Hormone interactions and transformations are also considered. The major information of this article is depicted in three figures summarizing the current discoveries on the HPG axis. This article systematically introduces the basic knowledge of the HPG axis and provides information of the current advances relating to reproductive hormones.
Collapse
Affiliation(s)
- Jia-Min Jin
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
20
|
Liu Z, Zheng Q, Zhang X, Lu L. Microarray analysis of genes involved with shell strength in layer shell gland at the early stage of active calcification. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2014; 26:609-24. [PMID: 25049830 PMCID: PMC4093333 DOI: 10.5713/ajas.2012.12398] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 10/08/2012] [Accepted: 09/15/2012] [Indexed: 01/13/2023]
Abstract
The objective of this study was to get a comprehensive understanding of how genes in chicken shell gland modulate eggshell strength at the early stage of active calcification. Four 32-week old of purebred Xianju hens with consistent high or low shell breakage strength were grouped into two pairs. Using Affymetrix Chicken Array, a whole-transcriptome analysis was performed on hen’s shell gland at 9 h post oviposition. Gene ontology enrichment analysis for differentially expressed (DE) transcripts was performed using the web-based GOEAST, and the validation of DE-transcripts was tested by qRT-PCR. 1,195 DE-transcripts, corresponding to 941 unique genes were identified in hens with strong eggshell compared to weak shell hens. According to gene ontology annotations, there are 77 DE-transcripts encoding ion transporters and secreted extracellular matrix proteins, and at least 26 DE-transcripts related to carbohydrate metabolism or post-translation glycosylation modification; furthermore, there are 88 signaling DE-transcripts. GO term enrichment analysis suggests that some DE-transcripts mediate reproductive hormones or neurotransmitters to affect eggshell quality through a complex suite of biophysical processes. These results reveal some candidate genes involved with eggshell strength at the early stage of active calcification which may facilitate our understanding of regulating mechanisms of eggshell quality.
Collapse
Affiliation(s)
- Zhangguo Liu
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Lin'an, Zhejiang, 311300, China
| | - Qi Zheng
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Lin'an, Zhejiang, 311300, China
| | - Xueyu Zhang
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Lin'an, Zhejiang, 311300, China
| | - Lizhi Lu
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Lin'an, Zhejiang, 311300, China
| |
Collapse
|
21
|
Maternal dexamethasone exposure during pregnancy in rats disrupts gonadotropin-releasing hormone neuronal development in the offspring. Cell Tissue Res 2013; 355:409-23. [PMID: 24374911 PMCID: PMC3921457 DOI: 10.1007/s00441-013-1765-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/29/2013] [Indexed: 12/02/2022]
Abstract
The migration of gonadotropin-releasing hormone (GnRH) neurons from the olfactory placode to the preoptic area (POA) from embryonic day 13 is important for successful reproduction during adulthood. Whether maternal glucocorticoid exposure alters GnRH neuronal morphology and number in the offspring is unknown. This study determines the effect of maternal dexamethasone (DEX) exposure on enhanced green fluorescent protein (EGFP) driven by GnRH promoter neurons (TG-GnRH) in transgenic rats dual-labelled with GnRH immunofluorescence (IF-GnRH). The TG-GnRH neurons were examined in intact male and female rats at different postnatal ages, as a marker for GnRH promoter activity. Pregnant females were subcutaneously injected with DEX (0.1 mg/kg) or vehicle daily during gestation days 13–20 to examine the number of GnRH neurons in P0 male offspring. The total number of TG-GnRH neurons and TG-GnRH/IF-GnRH neuronal ratio increased from P0 and P5 stages to P47–52 stages, suggesting temporal regulation of GnRH promoter activity during postnatal development in intact rats. In DEX-treated P0 males, the number of IF-GnRH neurons decreased within the medial septum, organum vasculosom of the lamina terminalis (OVLT) and anterior hypothalamus. The percentage of TG-GnRH neurons with branched dendritic structures decreased in the OVLT of DEX-P0 males. These results suggest that maternal DEX exposure affects the number and dendritic development of early postnatal GnRH neurons in the OVLT/POA, which may lead to altered reproductive functions in adults.
Collapse
|
22
|
Veilleux HD, Van Herwerden L, Cole NJ, Don EK, De Santis C, Dixson DL, Wenger AS, Munday PL. Otx2 expression and implications for olfactory imprinting in the anemonefish, Amphiprion percula. Biol Open 2013; 2:907-15. [PMID: 24143277 PMCID: PMC3773337 DOI: 10.1242/bio.20135496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/13/2013] [Indexed: 11/20/2022] Open
Abstract
The otx2 gene encodes a transcription factor (OTX2) essential in the formation of the brain and sensory systems. Specifically, OTX2-positive cells are associated with axons in the olfactory system of mice and otx2 is upregulated in odour-exposed zebrafish, indicating a possible role in olfactory imprinting. In this study, otx2 was used as a candidate gene to investigate the molecular mechanisms of olfactory imprinting to settlement cues in the coral reef anemonefish, Amphiprion percula. The A. percula otx2 (Ap-otx2) gene was elucidated, validated, and its expression tested in settlement-stage A. percula by exposing them to behaviourally relevant olfactory settlement cues in the first 24 hours post-hatching, or daily throughout the larval phase. In-situ hybridisation revealed expression of Ap-otx2 throughout the olfactory epithelium with increased transcript staining in odour-exposed settlement-stage larval fish compared to no-odour controls, in all scenarios. This suggests that Ap-otx2 may be involved in olfactory imprinting to behaviourally relevant settlement odours in A. percula.
Collapse
Affiliation(s)
- Heather D Veilleux
- School of Marine and Tropical Biology, James Cook University , Townsville QLD 4811 , Australia ; Centre for Tropical Fisheries and Aquaculture, James Cook University , Townsville QLD 4811 , Australia
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Larder R, Kimura I, Meadows J, Clark DD, Mayo S, Mellon PL. Gene dosage of Otx2 is important for fertility in male mice. Mol Cell Endocrinol 2013; 377:16-22. [PMID: 23811236 PMCID: PMC3771655 DOI: 10.1016/j.mce.2013.06.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 06/18/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
Together, the hypothalamus, pituitary and gonads direct the development and regulation of reproductive function in mammals. Gonadotropin-releasing hormone (GnRH) expression is limited to ∼800 neurons that originate in the olfactory placode then migrate to the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal (HPG) axis is dependent upon correct neuronal migration of GnRH neurons into the hypothalamus followed by proper synthesis and pulsatile secretion of GnRH. Defects in any one of these processes causes infertility. Otx2, the vertebrate homologue of Drosophila orthodenticle, is a transcription factor that has been shown to be critical for normal brain and eye development and is expressed in both the developing GnRH neurons and the pituitary, suggesting that this gene may play a critical role in development of the HPG axis. As Otx2-null mice are embryonic lethal, we have analyzed the reproductive capacity of heterozygous Otx2 mice to determine the contribution of Otx2 gene dosage to normal HPG axis function. Our data reveal that correct dosage of Otx2 is critical for normal fertility as loss of one allele of Otx2 leads to a discernible reproductive phenotype in male mice due to disruption of the migration of GnRH neurons during development.
