1
|
Zouaghi Y, Alpern D, Gardeux V, Russeil J, Deplancke B, Santoni F, Pitteloud N, Messina A. Transcriptomic profiling of murine GnRH neurons reveals developmental trajectories linked to human reproduction and infertility. Theranostics 2025; 15:3673-3692. [PMID: 40093908 PMCID: PMC11905127 DOI: 10.7150/thno.91873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/22/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: Neurons producing Gonadotropin-Releasing Hormone (GnRH) are essential for human reproduction and have to migrate from nose to brain during prenatal life. Impaired GnRH neuron biology results in alterations of the reproductive axis, including delayed puberty and infertility, with considerable effects on quality of life and metabolic health. Although various genes have been implicated, the molecular causes of these conditions remain elusive, with most patients lacking a genetic diagnosis. Methods: GnRH neurons and non-GnRH cells were FACS-isolated from mouse embryo microdissections to perform high-resolution transcriptomic profiling during mouse embryonic development. We analyzed our dataset to reveal GnRH neuron molecular identity, gene expression dynamics, and cell-to-cell communication. The spatial context of candidate genes was validated using in situ hybridization and spatial transcriptomic analysis. The possible links with human reproduction in health and disease were explored using enrichment analysis on GWAS data and analyzing the genetic burden of patients with congenital GnRH deficiency. Results: GnRH neurons undergo a profound transcriptional shift as they migrate from the nose to the brain and display expression trajectories associating with distinct biological processes, including cell migration, neuronal projections, and synapse formation. We revealed a timely and spatially restricted modulation of signaling pathways involving known and novel molecules, including Semaphorins and Neurexins, respectively. A particular set of genes, whose expression in GnRH neurons timely rises in late developmental stages, showed a strong association with GWAS genes linked with human reproductive onset. Finally, some of the identified trajectories harbor a diagnostic potential for congenital hypogonadism. This is supported by genetic analysis in a large cohort of patients affected by congenital GnRH deficiency, revealing a high mutation burden in patients compared to healthy controls. Conclusion: We charted the landscape of gene expression dynamics underlying murine GnRH neuron embryonic development. Our study highlights new genes in GnRH neuron development and provides novel insights linking those genes with human reproduction.
Collapse
Affiliation(s)
- Yassine Zouaghi
- Department of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vincent Gardeux
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Federico Santoni
- Department of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Andrea Messina
- Department of Endocrinology, Diabetes and Metabolism, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
2
|
Griffin C, Saint-Jeannet JP. In vitro modeling of cranial placode differentiation: Recent advances, challenges, and perspectives. Dev Biol 2024; 506:20-30. [PMID: 38052294 PMCID: PMC10843546 DOI: 10.1016/j.ydbio.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
Cranial placodes are transient ectodermal thickenings that contribute to a diverse array of organs in the vertebrate head. They develop from a common territory, the pre-placodal region that over time segregates along the antero-posterior axis into individual placodal domains: the adenohypophyseal, olfactory, lens, trigeminal, otic, and epibranchial placodes. These placodes terminally differentiate into the anterior pituitary, the lens, and contribute to sensory organs including the olfactory epithelium, and inner ear, as well as several cranial ganglia. To study cranial placodes and their derivatives and generate cells for therapeutic purposes, several groups have turned to in vitro derivation of placodal cells from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs). In this review, we summarize the signaling cues and mechanisms involved in cranial placode induction, specification, and differentiation in vivo, and discuss how this knowledge has informed protocols to derive cranial placodes in vitro. We also discuss the benefits and limitations of these protocols, and the potential of in vitro cranial placode modeling in regenerative medicine to treat cranial placode-related pathologies.
Collapse
Affiliation(s)
- Casey Griffin
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
3
|
Cho HJ, Gurbuz F, Stamou M, Kotan LD, Farmer SM, Can S, Tompkins MF, Mammadova J, Altincik SA, Gokce C, Catli G, Bugrul F, Bartlett K, Turan I, Balasubramanian R, Yuksel B, Seminara SB, Wray S, Topaloglu AK. POU6F2 mutation in humans with pubertal failure alters GnRH transcript expression. Front Endocrinol (Lausanne) 2023; 14:1203542. [PMID: 37600690 PMCID: PMC10436210 DOI: 10.3389/fendo.2023.1203542] [Citation(s) in RCA: 1] [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] [Received: 04/10/2023] [Accepted: 06/23/2023] [Indexed: 08/22/2023] Open
Abstract
Idiopathic hypogonadotropic hypogonadism (IHH) is characterized by the absence of pubertal development and subsequent impaired fertility often due to gonadotropin-releasing hormone (GnRH) deficits. Exome sequencing of two independent cohorts of IHH patients identified 12 rare missense variants in POU6F2 in 15 patients. POU6F2 encodes two distinct isoforms. In the adult mouse, expression of both isoform1 and isoform2 was detected in the brain, pituitary, and gonads. However, only isoform1 was detected in mouse primary GnRH cells and three immortalized GnRH cell lines, two mouse and one human. To date, the function of isoform2 has been verified as a transcription factor, while the function of isoform1 has been unknown. In the present report, bioinformatics and cell assays on a human-derived GnRH cell line reveal a novel function for isoform1, demonstrating it can act as a transcriptional regulator, decreasing GNRH1 expression. In addition, the impact of the two most prevalent POU6F2 variants, identified in five IHH patients, that were located at/or close to the DNA-binding domain was examined. Notably, one of these mutations prevented the repression of GnRH transcripts by isoform1. Normally, GnRH transcription increases as GnRH cells mature as they near migrate into the brain. Augmentation earlier during development can disrupt normal GnRH cell migration, consistent with some POU6F2 variants contributing to the IHH pathogenesis.
Collapse
Affiliation(s)
- Hyun-Ju Cho
- Cellular and Developmental Neurobiology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Fatih Gurbuz
- Division of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Maria Stamou
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Leman Damla Kotan
- Division of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Stephen Matthew Farmer
- Cellular and Developmental Neurobiology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Sule Can
- Division of Pediatric Endocrinology, İzmir Tepecik Training and Research Hospital, Health Sciences University, İzmir, Türkiye
| | - Miranda Faith Tompkins
- Cellular and Developmental Neurobiology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jamala Mammadova
- Division of Pediatric Endocrinology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Türkiye
| | - S. Ayca Altincik
- Division of Pediatric Endocrinology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - Cumali Gokce
- Division of Endocrinology, Faculty of Medicine, Mustafa Kemal University, Hatay, Türkiye
| | - Gonul Catli
- Division of Pediatric Endocrinology, İzmir Tepecik Training and Research Hospital, Health Sciences University, İzmir, Türkiye
| | - Fuat Bugrul
- Division of Pediatric Endocrinology, Faculty of Medicine, Selcuk University, Konya, Türkiye
| | - Keenan Bartlett
- Cellular and Developmental Neurobiology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ihsan Turan
- Division of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Ravikumar Balasubramanian
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Bilgin Yuksel
- Division of Pediatric Endocrinology, Faculty of Medicine, Cukurova University, Adana, Türkiye
| | - Stephanie B. Seminara
- Harvard Reproductive Sciences Center, The Reproductive Endocrine Unit and The Endocrine Unit of the Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - A. Kemal Topaloglu
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Mississippi Medical Center, Jackson, MS, United States
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MS, United States
| |
Collapse
|
4
|
Wang Y, Madhusudan S, Cotellessa L, Kvist J, Eskici N, Yellapragada V, Pulli K, Lund C, Vaaralahti K, Tuuri T, Giacobini P, Raivio T. Deciphering the Transcriptional Landscape of Human Pluripotent Stem Cell-Derived GnRH Neurons: The Role of Wnt Signaling in Patterning the Neural Fate. Stem Cells 2022; 40:1107-1121. [PMID: 36153707 PMCID: PMC9806769 DOI: 10.1093/stmcls/sxac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/14/2022] [Indexed: 01/05/2023]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons lay the foundation for human development and reproduction; however, the critical cell populations and the entangled mechanisms underlying the development of human GnRH neurons remain poorly understood. Here, by using our established human pluripotent stem cell-derived GnRH neuron model, we decoded the cellular heterogeneity and differentiation trajectories at the single-cell level. We found that a glutamatergic neuron population, which generated together with GnRH neurons, showed similar transcriptomic properties with olfactory sensory neuron and provided the migratory path for GnRH neurons. Through trajectory analysis, we identified a specific gene module activated along the GnRH neuron differentiation lineage, and we examined one of the transcription factors, DLX5, expression in human fetal GnRH neurons. Furthermore, we found that Wnt inhibition could increase DLX5 expression and improve the GnRH neuron differentiation efficiency through promoting neurogenesis and switching the differentiation fates of neural progenitors into glutamatergic neurons/GnRH neurons. Our research comprehensively reveals the dynamic cell population transition and gene regulatory network during GnRH neuron differentiation.