Collapse
Affiliation(s)
- Rachel Larder
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Ikuo Kimura
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
- Department of Genomic Drug Discovery Science, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo-ku, Kyoto 606-8501, Japan
| | - Jason Meadows
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Daniel. D. Clark
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Susan Mayo
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Pamela L. Mellon
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
- To whom correspondence should be addressed, , Telephone: 1-858-534-1312, Fax: 1-858-534-1438
| |
Collapse
|
24
|
Beby F, Lamonerie T. The homeobox gene Otx2 in development and disease. Exp Eye Res 2013; 111:9-16. [DOI: 10.1016/j.exer.2013.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 01/04/2023]
|
25
|
Abstract
Kisspeptins (Kiss) have been shown to be key components in the regulation of gonadotropin-releasing hormone (GnRH) secretion. In vitro studies have demonstrated an increase in GnRH gene expression by Kiss suggesting regulation of GnRH at both the secretory and pretranslational levels. Here, we define genetic mechanisms that mediate Kiss action on target gene expression. In vitro, sequential deletions of the mouse GnRH (mGnRH) gene promoter fused to the luciferase (LUC) reporter gene localized at kisspeptin-response element (KsRE) between -3446 and -2806 bp of the mGnRH gene. In vivo, transgenic mice bearing sequential deletions of the mGnRH gene promoter linked to the LUC reporter localized an identical KsRE. To define the mechanism of regulation, Kiss was first shown to induce nucleosome-depleted DNA within the KsRE, and a potential binding site for the transcription factor, Otx-2, was revealed. Furthermore, increased Otx-2 mRNA, protein, and binding to the KsRE after Kiss treatment were demonstrated. In conclusion, this work identified elements in GnRH-neuronal cell lines and in transgenic mice that mediate positive regulation of GnRH by Kiss. In addition, we show for the first time that Otx-2 is regulated by Kiss, and plays a role in mediating the transcriptional response of mGnRH gene.
Collapse
|
26
|
Beccari L, Marco-Ferreres R, Bovolenta P. The logic of gene regulatory networks in early vertebrate forebrain patterning. Mech Dev 2012; 130:95-111. [PMID: 23111324 DOI: 10.1016/j.mod.2012.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/09/2012] [Indexed: 01/19/2023]
Abstract
The vertebrate forebrain or prosencephalon is patterned at the beginning of neurulation into four major domains: the telencephalic, hypothalamic, retinal and diencephalic anlagen. These domains will then give rise to the majority of the brain structures involved in sensory integration and the control of higher intellectual and homeostatic functions. Understanding how forebrain pattering arises has thus attracted the interest of developmental neurobiologists for decades. As a result, most of its regulators have been identified and their hierarchical relationship is now the object of active investigation. Here, we summarize the main morphogenetic pathways and transcription factors involved in forebrain specification and propose the backbone of a possible gene regulatory network (GRN) governing its specification, taking advantage of the GRN principles elaborated by pioneer studies in simpler organisms. We will also discuss this GRN and its operational logic in the context of the remarkable morphological and functional diversification that the forebrain has undergone during evolution.
Collapse
Affiliation(s)
- Leonardo Beccari
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, c/Nicolas Cabrera, 1, Madrid 28049, Spain
| | | | | |
Collapse
|
27
|
Sabado V, Barraud P, Baker CVH, Streit A. Specification of GnRH-1 neurons by antagonistic FGF and retinoic acid signaling. Dev Biol 2012; 362:254-62. [PMID: 22200593 PMCID: PMC4561506 DOI: 10.1016/j.ydbio.2011.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 11/25/2022]
Abstract
A small population of neuroendocrine cells in the rostral hypothalamus and basal forebrain is the key regulator of vertebrate reproduction. They secrete gonadotropin-releasing hormone (GnRH-1), communicate with many areas of the brain and integrate multiple inputs to control gonad maturation, puberty and sexual behavior. In humans, disruption of the GnRH-1 system leads to hypogonadotropic gonadism and Kallmann syndrome. Unlike other neurons in the central nervous system, GnRH-1 neurons arise in the periphery, however their embryonic origin is controversial, and the molecular mechanisms that control their initial specification are not clear. Here, we provide evidence that in chick GnRH-1 neurons originate in the olfactory placode, where they are specified shortly after olfactory sensory neurons. FGF signaling is required and sufficient to induce GnRH-1 neurons, while retinoic acid represses their formation. Both pathways regulate and antagonize each other and our results suggest that the timing of signaling is critical for normal GnRH-1 neuron formation. While Kallmann's syndrome has generally been attributed to a failure of GnRH-1 neuron migration due to impaired FGF signaling, our findings suggest that in at least some Kallmann patients these neurons may never be specified. In addition, this study highlights the intimate embryonic relationship between GnRH-1 neurons and their targets and modulators in the adult.
Collapse
Affiliation(s)
- Virginie Sabado
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| | - Perrine Barraud
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Clare V. H. Baker
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Andrea Streit
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| |
Collapse
|
28
|
Gan L, Ni PY, Ge Y, Xiao YF, Sun CY, Deng L, Zhang W, Wu SS, Liu Y, Jiang W, Xin HB. Histone deacetylases regulate gonadotropin-releasing hormone I gene expression via modulating Otx2-driven transcriptional activity. PLoS One 2012; 7:e39770. [PMID: 22761896 PMCID: PMC3382570 DOI: 10.1371/journal.pone.0039770] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 05/30/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Precise coordination of the hypothalamic-pituitary-gonadal axis orchestrates the normal reproductive function. As a central regulator, the appropriate synthesis and secretion of gonadotropin-releasing hormone I (GnRH-I) from the hypothalamus is essential for the coordination. Recently, emerging evidence indicates that histone deacetylases (HDACs) play an important role in maintaining normal reproductive function. In this study, we identify the potential effects of HDACs on Gnrh1 gene transcription. METHODOLOGY/PRINCIPAL FINDINGS Inhibition of HDACs activities by trichostatin A (TSA) and valproic acid (VPA) promptly and dramatically repressed transcription of Gnrh1 gene in the mouse immortalized mature GnRH neuronal cells GT1-7. The suppression was connected with a specific region of Gnrh1 gene promoter, which contains two consensus Otx2 binding sites. Otx2 has been known to activate the basal and also enhancer-driven transcription of Gnrh1 gene. The transcriptional activity of Otx2 is negatively modulated by Grg4, a member of the Groucho-related-gene (Grg) family. In the present study, the expression of Otx2 was downregulated by TSA and VPA in GT1-7 cells, accompanied with the opposite changes of Grg4 expression. Chromatin immunoprecipitation and electrophoretic mobility shift assays demonstrated that the DNA-binding activity of Otx2 to Gnrh1 gene was suppressed by TSA and VPA. Overexpression of Otx2 partly abolished the TSA- and VPA-induced downregulation of Gnrh1 gene expression. CONCLUSIONS/SIGNIFICANCE Our data indicate that HDAC inhibitors downregulate Gnrh1 gene expression via repressing Otx2-driven transcriptional activity. This study should provide an insight for our understanding on the effects of HDACs in the reproductive system and suggests that HDACs could be potential novel targets for the therapy of GnRH-related diseases.
Collapse
Affiliation(s)
- Lu Gan
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Pei-Yan Ni
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yan Ge
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun-Fei Xiao
- Institute of Translational Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Chang-Yan Sun
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lin Deng
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wei Zhang
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Si-Si Wu
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ying Liu
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Wei Jiang
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hong-Bo Xin
- Laboratory of Cardiovascular Diseases and Laboratory of Cellular and Molecular Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Institute of Translational Medicine, Nanchang University, Nanchang, People's Republic of China
- * E-mail:
| |
Collapse
|
29
|
Beccari L, Conte I, Cisneros E, Bovolenta P. Sox2-mediated differential activation of Six3.2 contributes to forebrain patterning. Development 2012; 139:151-64. [PMID: 22096077 DOI: 10.1242/dev.067660] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The vertebrate forebrain is patterned during gastrulation into telencephalic, retinal, hypothalamic and diencephalic primordia. Specification of each of these domains requires the concerted activity of combinations of transcription factors (TFs). Paradoxically, some of these factors are widely expressed in the forebrain, which raises the question of how they can mediate regional differences. To address this issue, we focused on the homeobox TF Six3.2. With genomic and functional approaches we demonstrate that, in medaka fish, Six3.2 regulates, in a concentration-dependent manner, telencephalic and retinal specification under the direct control of Sox2. Six3.2 and Sox2 have antagonistic functions in hypothalamic development. These activities are, in part, executed by Foxg1 and Rx3, which seem to be differentially and directly regulated by Six3.2 and Sox2. Together, these data delineate the mechanisms by which Six3.2 diversifies its activity in the forebrain and highlight a novel function for Sox2 as one of the main regulators of anterior forebrain development. They also demonstrate that graded levels of the same TF, probably operating in partially independent transcriptional networks, pattern the vertebrate forebrain along the anterior-posterior axis.
Collapse
Affiliation(s)
- Leonardo Beccari
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/Nicolas Cabrera 1, Madrid 28049, Spain
| | | | | | | |
Collapse
|
30
|
Brayman MJ, Pepa PA, Berdy SE, Mellon PL. Androgen receptor repression of GnRH gene transcription. Mol Endocrinol 2012; 26:2-13. [PMID: 22074952 PMCID: PMC3248321 DOI: 10.1210/me.2011-1015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 10/13/2011] [Indexed: 11/19/2022] Open
Abstract
Alterations in androgen levels lead to reproductive defects in both males and females, including hypogonadotropic hypogonadism, anovulation, and infertility. Androgens have been shown to down-regulate GnRH mRNA levels through an androgen receptor (AR)-dependent mechanism. Here, we investigate how androgen regulates expression from the GnRH regulatory region in the GT1-7 cell line, a model of GnRH neurons. A synthetic androgen, R1881, repressed transcription from the GnRH promoter (GnRH-P) in an AR-dependent manner, and liganded AR associated with the chromatin at the GnRH-P in live GT1-7 cells. The three known octamer-binding transcription factor-1 (Oct-1) binding sites in GnRH-P were required for AR-mediated repression, although other sequences were also involved. Although a multimer of the consensus Oct-1 binding site was not repressed, a multimer of the cluster of Oct-1, Pre-B cell leukemia transcription factor (Pbx)/Prep, and NK2 homeobox 1 (Nkx2.1) binding sites, found at -106/-91 in GnRH-P, was sufficient for repression. In fact, overexpression of any of these factors disrupted the androgen response, indicating that a balance of factors in this tripartite complex is required for AR repression. AR bound to this region in EMSA, indicating a direct interaction of AR with DNA or with other transcription factors bound to GnRH-P at this sequence. Collectively, our data demonstrate that GnRH transcription is repressed by AR via multiple sequences in GnRH-P, including three Oct-1 binding sites, and that this repression requires the complex interaction of several transcription factors.
Collapse
Affiliation(s)
- Melissa J Brayman
- Department of Reproductive Medicine and The Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674, USA
| | | | | | | |
Collapse
|
31
|
Bunt J, Hasselt NE, Zwijnenburg DA, Koster J, Versteeg R, Kool M. Joint binding of OTX2 and MYC in promotor regions is associated with high gene expression in medulloblastoma. PLoS One 2011; 6:e26058. [PMID: 22016811 PMCID: PMC3189962 DOI: 10.1371/journal.pone.0026058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 09/16/2011] [Indexed: 01/19/2023] Open
Abstract
Both OTX2 and MYC are important oncogenes in medulloblastoma, the most common malignant brain tumor in childhood. Much is known about MYC binding to promoter regions, but OTX2 binding is hardly investigated. We used ChIP-on-chip data to analyze the binding patterns of both transcription factors in D425 medulloblastoma cells. When combining the data for all promoter regions in the genome, OTX2 binding showed a remarkable bi-modal distribution pattern with peaks around −250 bp upstream and +650 bp downstream of the transcription start sites (TSSs). Indeed, 40.2% of all OTX2-bound TSSs had more than one significant OTX2-binding peak. This OTX2-binding pattern was very different from the TSS-centered single peak binding pattern observed for MYC and other known transcription factors. However, in individual promoter regions, OTX2 and MYC have a strong tendency to bind in proximity of each other. OTX2-binding sequences are depleted near TSSs in the genome, providing an explanation for the observed bi-modal distribution of OTX2 binding. This contrasts to the enrichment of E-box sequences at TSSs. Both OTX2 and MYC binding independently correlated with higher gene expression. Interestingly, genes of promoter regions with multiple OTX2 binding as well as MYC binding showed the highest expression levels in D425 cells and in primary medulloblastomas. Genes within this class of promoter regions were enriched for medulloblastoma and stem cell specific genes. Our data suggest an important functional interaction between OTX2 and MYC in regulating gene expression in medulloblastoma.