Collapse
Affiliation(s)
- Yafei Wang
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Venkatram Yellapragada
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carina Lund
- Folkhälsan Research Center, Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland,New Children’s Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | | | - Taneli Raivio
- Corresponding author: Taneli Raivio, Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Yellapragada V, Eskici N, Wang Y, Madhusudan S, Vaaralahti K, Tuuri T, Raivio T. Time and dose-dependent effects of FGF8-FGFR1 signaling in GnRH neurons derived from human pluripotent stem cells. Dis Model Mech 2022; 15:276003. [PMID: 35833364 PMCID: PMC9403748 DOI: 10.1242/dmm.049436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper. Summary: This article demonstrates the essential role FGF8–FGFR1 signaling has in the development of gonadotropin-releasing hormone (GnRH)-expressing neurons by using a human stem cell model.
Collapse
Affiliation(s)
- Venkatram Yellapragada
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Yafei Wang
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, 00029 Helsinki University Hospital, Helsinki, Finland
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,New Children's Hospital, Pediatric Research Center, 00029 Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
6
|
Tanaka S, Zmora N, Levavi-Sivan B, Zohar Y. Chemogenetic Depletion of Hypophysiotropic GnRH Neurons Does Not Affect Fertility in Mature Female Zebrafish. Int J Mol Sci 2022; 23:ijms23105596. [PMID: 35628411 PMCID: PMC9143870 DOI: 10.3390/ijms23105596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
The hypophysiotropic gonadotropin-releasing hormone (GnRH) and its neurons are crucial for vertebrate reproduction, primarily in regulating luteinizing hormone (LH) secretion and ovulation. However, in zebrafish, which lack GnRH1, and instead possess GnRH3 as the hypophysiotropic form, GnRH3 gene knockout did not affect reproduction. However, early-stage ablation of all GnRH3 neurons causes infertility in females, implicating GnRH3 neurons, rather than GnRH3 peptides in female reproduction. To determine the role of GnRH3 neurons in the reproduction of adult females, a Tg(gnrh3:Gal4ff; UAS:nfsb-mCherry) line was generated to facilitate a chemogenetic conditional ablation of GnRH3 neurons. Following ablation, there was a reduction of preoptic area GnRH3 neurons by an average of 85.3%, which was associated with reduced pituitary projections and gnrh3 mRNA levels. However, plasma LH levels were unaffected, and the ablated females displayed normal reproductive capacity. There was no correlation between the number of remaining GnRH3 neurons and reproductive performance. Though it is possible that the few remaining GnRH3 neurons can still induce an LH surge, our findings are consistent with the idea that GnRH and its neurons are likely dispensable for LH surge in zebrafish. Altogether, our results resurrected questions regarding the functional homology of the hypophysiotropic GnRH1 and GnRH3 in controlling ovulation.
Collapse
Affiliation(s)
- Sakura Tanaka
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD 21202, USA; (S.T.); (N.Z.)
| | - Nilli Zmora
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD 21202, USA; (S.T.); (N.Z.)
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel;
| | - Yonathan Zohar
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD 21202, USA; (S.T.); (N.Z.)
- Correspondence:
| |
Collapse
|
7
|
Hoffmann HM, Meadows JD, Breuer JA, Yaw AM, Nguyen D, Tonsfeldt KJ, Chin AY, Devries BM, Trang C, Oosterhouse HJ, Lee JS, Doser JW, Gorman MR, Welsh DK, Mellon PL. The transcription factors SIX3 and VAX1 are required for suprachiasmatic nucleus circadian output and fertility in female mice. J Neurosci Res 2021; 99:2625-2645. [PMID: 34212416 PMCID: PMC8577618 DOI: 10.1002/jnr.24864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The homeodomain transcription factors sine oculis homeobox 3 (Six3) and ventral anterior homeobox 1 (Vax1) are required for brain development. Their expression in specific brain areas is maintained in adulthood, where their functions are poorly understood. To identify the roles of Six3 and Vax1 in neurons, we conditionally deleted each gene using Synapsincre , a promoter targeting maturing neurons, and generated Six3syn and Vax1syn mice. Six3syn and Vax1syn females, but not males, had reduced fertility, due to impairment of the luteinizing hormone (LH) surge driving ovulation. In nocturnal rodents, the LH surge requires a precise timing signal from the brain's circadian pacemaker, the suprachiasmatic nucleus (SCN), near the time of activity onset. Indeed, both Six3syn and Vax1syn females had impaired rhythmic SCN output, which was associated with weakened Period 2 molecular clock function in both Six3syn and Vax1syn mice. These impairments were associated with a reduction of the SCN neuropeptide vasoactive intestinal peptide in Vax1syn mice and a modest weakening of SCN timekeeping function in both Six3syn and Vax1syn mice. Changes in SCN function were associated with mistimed peak PER2::LUC expression in the SCN and pituitary in both Six3syn and Vax1syn females. Interestingly, Six3syn ovaries presented reduced sensitivity to LH, causing reduced ovulation during superovulation. In conclusion, we have identified novel roles of the homeodomain transcription factors SIX3 and VAX1 in neurons, where they are required for proper molecular circadian clock function, SCN rhythmic output, and female fertility.
Collapse
Affiliation(s)
- Hanne M. Hoffmann
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Jason D. Meadows
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Joseph A. Breuer
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra M. Yaw
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Karen J. Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Austin Y. Chin
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Brooke M. Devries
- Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA
| | - Crystal Trang
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Haley J. Oosterhouse
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Sora Lee
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey W. Doser
- CANR Statistical Consulting Center, Michigan State University, East Lansing, MI, USA
| | - Michael R. Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Psychology, University of California, San Diego, La Jolla, CA, USA
| | - David K. Welsh
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Pamela L. Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
8
|
Lavalle SN, Chou T, Hernandez J, Naing NCP, Tonsfeldt KJ, Hoffmann HM, Mellon PL. Kiss1 is differentially regulated in male and female mice by the homeodomain transcription factor VAX1. Mol Cell Endocrinol 2021; 534:111358. [PMID: 34098016 PMCID: PMC8319105 DOI: 10.1016/j.mce.2021.111358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Regulation of Kiss1 transcription is crucial to the development and function of the reproductive axis. The homeodomain transcription factor, ventral anterior homeobox 1 (VAX1), has been implicated as a potential regulator of Kiss1 transcription. However, it is unknown whether VAX1 directly mediates transcription within kisspeptin neurons or works indirectly by acting upstream of kisspeptin neuron populations. This study tested the hypothesis that VAX1 within kisspeptin neurons regulates Kiss1 gene expression. We found that VAX1 acts as a repressor of Kiss1 in vitro and within the male arcuate nucleus in vivo. In female mice, we found that the loss of VAX1 caused a reduction in Kiss1 expression and Kiss1-containing neurons in the anteroventral periventricular nucleus at the time of the preovulatory luteinizing hormone surge, but was compensated by an increase in Kiss1-cFos colocalization. Despite changes in Kiss1 transcription, gonadotropin levels were unaffected and there were no impairments to fertility.
Collapse
Affiliation(s)
- Shanna N Lavalle
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Teresa Chou
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jacqueline Hernandez
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nay Chi P Naing
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hanne M Hoffmann
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA; Department of Animal Science and the Reproductive and Developmental Sciences Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
9
|
Rjiba K, Ayech H, Kraiem O, Slimani W, Jelloul A, Ben Hadj Hmida I, Mahdhaoui N, Saad A, Mougou-Zerelli S. Disorders of sex development in Wolf-Hirschhorn syndrome: a genotype-phenotype correlation and MSX1 as candidate gene. Mol Cytogenet 2021; 14:12. [PMID: 33627176 PMCID: PMC7905666 DOI: 10.1186/s13039-021-00531-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/28/2021] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Wolf-Hirschhorn (WHS) is a set of congenital physical anomalies and mental retardation associated with a partial deletion of the short arm of chromosome 4. To establish a genotype-phenotype correlation; we carried out a molecular cytogenetic analysis on two Tunisian WHS patients. Patient 1 was a boy of 1-year-old, presented a typical WHS phenotype while patient 2, is a boy of 2 days presented an hypospadias, a micropenis and a cryptorchidie in addition to the typical WHS phenotype. Both the array comparative genomic hybridization and fluorescence in situ hybridization techniques were used. RESULTS Results of the analysis showed that patient 2 had a greater deletion size (4.8 Mb) of chromosome 4 than patient 1 (3.4 Mb). Here, we notice that the larger the deletion, the more genes are likely to be involved, and the more severe the phenotype is likely to be. If we analyze the uncommon deleted region between patient1 and patient 2 we found that the Muscle Segment Homeobox (MSX1) gene is included in this region. MSX1 is a critical transcriptional repressor factor, expressed in the ventral side of the developing anterior pituitary and implicated in gonadotrope differentiation. Msx1 acts as a negative regulatory pituitary development by repressing the gonadotropin releasing hormone (GnRH) genes during embryogenesis. We hypothesized that the deletion of MSX1 in our patient may deregulate the androgen synthesis. CONCLUSION Based on the MSX1 gene function, its absence might be indirectly responsible for the hypospadias phenotype by contributing to the spatiotemporal regulation of GnRH transcription during development.