Collapse
Affiliation(s)
- Jens Bunt
- Department of Oncogenomics, Academic Medical Center, Amsterdam, The Netherlands
| | - Nancy E. Hasselt
- Department of Oncogenomics, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, Amsterdam, The Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, Amsterdam, The Netherlands
| | - Marcel Kool
- Department of Oncogenomics, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
32
|
Layman WS, Hurd EA, Martin DM. Reproductive dysfunction and decreased GnRH neurogenesis in a mouse model of CHARGE syndrome. Hum Mol Genet 2011; 20:3138-50. [PMID: 21596839 DOI: 10.1093/hmg/ddr216] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CHARGE is a multiple congenital anomaly disorder and a common cause of pubertal defects, olfactory dysfunction, growth delays, deaf-blindness, balance disorders and congenital heart malformations. Mutations in CHD7, the gene encoding chromodomain helicase DNA binding protein 7, are present in 60-80% of individuals with the CHARGE syndrome. Mutations in CHD7 have also been reported in the Kallmann syndrome (olfactory dysfunction, delayed puberty and hypogonadotropic hypogonadism). CHD7 is a positive regulator of neural stem cell proliferation and olfactory sensory neuron formation in the olfactory epithelium, suggesting that the loss of CHD7 might also disrupt development of other neural populations. Here we report that female Chd7(Gt/+) mice have delays in vaginal opening and estrus onset, and erratic estrus cycles. Chd7(Gt/+) mice also have decreased circulating levels of luteinizing hormone and follicle-stimulating hormone but apparently normal responsiveness to gonadotropin-releasing hormone (GnRH) agonist and antagonist treatment. GnRH neurons in the adult Chd7(Gt/+) hypothalamus and embryonic nasal region are diminished, and there is decreased cellular proliferation in the embryonic olfactory placode. Expression levels of GnRH1 and Otx2 in the hypothalamus and GnRHR in the pituitary are significantly reduced in adult Chd7(Gt/+) mice. Additionally, Chd7 mutant embryos have CHD7 dosage-dependent reductions in expression levels of Fgfr1, Bmp4 and Otx2 in the olfactory placode. Together, these data suggest that CHD7 has critical roles in the development and maintenance of GnRH neurons for regulating puberty and reproduction.
Collapse
Affiliation(s)
- Wanda S Layman
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109-5652, USA
| | | | | |
Collapse
|
33
|
Kim HD, Choe HK, Chung S, Kim M, Seong JY, Son GH, Kim K. Class-C SOX transcription factors control GnRH gene expression via the intronic transcriptional enhancer. Mol Endocrinol 2011; 25:1184-96. [PMID: 21527504 DOI: 10.1210/me.2010-0332] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
GnRH is a pivotal hypothalamic neurohormone governing reproduction and sexual development. Because transcriptional regulation is crucial for the spatial and temporal expression of the GnRH gene, a region approximately 3.0 kb upstream of the mammalian GnRH promoter has been extensive studied. In the present study, we demonstrate a transcription-enhancer located in the first intron (intron A) region of the GnRH gene. This transcriptional enhancer harbors putative sex-determining region Y-related high-mobility-group box (SOX) family transcription factor-binding sites, which are well conserved across many mammalian species. The class-C SOX member proteins (SOX-C) (SOX4 and SOX11) specifically augment this transcriptional activation by binding to these SOX-binding sites. In accordance, SOX11 is highly enriched in immortalized GnRH-producing GT1-1 cells, and suppression of its expression significantly decreases GnRH gene expression as well as GnRH secretion. Chromatin immunoprecipitation shows that endogenous SOX-C factors recognize and bind to the intronic enhancer in GT1-1 cells and the hypothalamus. Accompanying immunohistochemical analysis demonstrates that SOX4 or SOX11 are highly expressed in the majority of hypothalamic GnRH neurons in adult mice. Taken together, these findings demonstrate that SOX-C transcription factors function as important transcriptional regulators of cell type-specific GnRH gene expression by acting on the intronic transcriptional enhancer.
Collapse
Affiliation(s)
- Hee-Dae Kim
- Department of Biological Sciences, Seoul National University, Brain Research Center for the 21st Century Frontier Program in Neuroscience, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Diaczok D, DiVall S, Matsuo I, Wondisford FE, Wolfe AM, Radovick S. Deletion of Otx2 in GnRH neurons results in a mouse model of hypogonadotropic hypogonadism. Mol Endocrinol 2011; 25:833-46. [PMID: 21436260 DOI: 10.1210/me.2010-0271] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH is the central regulator of reproductive function responding to central nervous system cues to control gonadotropin synthesis and secretion. GnRH neurons originate in the olfactory placode and migrate to the forebrain, in which they are found in a scattered distribution. Congenital idiopathic hypogonadotropic hypogonadism (CIHH) has been associated with mutations or deletions in a number of genes that participate in the development of GnRH neurons and expression of GnRH. Despite the critical role of GnRH in mammalian reproduction, a comprehensive understanding of the developmental factors that are responsible for regulating the establishment of mature GnRH neurons and the expression of GnRH is lacking. orthodenticle homeobox 2 (OTX2), a homeodomain protein required for the formation of the forebrain, has been shown to be expressed in GnRH neurons, up-regulated during GnRH neuronal development, and responsible for increased GnRH promoter activity in GnRH neuronal cell lines. Interestingly, mutations in Otx2 have been associated with human hypogonadotropic hypogonadism, but the mechanism by which Otx2 mutations cause CIHH is unknown. Here we show that deletion of Otx2 in GnRH neurons results in a significant decrease in GnRH neurons in the hypothalamus, a delay in pubertal onset, abnormal estrous cyclicity, and infertility. Taken together, these data provide in vivo evidence that Otx2 is critical for GnRH expression and reproductive competence.
Collapse
Affiliation(s)
- Daniel Diaczok
- Division of Pediatric Endocrinology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, Maryland 21287, USA
| | | | | | | | | | | |
Collapse
|
35
|
Procko C, Lu Y, Shaham S. Glia delimit shape changes of sensory neuron receptive endings in C. elegans. Development 2011; 138:1371-81. [PMID: 21350017 DOI: 10.1242/dev.058305] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuronal receptive endings, such as dendritic spines and sensory protrusions, are structurally remodeled by experience. How receptive endings acquire their remodeled shapes is not well understood. In response to environmental stressors, the nematode Caenorhabditis elegans enters a diapause state, termed dauer, which is accompanied by remodeling of sensory neuron receptive endings. Here, we demonstrate that sensory receptive endings of the AWC neurons in dauers remodel in the confines of a compartment defined by the amphid sheath (AMsh) glial cell that envelops these endings. AMsh glia remodel concomitantly with and independently of AWC receptive endings to delimit AWC receptive ending growth. Remodeling of AMsh glia requires the OTD/OTX transcription factor TTX-1, the fusogen AFF-1 and probably the vascular endothelial growth factor (VEGFR)-related protein VER-1, all acting within the glial cell. ver-1 expression requires direct binding of TTX-1 to ver-1 regulatory sequences, and is induced in dauers and at high temperatures. Our results demonstrate that stimulus-induced changes in glial compartment size provide spatial constraints on neuronal receptive ending growth.