Collapse
Affiliation(s)
- Khouloud Rjiba
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia.,Higher Institute of Biotechnology, Monastir University, Monastir, Tunisia.,Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Hédia Ayech
- Pediatric Department, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Olfa Kraiem
- Pediatric Department, Regional Hospital, Kairouan, Tunisia
| | - Wafa Slimani
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia.,Higher Institute of Biotechnology, Monastir University, Monastir, Tunisia.,Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Afef Jelloul
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Imen Ben Hadj Hmida
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Nabiha Mahdhaoui
- Pediatric Department, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Ali Saad
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia.,Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Soumaya Mougou-Zerelli
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia. .,Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia.
| |
Collapse
|
10
|
The Homeodomain Transcription Factors Vax1 and Six6 Are Required for SCN Development and Function. Mol Neurobiol 2019; 57:1217-1232. [PMID: 31705443 DOI: 10.1007/s12035-019-01781-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
The brain's primary circadian pacemaker, the suprachiasmatic nucleus (SCN), is required to translate day-length and circadian rhythms into neuronal, hormonal, and behavioral rhythms. Here, we identify the homeodomain transcription factor ventral anterior homeobox 1 (Vax1) as required for SCN development, vasoactive intestinal peptide expression, and SCN output. Previous work has shown that VAX1 is required for gonadotropin-releasing hormone (GnRH/LHRH) neuron development, a neuronal population controlling reproductive status. Surprisingly, the ectopic expression of a Gnrh-Cre allele (Gnrhcre) in the SCN confirmed the requirement of both VAX1 (Vax1flox/flox:Gnrhcre, Vax1Gnrh-cre) and sine oculis homeobox protein 6 (Six6flox/flox:Gnrhcre, Six6Gnrh-cre) in SCN function in adulthood. To dissociate the role of Vax1 and Six6 in GnRH neuron and SCN function, we used another Gnrh-cre allele that targets GnRH neurons, but not the SCN (Lhrhcre). Both Six6Lhrh-cre and Vax1Lhrh-cre were infertile, and in contrast to Vax1Gnrh-cre and Six6Gnrh-cre mice, Six6Lhrh-cre and Vax1Lhrh-cre had normal circadian behavior. Unexpectedly, ~ 1/4 of the Six6Gnrh-cre mice were unable to entrain to light, showing that ectopic expression of Gnrhcre impaired function of the retino-hypothalamic tract that relays light information to the brain. This study identifies VAX1, and confirms SIX6, as transcription factors required for SCN development and function and demonstrates the importance of understanding how ectopic CRE expression can impact the results.
Collapse
|
11
|
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
|
12
|
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
|
13
|
Pandolfi EC, Hoffmann HM, Schoeller EL, Gorman MR, Mellon PL. Haploinsufficiency of SIX3 Abolishes Male Reproductive Behavior Through Disrupted Olfactory Development, and Impairs Female Fertility Through Disrupted GnRH Neuron Migration. Mol Neurobiol 2018; 55:8709-8727. [PMID: 29589282 PMCID: PMC6156938 DOI: 10.1007/s12035-018-1013-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/16/2018] [Indexed: 12/17/2022]
Abstract
Mating behavior in males and females is dependent on olfactory cues processed through both the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). Signaling through the MOE is critical for the initiation of male mating behavior, and the loss of MOE signaling severely compromises this comportment. Here, we demonstrate that dosage of the homeodomain gene Six3 affects the degree of development of MOE but not the VNO. Anomalous MOE development in Six3 heterozygote mice leads to hyposmia, specifically disrupting male mounting behavior by impairing detection of volatile female estrus pheromones. Six3 is highly expressed in the MOE, main olfactory bulb (MOB), and hypothalamus; all regions essential in the proper migration of the gonadotropin-releasing hormone (GnRH) neurons, a key reproductive neuronal population that migrates along olfactory axons from the developing nose into the brain. Interestingly, we find that the reduction in Six3 expression in Six3 heterozygote mice compromises development of the MOE and MOB, resulting in mis-migration of GnRH neurons due to improper olfactory axon targeting. This reduction in the hypothalamic GnRH neuron population, by 45% in adulthood, leads to female subfertility, but does not impact male hormone levels, suggesting that male infertility is not related to GnRH neuron numbers, but exclusively linked to abnormal olfaction. We here determine that Six3 is haploinsufficient for MOE development, GnRH neuron migration, and fertility, and represents a novel candidate gene for Kallmann syndrome, a form of inherited infertility.
Collapse
Affiliation(s)
- Erica C Pandolfi
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hanne M Hoffmann
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Erica L Schoeller
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael R Gorman
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Psychology, 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, 9500 Gilman Drive, La Jolla, CA, 92093-0674, USA.
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
14
|
Weber A, Schwarz SC, Tost J, Trümbach D, Winter P, Busato F, Tacik P, Windhorst AC, Fagny M, Arzberger T, McLean C, van Swieten JC, Schwarz J, Vogt Weisenhorn D, Wurst W, Adhikary T, Dickson DW, Höglinger GU, Müller U. Epigenome-wide DNA methylation profiling in Progressive Supranuclear Palsy reveals major changes at DLX1. Nat Commun 2018; 9:2929. [PMID: 30050033 PMCID: PMC6062504 DOI: 10.1038/s41467-018-05325-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Genetic, epigenetic, and environmental factors contribute to the multifactorial disorder progressive supranuclear palsy (PSP). Here, we study epigenetic changes by genome-wide analysis of DNA from postmortem tissue of forebrains of patients and controls and detect significant (P < 0.05) methylation differences at 717 CpG sites in PSP vs. controls. Four-hundred fifty-one of these sites are associated with protein-coding genes. While differential methylation only affects a few sites in most genes, DLX1 is hypermethylated at multiple sites. Expression of an antisense transcript of DLX1, DLX1AS, is reduced in PSP brains. The amount of DLX1 protein is increased in gray matter of PSP forebrains. Pathway analysis suggests that DLX1 influences MAPT-encoded Tau protein. In a cell system, overexpression of DLX1 results in downregulation of MAPT while overexpression of DLX1AS causes upregulation of MAPT. Our observations suggest that altered DLX1 methylation and expression contribute to pathogenesis of PSP by influencing MAPT.
Collapse
Affiliation(s)
- Axel Weber
- Institute of Human Genetics, Justus-Liebig-Universität, Gießen, 35392, Germany.
| | - Sigrid C Schwarz
- Department of Neurology, Technische Universität München, Munich, 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
| | - Jörg Tost
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Evry, 91000, France
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Center München, Munich, 85764, Germany
| | - Pia Winter
- Institute of Human Genetics, Justus-Liebig-Universität, Gießen, 35392, Germany
| | - Florence Busato
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Evry, 91000, France
| | - Pawel Tacik
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, 53127, Germany
| | - Anita C Windhorst
- Institute of Medical Informatics, Justus-Liebig-Universität, Gießen, 35392, Germany
| | - Maud Fagny
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Evry, 91000, France
| | - Thomas Arzberger
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Department of Psychiatry, Ludwig-Maximilians-Universität, Munich, 81377, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Catriona McLean
- Alfred Anatomical Pathology and NNF, Victorian Brain Bank, Carlton, VIC, 3053, Australia
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Centre, Rotterdam, 3000, The Netherlands
| | - Johannes Schwarz
- Department of Neurology, Technische Universität München, Munich, 81377, Germany
| | - Daniela Vogt Weisenhorn
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Institute of Developmental Genetics, Helmholtz Center München, Munich, 85764, Germany
- Chair of Developmental Genetics, Technische Universität München-Weihenstephan, Neuherberg/Munich, 85764, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Institute of Developmental Genetics, Helmholtz Center München, Munich, 85764, Germany
- Chair of Developmental Genetics, Technische Universität München-Weihenstephan, Neuherberg/Munich, 85764, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, 81377, Germany
| | - Till Adhikary
- Institute for Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Marburg, 35043, Germany
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Günter U Höglinger
- Department of Neurology, Technische Universität München, Munich, 81377, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, 81377, Germany.