Collapse
Affiliation(s)
- Carl Procko
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | | | | |
Collapse
|
36
|
Abstract
The hypothalamus, pituitary, and gonads coordinate to direct the development and regulation of reproductive function in mammals. Control of the hypothalamic-pituitary-gonadal axis is dependent on correct migration of gonadotropin-releasing hormone (GnRH) neurons from the nasal placode to the hypothalamus, followed by proper synthesis and pulsatile secretion of GnRH, functions absent in patients with hypogonadal hypogonadism. In this study, we identify sine oculis-related homeobox 6 (Six6) as a novel factor necessary for proper targeting of GnRH expression to the limited population of GnRH neurons within the adult mouse hypothalamus and demonstrate that it is required for proper reproductive function in both male and female mice. Female Six6-null mice exhibit a striking decrease in fertility, failing to progress through the estrous cycle normally, show any signs of successful ovulation, or produce litters. Although basal gonadotropin production in these mice is relatively normal, analysis of GnRH expression reveals a dramatic decrease in total GnRH neuron numbers. We show that expression of Six6 is dramatically increased during GnRH neuronal maturation and that overexpression of Six6 induces GnRH transcription in neuronal cells. Finally, we demonstrate that this induction in GnRH expression is mediated via binding of Six6 to evolutionarily conserved ATTA sites located within the GnRH proximal promoter. Together, these data indicate that Six6 plays an important role in the regulation of GnRH expression and hypothalamic control of fertility.
Collapse
|
37
|
Iyer AK, Brayman MJ, Mellon PL. Dynamic chromatin modifications control GnRH gene expression during neuronal differentiation and protein kinase C signal transduction. Mol Endocrinol 2011; 25:460-73. [PMID: 21239613 DOI: 10.1210/me.2010-0403] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
GnRH, a neuropeptide produced by rare, specialized hypothalamic secretory neurons, is critical for reproduction. During development, GnRH gene expression increases as neurons migrate from the olfactory placode to the hypothalamus, with highest levels in the mature, postmitotic state. While neuronal differentiation is known to be controlled by chromatin modulations, the role of chromatin dynamics in GnRH gene regulation has not been studied. Here, we use mature and immature GnRH neuronal cell models to show that both neuron-specific and protein kinase C regulation of GnRH expression are mediated by chromatin structure and histone modifications. Only in GT1-7 mature GnRH neuronal cells did GnRH regulatory elements display high sensitivity to DNase and enrichment of active histone markers histone-H3 acetylation and H3 lysine 4 trimethylation (H3K4-Me3), as well as RNA polymerase II (RNAPII) binding and enhancer RNA transcription. In contrast, H3K9-Me2, a marker of inactive chromatin, was highest in nonneuronal cells, low in GT1-7 cells, and intermediate in immature GnRH neuronal cells. The chromatin of the GnRH gene was therefore active in mature GnRH neuronal cells, inactive in nonneuronal cells, but not fully inactive in immature GnRH neuronal cells. Activation of protein kinase C (PKC) potently represses GnRH expression. PKC activation caused closing of the chromatin and decreased RNAPII occupancy at the GnRH minimal promoter (-278/-97). At GnRH-Enhancer-1 (-2404/-2100), PKC activation decreased phosphorylated-RNAPII binding, enhancer RNA transcription, and H3 acetylation, and reciprocally increased H3K9-Me2. Chromatin modifications therefore participate in the dynamic regulation and specification of GnRH expression to differentiated hypothalamic neurons.
Collapse
Affiliation(s)
- Anita K Iyer
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | |
Collapse
|
38
|
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
|
39
|
Iyer AK, Miller NLG, Yip K, Tran BH, Mellon PL. Enhancers of GnRH transcription embedded in an upstream gene use homeodomain proteins to specify hypothalamic expression. Mol Endocrinol 2010; 24:1949-64. [PMID: 20667983 PMCID: PMC2954641 DOI: 10.1210/me.2010-0156] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 07/01/2010] [Indexed: 02/07/2023] Open
Abstract
GnRH, the central regulator of reproductive function, is produced by only approximately 800 highly specialized hypothalamic neurons. Previous studies identified a minimal promoter [GnRH minimal promoter (GnRH-P)] (-173/+1) and a neuron-specific enhancer [GnRH-enhancer (E)1] (-1863/-1571) as regulatory regions in the rat gene that confer this stringent specificity of GnRH expression to differentiated GnRH neurons. In transgenic mice, these two elements target only GnRH neurons but fail to drive expression in the entire population, suggesting the existence of additional regulatory regions. Here, we define two novel, highly conserved, upstream enhancers in the GnRH gene termed GnRH-E2 (-3135/-2631) and GnRH-E3 (-4199/-3895) that increase neuron-specific GnRH expression through interactions with GnRH-E1 and GnRH-P. GnRH-E2 and GnRH-E3 regulate GnRH expression through similar mechanisms via Oct-1, Msx1, and Dlx2, which bind both GnRH-E2 and the GnRH-E3 critical region at -3952/-3895. Overexpression of Dlx2 increases transcription through GnRH-E2 and GnRH-E3. Remarkably, these novel elements are contained within the 3' untranslated region of the neighboring upstream gene, yet are marked endogenously by histone modification signatures consistent with those of enhancers. Thus, GnRH-E2 and GnRH-E3 are novel regulatory elements that, together with GnRH-E1 and GnRH-P, confer the specificity of GnRH expression to differentiated and mature GnRH neurons.
Collapse
Affiliation(s)
- Anita K Iyer
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92093-0674, USA
| | | | | | | | | |
Collapse
|
40
|
Zhao S, Kelm RJ, Fernald RD. Regulation of gonadotropin-releasing hormone-1 gene transcription by members of the purine-rich element-binding protein family. Am J Physiol Endocrinol Metab 2010; 298:E524-33. [PMID: 19996387 PMCID: PMC2838525 DOI: 10.1152/ajpendo.00597.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gonadotropin-releasing hormone-1 (GnRH1) controls reproduction by stimulating the release of gonadotropins from the pituitary. To characterize regulatory factors governing GnRH1 gene expression, we employed biochemical and bioinformatics techniques to identify novel GnRH1 promoter-binding proteins from the brain of the cichlid fish, Astatotilapia burtoni (A. burtoni). Using an in vitro DNA-binding assay followed by mass spectrometric peptide mapping, we identified two members of the purine-rich element-binding (Pur) protein family, Puralpha and Purbeta, as candidates for GnRH1 promoter binding and regulation. We found that transcripts for both Puralpha and Purbeta colocalize in GnRH1-expressing neurons in the preoptic area of the hypothalamus in A. burtoni brain. Furthermore, we confirmed in vivo binding of endogenous Puralpha and Purbeta to the upstream region of the GnRH1 gene in A. burtoni brain and mouse neuronal GT1-7 cells. Consistent with the relative promoter occupancy exhibited by endogenous Pur proteins, overexpression of Purbeta, but not Puralpha, significantly downregulated GnRH1 mRNA levels in transiently transfected GT1-7 cells, suggesting that Purbeta acts as a repressor of GnRH1 gene transcription.