| | - Ulrich Müller
- Institute of Human Genetics, Justus-Liebig-Universität, Gießen, 35392, Germany.
| |
Collapse
|
15
|
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
|
16
|
GABAergic Interneuron Differentiation in the Basal Forebrain Is Mediated through Direct Regulation of Glutamic Acid Decarboxylase Isoforms by Dlx Homeobox Transcription Factors. J Neurosci 2017; 37:8816-8829. [PMID: 28821666 DOI: 10.1523/jneurosci.2125-16.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 06/25/2017] [Accepted: 08/03/2017] [Indexed: 01/09/2023] Open
Abstract
GABA is the key inhibitory neurotransmitter in the cortex but regulation of its synthesis during forebrain development is poorly understood. In the telencephalon, members of the distal-less (Dlx) homeobox gene family are expressed in, and regulate the development of, the basal ganglia primodia from which many GABAergic neurons originate and migrate to other forebrain regions. The Dlx1/Dlx2 double knock-out mice die at birth with abnormal cortical development, including loss of tangential migration of GABAergic inhibitory interneurons to the neocortex (Anderson et al., 1997a). We have discovered that specific promoter regulatory elements of glutamic acid decarboxylase isoforms (Gad1 and Gad2), which regulate GABA synthesis from the excitatory neurotransmitter glutamate, are direct transcriptional targets of both DLX1 and DLX2 homeoproteins in vivo Further gain- and loss-of-function studies in vitro and in vivo demonstrated that both DLX1 and DLX2 are necessary and sufficient for Gad gene expression. DLX1 and/or DLX2 activated the transcription of both Gad genes, and defects in Dlx function disrupted the differentiation of GABAergic interneurons with global reduction in GABA levels in the forebrains of the Dlx1/Dlx2 double knock-out mouse in vivo Identification of Gad genes as direct Dlx transcriptional targets is significant; it extends our understanding of Dlx gene function in the developing forebrain beyond the regulation of tangential interneuron migration to the differentiation of GABAergic interneurons arising from the basal telencephalon, and may help to unravel the pathogenesis of several developmental brain disorders.SIGNIFICANCE STATEMENT GABA is the major inhibitory neurotransmitter in the brain. We show that Dlx1/Dlx2 homeobox genes regulate GABA synthesis during forebrain development through direct activation of glutamic acid decarboxylase enzyme isoforms that convert glutamate to GABA. This discovery helps explain how Dlx mutations result in abnormal forebrain development, due to defective differentiation, in addition to the loss of tangential migration of GABAergic inhibitory interneurons to the neocortex. Reduced numbers or function of cortical GABAergic neurons may lead to hyperactivity states such as seizures (Cobos et al., 2005) or contribute to the pathogenesis of some autism spectrum disorders. GABAergic dysfunction in the basal ganglia could disrupt the learning and development of complex motor and cognitive behaviors (Rubenstein and Merzenich, 2003).
Collapse
|
17
|
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
|
18
|
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]
|
19
|
Stallings CE, Kapali J, Ellsworth BS. Mouse Models of Gonadotrope Development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:1-48. [PMID: 27697200 DOI: 10.1016/bs.pmbts.2016.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotrope is central to reproductive function. Gonadotropes develop in a systematic process dependent on signaling factors secreted from surrounding tissues and those produced within the pituitary gland itself. These signaling pathways are important for stimulating specific transcription factors that ultimately regulate the expression of genes and define gonadotrope identity. Proper gonadotrope development and ultimately gonadotrope function are essential for normal sexual maturation and fertility. Understanding the mechanisms governing differentiation programs of gonadotropes is important to improve treatment and molecular diagnoses for patients with gonadotrope abnormalities. Much of what is known about gonadotrope development has been elucidated from mouse models in which important factors contributing to gonadotrope development and function have been deleted, ectopically expressed, or modified. This chapter will focus on many of these mouse models and their contribution to our current understanding of gonadotrope development.
Collapse
Affiliation(s)
- C E Stallings
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - J Kapali
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - B S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
20
|
Deletion of Vax1 from Gonadotropin-Releasing Hormone (GnRH) Neurons Abolishes GnRH Expression and Leads to Hypogonadism and Infertility. J Neurosci 2016; 36:3506-18. [PMID: 27013679 DOI: 10.1523/jneurosci.2723-15.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Hypothalamic gonadotropin-releasing hormone (GnRH) neurons are at the apex of the hypothalamic-pituitary-gonadal axis that regulates mammalian fertility. Herein we demonstrate a critical role for the homeodomain transcription factor ventral anterior homeobox 1 (VAX1) in GnRH neuron maturation and show that Vax1 deletion from GnRH neurons leads to complete infertility in males and females. Specifically, global Vax1 knock-out embryos had normal numbers of GnRH neurons at 13 d of gestation, but no GnRH staining was detected by embryonic day 17. To identify the role of VAX1 specifically in GnRH neuron development,Vax1(flox)mice were generated and lineage tracing performed in Vax1(flox/flox):GnRH(cre):RosaLacZ mice. This identified VAX1 as essential for maintaining expression of Gnrh1 The absence of GnRH staining in adult Vax1(flox/flox):GnRH(cre)mice led to delayed puberty, hypogonadism, and infertility. To address the mechanism by which VAX1 maintains Gnrh1 transcription, the capacity of VAX1 to regulate Gnrh1 transcription was evaluated in the GnRH cell lines GN11 and GT1-7. As determined by luciferase and electrophoretic mobility shift assays, we found VAX1 to be a direct activator of the GnRH promoter through binding to four ATTA sites in the GnRH enhancer (E1) and proximal promoter (P), and able to compete with the homeoprotein SIX6 for occupation of the identified ATTA sites in the GnRH promoter. We conclude that VAX1 is expressed in GnRH neurons where it is required for GnRH neuron expression of GnRH and maintenance of fertility in mice. SIGNIFICANCE STATEMENT Infertility classified as idiopathic hypogonadotropic hypogonadism (IHH) is characterized by delayed or absent sexual maturation and low sex steroid levels due to alterations in neuroendocrine control of the hypothalamic-pituitary-gonadal axis. The incidence of IHH is 1-10 cases per 100,000 births. Although extensive efforts have been invested in identifying genes giving rise to IHH, >50% of cases have unknown genetic origins. We recently showed that haploinsufficiency of ventral anterior homeobox 1 (Vax1) leads to subfertility, making it a candidate in polygenic IHH. In this study, we investigate the mechanism by which VAX1 controls fertility finding that VAX1 is required for maintenance of Gnrh1 gene expression and deletion of Vax1 from GnRH neurons leads to complete infertility.
Collapse
|
21
|
Lund C, Pulli K, Yellapragada V, Giacobini P, Lundin K, Vuoristo S, Tuuri T, Noisa P, Raivio T. Development of Gonadotropin-Releasing Hormone-Secreting Neurons from Human Pluripotent Stem Cells. Stem Cell Reports 2016; 7:149-57. [PMID: 27426041 PMCID: PMC4982984 DOI: 10.1016/j.stemcr.2016.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 12/18/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons regulate human puberty and reproduction. Modeling their development and function in vitro would be of interest for both basic research and clinical translation. Here, we report a three-step protocol to differentiate human pluripotent stem cells (hPSCs) into GnRH-secreting neurons. Firstly, hPSCs were differentiated to FOXG1, EMX2, and PAX6 expressing anterior neural progenitor cells (NPCs) by dual SMAD inhibition. Secondly, NPCs were treated for 10 days with FGF8, which is a key ligand implicated in GnRH neuron ontogeny, and finally, the cells were matured with Notch inhibitor to bipolar TUJ1-positive neurons that robustly expressed GNRH1 and secreted GnRH decapeptide into the culture medium. The protocol was reproducible both in human embryonic stem cells and induced pluripotent stem cells, and thus provides a translational tool for investigating the mechanisms of human puberty and its disorders. GnRH neurons regulate puberty and reproduction We generated GnRH-expressing and secreting neurons from hPSCs These neurons can be used to study diseases affecting the reproductive system
Collapse
Affiliation(s)
- Carina Lund
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki 00014, Finland
| | - Kristiina Pulli
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki 00014, Finland
| | - Venkatram Yellapragada
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki 00014, Finland
| | - Paolo Giacobini
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, Unité 1172, 59045 Lille Cedex, France; School of Medicine, University of Lille, Lille 59000, France
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, HUCH, Helsinki 00029, Finland
| | - Sanna Vuoristo
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki 00014, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, HUCH, Helsinki 00029, Finland
| | - Parinya Noisa
- School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; Faculty of Medicine/Physiology, University of Helsinki, Biomedicum Helsinki, PO Box 63 (Haartmaninkatu 8), Helsinki 00014, Finland
| | - Taneli Raivio
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki 00014, Finland; Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), Helsinki 00029, Finland; Faculty of Medicine/Physiology, University of Helsinki, Biomedicum Helsinki, PO Box 63 (Haartmaninkatu 8), Helsinki 00014, Finland.
| |
Collapse
|
22
|
A Novel Gonadotropin-Releasing Hormone 1 (Gnrh1) Enhancer-Derived Noncoding RNA Regulates Gnrh1 Gene Expression in GnRH Neuronal Cell Models. PLoS One 2016; 11:e0158597. [PMID: 27389022 PMCID: PMC4936741 DOI: 10.1371/journal.pone.0158597] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/18/2016] [Indexed: 12/22/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH), a neuropeptide released from a small population of neurons in the hypothalamus, is the central mediator of the hypothalamic-pituitary-gonadal axis, and is required for normal reproductive development and function. Evolutionarily conserved regulatory elements in the mouse, rat, and human Gnrh1 gene include three enhancers and the proximal promoter, which confer Gnrh1 gene expression specifically in GnRH neurons. In immortalized mouse hypothalamic GnRH (GT1-7) neurons, which show pulsatile GnRH release in culture, RNA sequencing and RT-qPCR revealed that expression of a novel long noncoding RNA at Gnrh1 enhancer 1 correlates with high levels of GnRH mRNA expression. In GT1-7 neurons, which contain a transgene carrying 3 kb of the rat Gnrh1 regulatory region, both the mouse and rat Gnrh1 enhancer-derived noncoding RNAs (GnRH-E1 RNAs) are expressed. We investigated the characteristics and function of the endogenous mouse GnRH-E1 RNA. Strand-specific RT-PCR analysis of GnRH-E1 RNA in GT1-7 cells revealed GnRH-E1 RNAs that are transcribed in the sense and antisense directions from distinct 5’ start sites, are 3’ polyadenylated, and are over 2 kb in length. These RNAs are localized in the nucleus and have a half-life of over 8 hours. In GT1-7 neurons, siRNA knockdown of mouse GnRH-E1 RNA resulted in a significant decrease in the expression of the Gnrh1 primary transcript and Gnrh1 mRNA. Over-expression of either the sense or antisense mouse GnRH-E1 RNA in immature, migratory GnRH (GN11) neurons, which do not express either GnRH-E1 RNA or GnRH mRNA, induced the transcriptional activity of co-transfected rat Gnrh1 gene regulatory elements, where the induction requires the presence of the rat Gnrh1 promoter. Together, these data indicate that GnRH-E1 RNA is an inducer of Gnrh1 gene expression. GnRH-E1 RNA may play an important role in the development and maturation of GnRH neurons.
Collapse
|
23
|
Tran DQ, Ramos EH, Belsham DD. Induction of Gnrh mRNA expression by the ω-3 polyunsaturated fatty acid docosahexaenoic acid and the saturated fatty acid palmitate in a GnRH-synthesizing neuronal cell model, mHypoA-GnRH/GFP. Mol Cell Endocrinol 2016; 426:125-35. [PMID: 26923440 DOI: 10.1016/j.mce.2016.02.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 12/18/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons coordinate reproduction. However, whether GnRH neurons directly sense free fatty acids (FFAs) is unknown. We investigated the individual effects of the FFAs docosahexaenoic acid (DHA), palmitate, palmitoleate, and oleate (100 μM each) on Gnrh mRNA expression in the mHypoA-GnRH/GFP neuronal cell model. We report that 2 h exposure to palmitate or DHA increases Gnrh transcription. Using the inhibitors AH7614, K252c, U0126, wortmannin, and LY294002, we demonstrate that the effect of DHA is mediated through GPR120 to downstream PKC/MAPK and PI3K signaling. Our results indicate that the effect of palmitate may depend on palmitoyl-coA synthesis and PI3K signaling. Finally, we demonstrate that both DHA and palmitate increase Gnrh enhancer-derived RNA levels. Overall, these studies provide evidence that GnRH neurons directly sense FFAs. This will advance our understanding of the mechanisms underlying FFA sensing in the brain and provides insight into the links between nutrition and reproductive function.
Collapse
Affiliation(s)
- Dean Q Tran
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ernesto H Ramos
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Denise D Belsham
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
24
|
Messina A, Langlet F, Chachlaki K, Roa J, Rasika S, Jouy N, Gallet S, Gaytan F, Parkash J, Tena-Sempere M, Giacobini P, Prevot V. A microRNA switch regulates the rise in hypothalamic GnRH production before puberty. Nat Neurosci 2016; 19:835-44. [DOI: 10.1038/nn.4298] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
|
25
|
Hoffmann HM, Mellon PL. A small population of hypothalamic neurons govern fertility: the critical role of VAX1 in GnRH neuron development and fertility maintenance. NEUROSCIENCE COMMUNICATIONS 2016; 2:e1373. [PMID: 28164172 PMCID: PMC5287408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fertility depends on the correct maturation and function of approximately 800 gonadotropin-releasing hormone (GnRH) neurons in the brain. GnRH neurons are at the apex of the hypothalamic-pituitary-gonadal axis that regulates fertility. In adulthood, GnRH neurons are scattered throughout the anterior hypothalamic area and project to the median eminence, where GnRH is released into the portal vasculature to stimulate release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the pituitary. LH and FSH then regulate gonadal steroidogenesis and gametogenesis. Absence of GnRH neurons or inappropriate GnRH release leads to infertility. Despite the critical role of GnRH neurons in fertility, we still have a limited understanding of the genes responsible for proper GnRH neuron development and function in adulthood. GnRH neurons originate in the olfactory placode then migrate into the brain. Homeodomain transcription factors expressed within GnRH neurons or along their migratory path are candidate genes for inherited infertility. Using a combined in vitro and in vivo approach, we have identified Ventral Anterior Homeobox 1 (Vax1) as a novel homeodomain transcription factor responsible for GnRH neuron maturation and fertility. GnRH neuron counts in Vax1 knock-out embryos revealed Vax1 to be required for the presence of GnRH-expressing cells at embryonic day 17.5 (E17.5), but not at E13.5. To localize the effects of Vax1 on fertility, we generated Vax1flox mice and crossed them with Gnrhcre mice to specifically delete Vax1 within GnRH neurons. GnRH staining in Vax1flox/flox:GnRHcre mice show a total absence of GnRH expression in the adult. We performed lineage tracing in Vax1flox/flox:GnRHcre:RosaLacZ mice which proved GnRH neurons to be alive, but incapable of expressing GnRH. The absence of GnRH leads to delayed puberty, hypogonadism and complete infertility in both sexes. Finally, using the immortalized model GnRH neuron cell lines, GN11 and GT1-7, we show that VAX1 is a direct regulator of Gnrh1 transcription by binding key ATTA sites within the Gnrh1 promoter. This study identifies VAX1 as a key transcription factor regulating GnRH expression and establishes VAX1 as a novel candidate gene implicated in heritable infertility.
Collapse
|
26
|
Xie H, Hoffmann HM, Meadows JD, Mayo SL, Trang C, Leming SS, Maruggi C, Davis SW, Larder R, Mellon PL. Homeodomain Proteins SIX3 and SIX6 Regulate Gonadotrope-specific Genes During Pituitary Development. Mol Endocrinol 2015; 29:842-55. [PMID: 25915183 PMCID: PMC4447639 DOI: 10.1210/me.2014-1279] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 04/20/2015] [Indexed: 12/15/2022] Open
Abstract
Sine oculis-related homeobox 3 (SIX3) and SIX6, 2 closely related homeodomain transcription factors, are involved in development of the mammalian neuroendocrine system and mutations of Six6 adversely affect fertility in mice. We show that both small interfering RNA knockdown in gonadotrope cell lines and knockout of Six6 in both embryonic and adult male mice (Six6 knockout) support roles for SIX3 and SIX6 in transcriptional regulation in gonadotrope gene expression and that SIX3 and SIX6 can functionally compensate for each other. Six3 and Six6 expression patterns in gonadotrope cell lines reflect the timing of the expression of pituitary markers they regulate. Six3 is expressed in an immature gonadotrope cell line and represses transcription of the early lineage-specific pituitary genes, GnRH receptor (GnRHR) and the common α-subunit (Cga), whereas Six6 is expressed in a mature gonadotrope cell line and represses the specific β-subunits of LH and FSH (LHb and FSHb) that are expressed later in development. We show that SIX6 repression requires interaction with transducin-like enhancer of split corepressor proteins and competition for DNA-binding sites with the transcriptional activator pituitary homeobox 1. Our studies also suggest that estradiol and circadian rhythm regulate pituitary expression of Six6 and Six3 in adult females but not in males. In summary, SIX3 and SIX6 play distinct but compensatory roles in regulating transcription of gonadotrope-specific genes as gonadotrope cells differentiate.