Collapse
Affiliation(s)
- Sheng Zhao
- Dept. of Biology, Stanford University, California, 94305-5020, USA
| | | | | |
Collapse
|
41
|
Dateki S, Kosaka K, Hasegawa K, Tanaka H, Azuma N, Yokoya S, Muroya K, Adachi M, Tajima T, Motomura K, Kinoshita E, Moriuchi H, Sato N, Fukami M, Ogata T. Heterozygous orthodenticle homeobox 2 mutations are associated with variable pituitary phenotype. J Clin Endocrinol Metab 2010; 95:756-64. [PMID: 19965921 DOI: 10.1210/jc.2009-1334] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Although recent studies have suggested a positive role of OTX2 in pituitary as well as ocular development and function, detailed pituitary phenotypes in OTX2 mutations and OTX2 target genes for pituitary function other than HESX1 and POU1F1 remain to be determined. OBJECTIVE We aimed to examine such unresolved issues. SUBJECTS We studied 94 Japanese patients with various ocular or pituitary abnormalities. RESULTS We identified heterozygous p.K74fsX103 in case 1, p.A72fsX86 in case 2, p.G188X in two unrelated cases (3 and 4), and a 2,860,561-bp microdeletion involving OTX2 in case 5. Clinical studies revealed isolated GH deficiency in cases 1 and 5; combined pituitary hormone deficiency in case 3; abnormal pituitary structures in cases 1, 3, and 5; and apparently normal pituitary function in cases 2 and 4, together with ocular anomalies in cases 1-5. The wild-type Orthodenticle homeobox 2 (OTX2) protein transactivated the GNRH1 promoter as well as the HESX1, POU1F1, and IRBP (interstitial retinoid-binding protein) promoters, whereas the p.K74fsX103-OTX2 and p.A72fsX86-OTX2 proteins had no transactivation functions and the p.G188X-OTX2 protein had reduced ( approximately 50%) transactivation functions for the four promoters, with no dominant-negative effect. cDNA screening identified positive OTX2 expression in the hypothalamus. CONCLUSIONS The results imply that OTX2 mutations are associated with variable pituitary phenotype, with no genotype-phenotype correlations, and that OTX2 can transactivate GNRH1 as well as HESX1 and POU1F1.
Collapse
Affiliation(s)
- Sumito Dateki
- Department of Endocrinology and Metabolism, National Research Institute for Child Health and Development, 2-10-1 Ohkura, Setagaya, Tokyo 157-8535, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Larder R, Mellon PL. Otx2 induction of the gonadotropin-releasing hormone promoter is modulated by direct interactions with Grg co-repressors. J Biol Chem 2009; 284:16966-16978. [PMID: 19401468 PMCID: PMC2719334 DOI: 10.1074/jbc.m109.002485] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Indexed: 12/12/2022] Open
Abstract
Hormonal communication between the hypothalamus, pituitary, and gonads orchestrates the development and regulation of mammalian reproductive function. In mice, gonadotropin-releasing hormone (GnRH) expression is limited to approximately 1000 neurons that originate in the olfactory placode then migrate to specific positions scattered throughout the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal axis is dependent upon correct migration of GnRH neurons into the hypothalamus followed by the appropriate synthesis and pulsatile secretion of GnRH. Defects in any one of these processes can cause infertility. Recently, substantial progress has been made in identifying transcription factors, and their cofactors, that regulate not only adult expression of GnRH, but also the maturation of GnRH neurons. Here, we show that expression of Otx2, a homeodomain protein required for the formation of the forebrain, is dramatically up-regulated during GnRH neuronal maturation and that overexpression of Otx2 increases GnRH promoter activity in GnRH neuronal cell lines. Furthermore, Otx2 transcriptional activity is modulated by Grg4, a member of the Groucho-related-gene (Grg) family. Using mutational analysis, we show that a WRPW peptide motif within the Otx2 protein is required for physical interaction between Otx2 and Grg4. Without this physical interaction, Grg4 cannot repress Otx2-dependent activation of GnRH gene transcription. Taken together, these data show that Otx2 is important for GnRH expression and that direct interaction between Otx2 and Grg co-repressors regulates GnRH gene expression in hypothalamic neurons.
Collapse
Affiliation(s)
- Rachel Larder
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674
| | - Pamela L Mellon
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674.
| |
Collapse
|
43
|
Ng Y, Wolfe A, Novaira HJ, Radovick S. Estrogen regulation of gene expression in GnRH neurons. Mol Cell Endocrinol 2009; 303:25-33. [PMID: 19428988 PMCID: PMC2680765 DOI: 10.1016/j.mce.2009.01.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 01/16/2009] [Accepted: 01/16/2009] [Indexed: 12/27/2022]
Abstract
Estrogen plays an essential role in the regulation of the female reproductive hormone axis, and specifically is a major regulator of GnRH neuronal function in the female brain. GnRH neuronal cell lines were used to explore the direct effects of estradiol on gene expression in GnRH neurons. The presence of estrogen receptor (ER) binding sites was established by a receptor-binding assay, and estrogen receptor alpha and beta mRNA were identified in GN11 cells and ERbeta in GT1-7 cells using RT-PCR analysis of mRNA. ERalpha was more abundantly expressed in GN11 cells than ERbeta as assessed by real-time PCR. Additionally, GN11 cells expressed significantly more of both ERalpha and beta than GT1-7 cells. Functional studies in GN11 and GT1-7 demonstrated estrogen down regulation of endogenous mouse GnRH mRNA levels using quantitative real-time PCR (qRT-PCR). Correspondingly, estradiol also reduced secretion of GnRH from both the GN11 and GT1-7 cell lines. Since estradiol has been shown to regulate progesterone receptor (PR) expression; similar studies were performed demonstrating an estradiol mediated increase in PR in both cell lines. Estradiol regulation of ER expression was also explored and these studies indicated that estradiol decreased ERalpha and ERbeta mRNA levels in a dose-dependent manner in GN11 and GT1-7 cells. These effects were blocked by the addition of the estrogen receptor antagonist ICI 182,780. Both PPT, a specific ERalpha agonist, and DPN, a specific ERbeta agonist, inhibited GnRH gene expression in GN11 cells, but only DPN inhibited GnRH gene expression in GT1-7 cells, consistent with their undetectable levels of ERalpha expression. These studies characterize a direct inhibitory effect of estradiol on GnRH in GnRH neurons, and a direct stimulatory effect of estradiol on PR gene expression. In addition, the agonist studies indicate that there is a functional overlap of ERalpha and ERbeta regulation in GnRH neurons. These studies may give insight into the molecular regulation of estrogen negative feedback in the central reproductive axis.