Collapse
Affiliation(s)
- Huimin Xie
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Hanne M Hoffmann
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Jason D Meadows
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Susan L Mayo
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Crystal Trang
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Sunamita S Leming
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Chiara Maruggi
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Shannon W Davis
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Rachel Larder
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Pamela L Mellon
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
27
|
Garaffo G, Conte D, Provero P, Tomaiuolo D, Luo Z, Pinciroli P, Peano C, D'Atri I, Gitton Y, Etzion T, Gothilf Y, Gays D, Santoro MM, Merlo GR. The Dlx5 and Foxg1 transcription factors, linked via miRNA-9 and -200, are required for the development of the olfactory and GnRH system. Mol Cell Neurosci 2015; 68:103-19. [PMID: 25937343 PMCID: PMC4604252 DOI: 10.1016/j.mcn.2015.04.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 04/20/2015] [Accepted: 04/29/2015] [Indexed: 01/26/2023] Open
Abstract
During neuronal development and maturation, microRNAs (miRs) play diverse functions ranging from early patterning, proliferation and commitment to differentiation, survival, homeostasis, activity and plasticity of more mature and adult neurons. The role of miRs in the differentiation of olfactory receptor neurons (ORNs) is emerging from the conditional inactivation of Dicer in immature ORN, and the depletion of all mature miRs in this system. Here, we identify specific miRs involved in olfactory development, by focusing on mice null for Dlx5, a homeogene essential for both ORN differentiation and axon guidance and connectivity. Analysis of miR expression in Dlx5−/− olfactory epithelium pointed to reduced levels of miR-9, miR-376a and four miRs of the -200 class in the absence of Dlx5. To functionally examine the role of these miRs, we depleted miR-9 and miR-200 class in reporter zebrafish embryos and observed delayed ORN differentiation, altered axonal trajectory/targeting, and altered genesis and position of olfactory-associated GnRH neurons, i.e. a phenotype known as Kallmann syndrome in humans. miR-9 and miR-200-class negatively control Foxg1 mRNA, a fork-head transcription factor essential for development of the olfactory epithelium and of the forebrain, known to maintain progenitors in a stem state. Increased levels of z-foxg1 mRNA resulted in delayed ORN differentiation and altered axon trajectory, in zebrafish embryos. This work describes for the first time the role of specific miR (-9 and -200) in olfactory/GnRH development, and uncovers a Dlx5–Foxg1 regulation whose alteration affects receptor neuron differentiation, axonal targeting, GnRH neuron development, the hallmarks of the Kallmann syndrome. Dlx5 controls the expressions of miR9 and miR-200, which target the Foxg1 mRNA miR-9 and -200 are needed for olfactory neurons differentiation and axon extension miR-9 and -200 are required for the genesis and position of GnRH neurons. Altered expression of miR-9 and -200 might contribute to the Kallmann disease.
Collapse
Affiliation(s)
- Giulia Garaffo
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Daniele Conte
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Paolo Provero
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Daniela Tomaiuolo
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Zheng Luo
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Patrizia Pinciroli
- Doctorate School in Molecular Medicine, Dept. Medical Biotechnology Translational Medicine (BIOMETRA), University of Milano, Italy
| | - Clelia Peano
- Inst. of Biomedical Technology, National Research Council, ITB-CNR Segrate (MI) Italy
| | - Ilaria D'Atri
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Yorick Gitton
- UMR7221 CNRS/MNHN - Evolution des régulations endocriniennes - Paris, France
| | - Talya Etzion
- Dept. Neurobiology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; VIB, Vesalius Research Center, KU Leuven, Belgium
| | - Yoav Gothilf
- Dept. Neurobiology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; VIB, Vesalius Research Center, KU Leuven, Belgium
| | - Dafne Gays
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Massimo M Santoro
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy; Dept. Neurobiology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; VIB, Vesalius Research Center, KU Leuven, Belgium
| | - Giorgio R Merlo
- Dept. Molecular Biotechnology and Health Sciences, University of Torino, Italy.
| |
Collapse
|
28
|
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
|
29
|
Abstract
Cranial sensory placodes derive from discrete patches of the head ectoderm and give rise to numerous sensory structures. During gastrulation, a specialized "neural border zone" forms around the neural plate in response to interactions between the neural and nonneural ectoderm and signals from adjacent mesodermal and/or endodermal tissues. This zone subsequently gives rise to two distinct precursor populations of the peripheral nervous system: the neural crest and the preplacodal ectoderm (PPE). The PPE is a common field from which all cranial sensory placodes arise (adenohypophyseal, olfactory, lens, trigeminal, epibranchial, otic). Members of the Six family of transcription factors are major regulators of PPE specification, in partnership with cofactor proteins such as Eya. Six gene activity also maintains tissue boundaries between the PPE, neural crest, and epidermis by repressing genes that specify the fates of those adjacent ectodermally derived domains. As the embryo acquires anterior-posterior identity, the PPE becomes transcriptionally regionalized, and it subsequently becomes subdivided into specific placodes with distinct developmental fates in response to signaling from adjacent tissues. Each placode is characterized by a unique transcriptional program that leads to the differentiation of highly specialized cells, such as neurosecretory cells, sensory receptor cells, chemosensory neurons, peripheral glia, and supporting cells. In this review, we summarize the transcriptional and signaling factors that regulate key steps of placode development, influence subsequent sensory neuron specification, and discuss what is known about mutations in some of the essential PPE genes that underlie human congenital syndromes.
Collapse
Affiliation(s)
- Sally A Moody
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA; George Washington University Institute for Neuroscience, Washington, DC, USA.
| | - Anthony-Samuel LaMantia
- George Washington University Institute for Neuroscience, Washington, DC, USA; Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
30
|
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
|
31
|
Vastagh C, Schwirtlich M, Kwakowsky A, Erdélyi F, Margolis FL, Yanagawa Y, Katarova Z, Szabó G. The spatiotemporal segregation of GAD forms defines distinct GABA signaling functions in the developing mouse olfactory system and provides novel insights into the origin and migration of GnRH neurons. Dev Neurobiol 2014; 75:249-70. [PMID: 25125027 DOI: 10.1002/dneu.22222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/18/2014] [Accepted: 08/06/2014] [Indexed: 01/26/2023]
Abstract
Gamma-aminobutyric acid (GABA) has a dual role as an inhibitory neurotransmitter in the adult central nervous system (CNS) and as a signaling molecule exerting largely excitatory actions during development. The rate-limiting step of GABA synthesis is catalyzed by two glutamic acid decarboxylase isoforms GAD65 and GAD67 coexpressed in the GABAergic neurons of the CNS. Here we report that the two GADs show virtually nonoverlapping expression patterns consistent with distinct roles in the developing peripheral olfactory system. GAD65 is expressed exclusively in undifferentiated neuronal progenitors confined to the proliferative zones of the sensory vomeronasal and olfactory epithelia In contrast GAD67 is expressed in a subregion of the nonsensory epithelium/vomeronasal organ epithelium containing the putative Gonadotropin-releasing hormone (GnRH) progenitors and GnRH neurons migrating from this region through the frontonasal mesenchyme into the basal forebrain. Only GAD67+, but not GAD65+ cells accumulate detectable GABA. We further demonstrate that GAD67 and its embryonic splice variant embryonic GAD (EGAD) concomitant with GnRH are dynamically regulated during GnRH neuronal migration in vivo and in two immortalized cell lines representing migratory (GN11) and postmigratory (GT1-7) stage GnRH neurons, respectively. Analysis of GAD65/67 single and double knock-out embryos revealed that the two GADs play complementary (inhibitory) roles in GnRH migration ultimately modulating the speed and/or direction of GnRH migration. Our results also suggest that GAD65 and GAD67/EGAD characterized by distinct subcellular localization and kinetics have disparate functions during olfactory system development mediating proliferative and migratory responses putatively through specific subcellular GABA pools.