Collapse
Affiliation(s)
| | | | - Horacio J. Novaira
- Johns Hopkins University School of Medicine Department of Pediatrics, Division of Endocrinology
| | - Sally Radovick
- Johns Hopkins University School of Medicine Department of Pediatrics, Division of Endocrinology
| |
Collapse
|
44
|
Abstract
Homeobox genes are an evolutionarily conserved class of transcription factors that are key regulators of developmental processes such as regional specification, patterning, migration and differentiation. In both mouse and humans, the developing forebrain is marked by distinct boundaries of homeobox gene expression at different developmental time points. These genes regulate the patterning of the forebrain along the dorsal/ventral and rostral/caudal axes and are also essential for the differentiation of specific neuronal subtypes. Inhibitory interneurons that arise from the ganglionic eminences and migrate tangentially to the neocortex and hippocampus are dramatically affected by mutations in several homeobox genes. In this review, we discuss the identification, expression patterns, loss- and/or gain-of-function models, and confirmed transcriptional targets for a set of homeobox genes required for the correct development of the forebrain in the mouse. In humans, mutations of homeobox genes expressed in the forebrain have been shown to result in mental retardation, epilepsy or movement disorders. The number of homeobox genes currently linked to human nervous system disease is surprisingly low, perhaps reflecting the essential functions of these genes throughout embryogenesis or the degree of functional redundancy during central nervous system development.
Collapse
Affiliation(s)
- J T Wigle
- Department of Biochemistry & Medical Genetics; Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
45
|
Zhao S, Korzan WJ, Chen CC, Fernald RD. Heterogeneous nuclear ribonucleoprotein A/B and G inhibits the transcription of gonadotropin-releasing-hormone 1. Mol Cell Neurosci 2008; 37:69-84. [PMID: 17920292 PMCID: PMC2262047 DOI: 10.1016/j.mcn.2007.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 08/14/2007] [Accepted: 08/21/2007] [Indexed: 12/16/2022] Open
Abstract
Gonadotropin-releasing hormone 1 (GnRH1) causes the release of gonadotropins from the pituitary to control reproduction. Here we report that two heterogeneous nuclear ribonucleoproteins (hnRNP-A/B and hnRNP-G) bind to the GnRH-I upstream promoter region in a cichlid fish Astatotilapia burtoni. We identified these binding proteins using a newly developed homology based method of mass spectrometric peptide mapping. We show that both hnRNP-A/B and hnRNP-G co-localize with GnRH1 in the pre-optic area of the hypothalamus in the brain. We also demonstrated that these ribonucleoproteins exhibit similar binding capacity in vivo, using immortalized mouse GT1-7 cells where overexpression of either hnRNP-A/B or hnRNP-G significantly down-regulates GnRH1 mRNA levels in GT1-7 cells, suggesting that both act as repressors in GnRH1 transcriptional regulation.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Biological Sciences & Program in Neuroscience, Stanford University, Stanford, California 94305-5020
| | - Wayne J. Korzan
- Department of Biological Sciences & Program in Neuroscience, Stanford University, Stanford, California 94305-5020
| | - Chun-Chun Chen
- Department of Biological Sciences & Program in Neuroscience, Stanford University, Stanford, California 94305-5020
| | - Russell D. Fernald
- Department of Biological Sciences & Program in Neuroscience, Stanford University, Stanford, California 94305-5020
| |
Collapse
|
46
|
Rave-Harel N, Miller NLG, Givens ML, Mellon PL. The Groucho-related gene family regulates the gonadotropin-releasing hormone gene through interaction with the homeodomain proteins MSX1 and OCT1. J Biol Chem 2005; 280:30975-83. [PMID: 16002402 PMCID: PMC2773698 DOI: 10.1074/jbc.m502315200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is exclusively expressed in a unique population of hypothalamic neurons that controls reproductive function. GnRH gene expression is highly dynamic. Its transcriptional activity is regulated in a complex spatiotemporal manner during embryonic development and postnatal life. Although a variety of transcription factors have been identified as regulators of GnRH transcription, most are promiscuous in their DNA-binding requirements, and none are solely expressed in GnRH neurons. Their specific activity is probably determined by interactions with distinct cofactors. Here we find that the Groucho-related gene (GRG) family of co-repressors is expressed in a model cell line for the GnRH neuron and co-expresses with GnRH during prenatal development. GRG proteins associate in vivo with the GnRH promoter. Furthermore, GRG proteins interact with two regulators of GnRH transcription, the homeodomain proteins MSX1 and OCT1. Co-transfection experiments indicate that GRG proteins regulate GnRH promoter activity. The long GRG forms enhance MSX1 repression and counteract OCT1 activation of the GnRH gene. In contrast, the short form, GRG5, has a dominant-negative effect on MSX1-dependent repression. Taken together, these data suggest that the dynamic switch between activation and repression of GnRH transcription is mediated by recruitment of the GRG co-regulators.
Collapse
Affiliation(s)
- Naama Rave-Harel
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, 92093-0674
| | - Nichol L. G. Miller
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, 92093-0674
| | - Marjory L. Givens
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, 92093-0674
| | - Pamela L. Mellon
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, 92093-0674
- Department of Neurosciences, University of California, San Diego, La Jolla, California, 92093-0674
- To whom correspondence should be addressed: Dept. of Reproductive Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0674. Tel.: 858-534-1312; Fax: 858-534-1438;
| |
Collapse
|
47
|
Tang Q, Mazur M, Mellon PL. The protein kinase C pathway acts through multiple transcription factors to repress gonadotropin-releasing hormone gene expression in hypothalamic GT1-7 neuronal cells. Mol Endocrinol 2005; 19:2769-79. [PMID: 15994198 PMCID: PMC2935804 DOI: 10.1210/me.2004-0463] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The GnRH gene uses two well-defined regions to target expression to a small population of hypothalamic GnRH neurons: a 173-bp proximal promoter and a 300-bp enhancer localized at approximately -1800 to -1500 bp from the start site. Interaction of multiple factors with the GnRH enhancer and promoter is required to confer neuron-specific expression in vivo and in cells in culture. In addition, the expression of the GnRH gene is regulated by numerous neurotransmitters and hormones. Several of these effectors act through membrane receptors to trigger the protein kinase C pathway, and 12-O-tetradecanoyl phorbol-13-acetate (TPA), a modulator of this pathway, has been shown to suppress GnRH gene expression through the promoter. We find that TPA suppresses expression through the GnRH enhancer as well as the promoter. In the enhancer, an Oct-1 binding site, a Pbx/Prep binding site, Msx/Dlx binding sites, and a previously unidentified protein-binding element at -1793, all contribute to TPA suppression. TPA treatment leads to decreased binding of Oct-1 and Pbx1a/Prep to their sites. However, a complex formed by GT1-7 nuclear extracts on the -1793 site is not affected by TPA treatment. It is known that cooperative interaction among multiple factors is necessary for GnRH gene expression; thus, one mechanism by which TPA suppresses GnRH gene expression is to disengage some of these factors from their cis-regulatory elements.