Collapse
Affiliation(s)
- Csaba Vastagh
- Division of Medical Gene Technology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Duval N, Daubas P, Bourcier de Carbon C, St Cloment C, Tinevez JY, Lopes M, Ribes V, Robert B. Msx1 and Msx2 act as essential activators of Atoh1 expression in the murine spinal cord. Development 2014; 141:1726-36. [PMID: 24715462 DOI: 10.1242/dev.099002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dorsal spinal neurogenesis is orchestrated by the combined action of signals secreted from the roof plate organizer and a downstream transcriptional cascade. Within this cascade, Msx1 and Msx2, two homeodomain transcription factors (TFs), are induced earlier than bHLH neuralizing TFs. Whereas bHLH TFs have been shown to specify neuronal cell fate, the function of Msx genes remains poorly defined. We describe dramatic alterations of neuronal patterning in Msx1/Msx2 double-mutant mouse embryos. The most dorsal spinal progenitor pool fails to express the bHLH neuralizing TF Atoh1, which results in a lack of Lhx2-positive and Barhl2-positive dI1 interneurons. Neurog1 and Ascl1 expression territories are dorsalized, leading to ectopic dorsal differentiation of dI2 and dI3 interneurons. In proportion, the amount of Neurog1-expressing progenitors appears unaffected, whereas the number of Ascl1-positive cells is increased. These defects occur while BMP signaling is still active in the Msx1/Msx2 mutant embryos. Cell lineage analysis and co-immunolabeling demonstrate that Atoh1-positive cells derive from progenitors expressing both Msx1 and Msx2. In vitro, Msx1 and Msx2 proteins activate Atoh1 transcription by specifically interacting with several homeodomain binding sites in the Atoh1 3' enhancer. In vivo, Msx1 and Msx2 are required for Atoh1 3' enhancer activity and ChIP experiments confirm Msx1 binding to this regulatory sequence. These data support a novel function of Msx1 and Msx2 as transcriptional activators. Our study provides new insights into the transcriptional control of spinal cord patterning by BMP signaling, with Msx1 and Msx2 acting upstream of Atoh1.
Collapse
Affiliation(s)
- Nathalie Duval
- Institut Pasteur, Morphogenesis Molecular Genetics, CNRS URA 2578, 75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
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
|
34
|
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
|
35
|
A network of transcription factors operates during early tooth morphogenesis. Mol Cell Biol 2013; 33:3099-112. [PMID: 23754753 DOI: 10.1128/mcb.00524-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Improving the knowledge of disease-causing genes is a unique challenge in human health. Although it is known that genes causing similar diseases tend to lie close to one another in a network of protein-protein or functional interactions, the identification of these protein-protein networks is difficult to unravel. Here, we show that Msx1, Snail, Lhx6, Lhx8, Sp3, and Lef1 interact in vitro and in vivo, revealing the existence of a novel context-specific protein network. These proteins are all expressed in the neural crest-derived dental mesenchyme and cause tooth agenesis disorder when mutated in mouse and/or human. We also identified an in vivo direct target for Msx1 function, the cyclin D-dependent kinase (CDK) inhibitor p19(ink4d), whose transcription is differentially modulated by the protein network. Considering the important role of p19(ink4d) as a cell cycle regulator, these results provide evidence for the first time of the unique plasticity of the Msx1-dependent network of proteins in conferring differential transcriptional output and in controlling the cell cycle through the regulation of a cyclin D-dependent kinase inhibitor. Collectively, these data reveal a novel protein network operating in the neural crest-derived dental mesenchyme that is relevant for many other areas of developmental and evolutionary biology.
Collapse
|
36
|
Glidewell-Kenney CA, Shao PP, Iyer AK, Grove AMH, Meadows JD, Mellon PL. Neurokinin B causes acute GnRH secretion and repression of GnRH transcription in GT1-7 GnRH neurons. Mol Endocrinol 2013; 27:437-54. [PMID: 23393128 DOI: 10.1210/me.2012-1271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Genetic studies in human patients with idiopathic hypogonadotropic hypogonadism (IHH) identified mutations in the genes that encode neurokinin B (NKB) and the neurokinin 3 receptor (NK3R). However, determining the mechanism whereby NKB regulates gonadotropin secretion has been difficult because of conflicting results from in vivo studies investigating the luteinizing hormone (LH) response to senktide, a NK3R agonist. NK3R is expressed in a subset of GnRH neurons and in kisspeptin neurons that are known to regulate GnRH secretion. Thus, one potential source of inconsistency is that NKB could produce opposing direct and indirect effects on GnRH secretion. Here, we employ the GT1-7 cell model to elucidate the direct effects of NKB on GnRH neuron function. We find that GT1-7 cells express NK3R and respond to acute senktide treatment with c-Fos induction and increased GnRH secretion. In contrast, long-term senktide treatment decreased GnRH secretion. Next, we focus on the examination of the mechanism underlying the long-term decrease in secretion and determine that senktide treatment represses transcription of GnRH. We further show that this repression of GnRH transcription may involve enhanced c-Fos protein binding at novel activator protein-1 (AP-1) half-sites identified in enhancer 1 and the promoter, as well as chromatin remodeling at the promoter of the GnRH gene. These data indicate that NKB could directly regulate secretion from NK3R-expressing GnRH neurons. Furthermore, whether the response is inhibitory or stimulatory toward GnRH secretion could depend on the history or length of exposure to NKB because of a repressive effect on GnRH transcription.
Collapse
Affiliation(s)
- Christine A Glidewell-Kenney
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | | | | | | | | | | |
Collapse
|
37
|
Xie H, Cherrington BD, Meadows JD, Witham EA, Mellon PL. Msx1 homeodomain protein represses the αGSU and GnRH receptor genes during gonadotrope development. Mol Endocrinol 2013; 27:422-36. [PMID: 23371388 DOI: 10.1210/me.2012-1289] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple homeodomain transcription factors are crucial for pituitary organogenesis and cellular differentiation. A homeodomain repressor, Msx1, is expressed from the ventral aspect of the developing anterior pituitary and implicated in gonadotrope differentiation. Here, we find that Msx1 represses transcription of lineage-specific pituitary genes such as the common α-glycoprotein subunit (αGSU) and GnRH receptor (GnRHR) promoters in the mouse gonadotrope-derived cell lines, αT3-1 and LβT2. Repression of the mouse GnRHR promoter by Msx1 is mediated through a consensus-binding motif in the downstream activin regulatory element (DARE). Truncation and mutation analyses of the human αGSU promoter map Msx1 repression to a site at -114, located at the junctional regulatory element (JRE). Dlx activators are closely related to the Msx repressors, acting through the same elements, and Dlx3 and Dlx2 act as transcriptional activators for GnRHR and αGSU, respectively. Small interfering RNA knockdown of Msx1 in αT3-1 cells increases endogenous αGSU and GnRHR mRNA expression. Msx1 gene expression reaches its maximal expression at the rostral edge at e13.5. The subsequent decline in Msx1 expression specifically coincides with the onset of expression of both αGSU and GnRHR. The expression levels of both αGSU and GnRHR in Msx1-null mice at e18.5 are higher compared with wild type, further confirming a role for Msx1 in the repression of αGSU and GnRHR. In summary, Msx1 functions as a negative regulator early in pituitary development by repressing the gonadotrope-specific αGSU and GnRHR genes, but a temporal decline in Msx1 expression alleviates this repression allowing induction of GnRHR and αGSU, thus serving to time the onset of gonadotrope-specific gene program.
Collapse
Affiliation(s)
- Huimin Xie
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, CA 92093-0674, USA
| | | | | | | | | |
Collapse
|
38
|
Analysis of genome-wide structure, diversity and fine mapping of Mendelian traits in traditional and village chickens. Heredity (Edinb) 2012; 109:6-18. [PMID: 22395157 DOI: 10.1038/hdy.2012.9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Extensive phenotypic variation is a common feature among village chickens found throughout much of the developing world, and in traditional chicken breeds that have been artificially selected for traits such as plumage variety. We present here an assessment of traditional and village chicken populations, for fine mapping of Mendelian traits using genome-wide single-nucleotide polymorphism (SNP) genotyping while providing information on their genetic structure and diversity. Bayesian clustering analysis reveals two main genetic backgrounds in traditional breeds, Kenyan, Ethiopian and Chilean village chickens. Analysis of linkage disequilibrium (LD) reveals useful LD (r(2) ≥ 0.3) in both traditional and village chickens at pairwise marker distances of ~10 Kb; while haplotype block analysis indicates a median block size of 11-12 Kb. Association mapping yielded refined mapping intervals for duplex comb (Gga 2:38.55-38.89 Mb) and rose comb (Gga 7:18.41-22.09 Mb) phenotypes in traditional breeds. Combined mapping information from traditional breeds and Chilean village chicken allows the oocyan phenotype to be fine mapped to two small regions (Gga 1:67.25-67.28 Mb, Gga 1:67.28-67.32 Mb) totalling ~75 Kb. Mapping the unmapped earlobe pigmentation phenotype supports previous findings that the trait is sex-linked and polygenic. A critical assessment of the number of SNPs required to map simple traits indicate that between 90 and 110K SNPs are required for full genome-wide analysis of haplotype block structure/ancestry, and for association mapping in both traditional and village chickens. Our results demonstrate the importance and uniqueness of phenotypic diversity and genetic structure of traditional chicken breeds for fine-scale mapping of Mendelian traits in the species, with village chicken populations providing further opportunities to enhance mapping resolutions.
Collapse
|
39
|
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
|
40
|
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
|
41
|
Neural crest and ectodermal cells intermix in the nasal placode to give rise to GnRH-1 neurons, sensory neurons, and olfactory ensheathing cells. J Neurosci 2011; 31:6915-27. [PMID: 21543621 DOI: 10.1523/jneurosci.6087-10.2011] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The origin of GnRH-1 cells and olfactory ensheathing cells has been controversial. Genetic Cre-lox lineage tracing of the neural crest (NC) versus ectodermal contribution to the developing nasal placode was performed using two complementary mouse models, the NC-specific Wnt1Cre mouse line and an ectodermal-specific Crect mouse line. Using these lines we prove that the NC give rise to the olfactory ensheathing cells and subpopulations of GnRH-1 neurons, olfactory and vomeronasal cells. These data demonstrate that Schwann cells and olfactory ensheathing cells share a common developmental origin. Furthermore, the results indicate that certain conditions that impact olfaction and sexual development, such as Kallmann syndrome, may be in part neurocristopathies.
Collapse
|
42
|
Beneduzzi D, Iyer AK, Trarbach EB, Silveira-Neto AP, Silveira LG, Tusset C, Yip K, Mendonça BB, Mellon PL, Latronico AC. Mutational analysis of the necdin gene in patients with congenital isolated hypogonadotropic hypogonadism. Eur J Endocrinol 2011; 165:145-50. [PMID: 21543378 PMCID: PMC3203643 DOI: 10.1530/eje-11-0199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CONTEXT Necdin activates GNRH gene expression and is fundamental for the development, migration, and axonal extension of murine GNRH neurons. In humans, necdin plays a potential role in the hypogonadotropic hypogonadism phenotype in patients with Prader-Willi syndrome. AIM To investigate necdin gene (NDN) variants in patients with isolated hypogonadotropic hypogonadism (IHH). PATIENTS AND METHODS We studied 160 Brazilian patients with IHH, which includes 92 with Kallmann syndrome and 68 with normosmic IHH. Genomic DNA was extracted and the single NDN exon was amplified and sequenced. To measure GNRH transcriptional activity, luciferase reporter plasmids containing GNRH regulatory regions were transiently transfected into GT1-7 cells in the presence and absence of overexpressed wild-type or mutant necdin. RESULTS A heterozygous variant of necdin, p.V318A, was identified in a 23-year-old male with Kallmann syndrome. The p.V318A was also present in affected aunt and his father and was absent in 100 Brazilian control subjects. Previous FGFR1 gene analysis revealed a missense mutation (p.P366L) in this family. Functional studies revealed a minor difference in the activation of GNRH transcription by mutant protein compared with wild type in that a significant impairment of the necdin protein activity threshold was observed. CONCLUSION A rare variant of necdin (p.V318A) was described in a family with Kallmann syndrome associated with a FGFR1 mutation. Familial segregation and in vitro analysis suggested that this non-synonymous variant did not have a direct causative role in the hypogonadism phenotype. NDN mutations are not a frequent cause of congenital IHH.
Collapse
Affiliation(s)
- Daiane Beneduzzi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, Avenida Doutor Eneas de Carvalho Aguiar 155, 2° Andar, Bloco 6, CEP 05403-900 São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
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 DOI: 10.1210/me.2010-0156] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|
47
|
Sato S, Ikeda K, Shioi G, Ochi H, Ogino H, Yajima H, Kawakami K. Conserved expression of mouse Six1 in the pre-placodal region (PPR) and identification of an enhancer for the rostral PPR. Dev Biol 2010; 344:158-71. [PMID: 20471971 DOI: 10.1016/j.ydbio.2010.04.029] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 04/24/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
Abstract
All cranial sensory organs and sensory neurons of vertebrates develop from cranial placodes. In chick, amphibians and zebrafish, all placodes originate from a common precursor domain, the pre-placodal region (PPR), marked by the expression of Six1/4 and Eya1/2. However, the PPR has never been described in mammals and the mechanism involved in the formation of PPR is poorly defined. Here, we report the expression of Six1 in the horseshoe-shaped mouse ectoderm surrounding the anterior neural plate in a pattern broadly similar to that of non-mammalian vertebrates. To elucidate the identity of Six1-positive mouse ectoderm, we searched for enhancers responsible for Six1 expression by in vivo enhancer assays. One conserved non-coding sequence, Six1-14, showed specific enhancer activity in the rostral PPR of chick and Xenopus and in the mouse ectoderm. These results strongly suggest the presence of PPR in mouse and that it is conserved in vertebrates. Moreover, we show the importance of the homeodomain protein-binding sites of Six1-14, the Six1 rostral PPR enhancer, for enhancer activity, and that Dlx5, Msx1 and Pax7 are candidate binding factors that regulate the level and area of Six1 expression, and thereby the location of the PPR. Our findings provide critical information and tools to elucidate the molecular mechanism of early sensory development and have implications for the development of sensory precursor/stem cells.
Collapse
Affiliation(s)
- Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Necdin promotes tangential migration of neocortical interneurons from basal forebrain. J Neurosci 2010; 30:3709-14. [PMID: 20220004 DOI: 10.1523/jneurosci.5797-09.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Necdin is a pleiotropic protein that promotes neuronal differentiation and survival. In mammals, the necdin gene on the maternal chromosome is silenced by genomic imprinting, and only the paternal necdin gene is expressed in virtually all postmitotic neurons. Necdin forms a complex with the homeodomain protein Dlx2 to enhance its transcriptional activity. Dlx2 plays a major role in controlling tangential migration of GABAergic interneurons from the basal forebrain to the neocortex. Here, we examined whether Dlx2-expressing interneurons migrate properly in vivo in mutant mice lacking the paternal necdin gene. In necdin-deficient mice at birth, the population of Dlx2-expressing cells significantly decreased in the neocortex but increased in the preoptic area. DiI-labeled cell migration assay using organotypic forebrain slice cultures revealed that the number of cells migrating from the medial ganglionic eminence into the neocortex was significantly reduced in necdin-deficient embryos. Furthermore, necdin-deficient mice had a decreased population of neocortical GABA-containing neurons and were highly susceptible to pentylenetetrazole-induced seizures. These results suggest that necdin promotes tangential migration of neocortical GABAergic interneurons during mammalian forebrain development.
Collapse
|
49
|
Schlosser G. Making senses development of vertebrate cranial placodes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 283:129-234. [PMID: 20801420 DOI: 10.1016/s1937-6448(10)83004-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cranial placodes (which include the adenohypophyseal, olfactory, lens, otic, lateral line, profundal/trigeminal, and epibranchial placodes) give rise to many sense organs and ganglia of the vertebrate head. Recent evidence suggests that all cranial placodes may be developmentally related structures, which originate from a common panplacodal primordium at neural plate stages and use similar regulatory mechanisms to control developmental processes shared between different placodes such as neurogenesis and morphogenetic movements. After providing a brief overview of placodal diversity, the present review summarizes current evidence for the existence of a panplacodal primordium and discusses the central role of transcription factors Six1 and Eya1 in the regulation of processes shared between different placodes. Upstream signaling events and transcription factors involved in early embryonic induction and specification of the panplacodal primordium are discussed next. I then review how individual placodes arise from the panplacodal primordium and present a model of multistep placode induction. Finally, I briefly summarize recent advances concerning how placodal neurons and sensory cells are specified, and how morphogenesis of placodes (including delamination and migration of placode-derived cells and invagination) is controlled.
Collapse
Affiliation(s)
- Gerhard Schlosser
- Zoology, School of Natural Sciences & Martin Ryan Institute, National University of Ireland, Galway, Ireland
| |
Collapse
|
50
|
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] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|