Collapse
Affiliation(s)
- Qingbo Tang
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
48
|
Givens ML, Kurotani R, Rave-Harel N, Miller NLG, Mellon PL. Phylogenetic footprinting reveals evolutionarily conserved regions of the gonadotropin-releasing hormone gene that enhance cell-specific expression. Mol Endocrinol 2004; 18:2950-66. [PMID: 15319450 PMCID: PMC2932476 DOI: 10.1210/me.2003-0437] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Reproductive function is controlled by the hypothalamic neuropeptide, GnRH, which serves as the central regulator of the hypothalamic-pituitary-gonadal axis. GnRH expression is limited to a small population of neurons in the hypothalamus. Targeting this minute population of neurons (as few as 800 in the mouse) requires regulatory elements upstream of the GnRH gene that remain to be fully characterized. Previously, we have identified an evolutionarily conserved promoter region (-173 to +1) and an enhancer (-1863 to -1571) in the rat gene that targets a subset of the GnRH neurons in vivo. In the present study, we used phylogenetic sequence comparison between human and rodents and analysis of the transcription factor clusters within conserved regions in an attempt to identify additional upstream regulatory elements. This approach led to the characterization of a new upstream enhancer that regulates expression of GnRH in a cell-specific manner. Within this upstream enhancer are nine binding sites for Octamer-binding transcription factor 1 (OCT1), known to be an important transcriptional regulator of GnRH gene expression. In addition, we have identified nuclear factor I (NF1) binding to multiple elements in the GnRH-regulatory regions, each in close proximity to OCT1. We show that OCT1 and NF1 physically and functionally interact. Moreover, the OCT1 and NF1 binding sites in the regulatory regions appear to be essential for appropriate GnRH gene expression. These findings indicate a role for this upstream enhancer and novel OCT1/NF1 complexes in neuron-restricted expression of the GnRH gene.
Collapse
Affiliation(s)
- Marjory L Givens
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | | | | | |
Collapse
|
49
|
Rave-Harel N, Givens ML, Nelson SB, Duong HA, Coss D, Clark ME, Hall SB, Kamps MP, Mellon PL. TALE homeodomain proteins regulate gonadotropin-releasing hormone gene expression independently and via interactions with Oct-1. J Biol Chem 2004; 279:30287-97. [PMID: 15138251 PMCID: PMC2935805 DOI: 10.1074/jbc.m402960200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is the central regulator of reproductive function. Expression of the GnRH gene is confined to a rare population of neurons scattered throughout the hypothalamus. Restricted expression of the rat GnRH gene is driven by a multicomponent enhancer and an evolutionarily conserved promoter. Oct-1, a ubiquitous POU homeodomain transcription factor, was identified as an essential factor regulating GnRH transcription in the GT1-7 hypothalamic neuronal cell line. In this study, we conducted a two-hybrid interaction screen in yeast using a GT1-7 cDNA library to search for specific Oct-1 cofactors. Using this approach, we isolated Pbx1b, a TALE homeodomain transcription factor that specifically associates with Oct-1. We show that heterodimers containing Pbx/Prep1 or Pbx/Meis1 TALE homeodomain proteins bind to four functional elements within the GnRH regulatory region, each in close proximity to an Oct-1-binding site. Cotransfection experiments indicate that TALE proteins are essential for GnRH promoter activity in the GT1-7 cells. Moreover, Pbx1 and Oct-1, as well as Prep1 and Oct-1, form functional complexes that enhance GnRH gene expression. Finally, Pbx1 is expressed in GnRH neurons in embryonic as well as mature mice, suggesting that the associations between TALE homeodomain proteins and Oct-1 regulate neuron-specific expression of the GnRH gene in vivo.
Collapse
Affiliation(s)
- Naama Rave-Harel
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Marjory L. Givens
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Shelley B. Nelson
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Hao A. Duong
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Djurdjica Coss
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Melody E. Clark
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Sara Barth Hall
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
| | - Mark P. Kamps
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, California 92903
| | - Pamela L. Mellon
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92903
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, California 92903
- To whom correspondence should be addressed: Dept. of Reproductive Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0674; Tel.: 858-534-1312; Fax: 858-534-1438;
| |
Collapse
|
50
|
Nédélec S, Foucher I, Brunet I, Bouillot C, Prochiantz A, Trembleau A. Emx2 homeodomain transcription factor interacts with eukaryotic translation initiation factor 4E (eIF4E) in the axons of olfactory sensory neurons. Proc Natl Acad Sci U S A 2004; 101:10815-20. [PMID: 15247416 PMCID: PMC490017 DOI: 10.1073/pnas.0403824101] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We report that Emx2 homeogene is expressed at the mRNA and protein levels in the adult mouse olfactory neuroepithelium. As expected for a transcription factor, Emx2 is present in the nucleus of immature and mature olfactory sensory neurons. However, the protein is also detected in the axonal compartment of these neurons, both in the olfactory mucosa axon bundles and in axon terminals within the olfactory bulb. Emx2 axonal staining is heterogeneous, suggesting an association with particles. Subcellular fractionations of olfactory bulb synaptosomes, combined with chemical lesions of olfactory neurons, confirm the presence of Emx2 in axon terminals. Significant amounts of Emx2 protein cosediment with high density synaptosomal subfractions containing eukaryotic translation initiation factor 4E (eIF4E). Nonionic detergents and RNase treatments failed to detach eIF4E and Emx2 from these high-density fractions enriched in vesicles and granular structures. In addition, Emx2 and eIF4E can be coimmunoprecipitated from olfactory mucosa and bulb extracts and interact directly, as demonstrated in pull-down experiments. Emx2 axonal localization, association with high-density particles and interaction with eIF4E strongly suggest that this transcription factor has new nonnuclear functions most probably related to the local control of protein translation in the olfactory sensory neuron axons. Finally, we show that two other brain-expressed homeoproteins, Otx2 and Engrailed 2, also bind eIF4E, indicating that several homeoproteins may modulate eIF4E functions in the developing and adult nervous system.
Collapse
Affiliation(s)
- Stéphane Nédélec
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8542, Neuropharmacology and Development, Ecole Normale Supérieure, 46 Rue d'Ulm, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|