51
|
Garcia-Moreno SA, Plebanek MP, Capel B. Epigenetic regulation of male fate commitment from an initially bipotential system. Mol Cell Endocrinol 2018; 468:19-30. [PMID: 29410272 PMCID: PMC6084468 DOI: 10.1016/j.mce.2018.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/21/2022]
Abstract
A fundamental goal in biology is to understand how distinct cell types containing the same genetic information arise from a single stem cell throughout development. Sex determination is a key developmental process that requires a unidirectional commitment of an initially bipotential gonad towards either the male or female fate. This makes sex determination a unique model to study cell fate commitment and differentiation in vivo. We have focused this review on the accumulating evidence that epigenetic mechanisms contribute to the bipotential state of the fetal gonad and to the regulation of chromatin accessibility during and immediately downstream of the primary sex-determining switch that establishes the male fate.
Collapse
Affiliation(s)
| | | | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
52
|
Carré GA, Siggers P, Xipolita M, Brindle P, Lutz B, Wells S, Greenfield A. Loss of p300 and CBP disrupts histone acetylation at the mouse Sry promoter and causes XY gonadal sex reversal. Hum Mol Genet 2018; 27:190-198. [PMID: 29145650 PMCID: PMC5886154 DOI: 10.1093/hmg/ddx398] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023] Open
Abstract
CREB-binding protein (CBP, CREBBP, KAT3A) and its closely related paralogue p300 (EP300, KAT3B), together termed p300/CBP, are histone/lysine acetyl-transferases that control gene expression by modifying chromatin-associated proteins. Here, we report roles for both of these chromatin-modifying enzymes in mouse sex determination, the process by which the embryonic gonad develops into a testis or an ovary. By targeting gene ablation to embryonic gonadal somatic cells using an inducible Cre line, we show that gonads lacking either gene exhibit major abnormalities of XY gonad development at 14.5 dpc, including partial sex reversal. Embryos lacking three out of four functional copies of p300/Cbp exhibit complete XY gonadal sex reversal and have greatly reduced expression of the key testis-determining genes Sry and Sox9. An analysis of histone acetylation at the Sry promoter in mutant gonads at 11.5 dpc shows a reduction in levels of the positive histone mark H3K27Ac. Our data suggest a role for CBP/p300 in testis determination mediated by control of histone acetylation at the Sry locus and reveal a novel element in the epigenetic control of Sry and mammalian sex determination. They also suggest possible novel causes of human disorders of sex development (DSD).
Collapse
Affiliation(s)
- Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Marilena Xipolita
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Paul Brindle
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center Mainz, 55128 Mainz, Germany
| | - Sara Wells
- Mary Lyon Centre, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| |
Collapse
|
53
|
Del Barco Barrantes I, Stephan-Otto Attolini C, Slobodnyuk K, Igea A, Gregorio S, Gawrzak S, Gomis RR, Nebreda AR. Regulation of Mammary Luminal Cell Fate and Tumorigenesis by p38α. Stem Cell Reports 2017; 10:257-271. [PMID: 29290625 PMCID: PMC5768988 DOI: 10.1016/j.stemcr.2017.11.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022] Open
Abstract
Mammary stem and progenitor cells are essential for mammary gland homeostasis and are also candidates for cells of origin of mammary tumors. Here, we have investigated the function of the protein kinase p38α in the mammary gland using mice that delete this protein in the luminal epithelial cells. We show that p38α regulates the fate of luminal progenitor cells through modulation of the transcription factor RUNX1, an important controller of the estrogen receptor-positive cell lineage. We also provide evidence that the regulation of RUNX1 by p38α probably involves the kinase MSK1, which phosphorylates histone H3 at the RUNX1 promoter. Moreover, using a mouse model for breast cancer initiated by luminal cells, we show that p38α downregulation in mammary epithelial cells reduces tumor burden, which correlates with decreased numbers of tumor-initiating cells. Collectively, our results define a key role for p38α in luminal progenitor cell fate that affects mammary tumor formation. Luminal progenitor cell fate in the mammary gland is regulated by p38α p38α controls the ER transcriptional program by modulating RUNX1 p38α regulates H3 phosphorylation at the RUNX1 promoter through the kinase MSK1 p38α promotes mammary tumorigenesis by maintaining luminal tumor-initiating cells
Collapse
Affiliation(s)
- Ivan Del Barco Barrantes
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Konstantin Slobodnyuk
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Ana Igea
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Sara Gregorio
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Sylwia Gawrzak
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Roger R Gomis
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; ICREA, Passeig de Lluís Companys 23, 08010 Barcelona, Spain; CIBERONC, 08028 Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; ICREA, Passeig de Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
54
|
Upadhyay K, Loke J, O V, Taragin B, Ostrer H. Biallelic mutations in FLNB cause a skeletal dysplasia with 46,XY gonadal dysgenesis by activating β-catenin. Clin Genet 2017; 93:412-416. [PMID: 29095481 DOI: 10.1111/cge.13165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 01/05/2023]
Abstract
Filamin B (FLNB) functions as a switch that can affect chrondrocyte development and endochondral bone formation through a series of signaling molecules and transcription factors that also affect Sertoli cell development. Here, we report a subject with a novel skeletal dysplasia and co-existing 46,XY gonadal dysgenesis and biallelic mutations in FLNB. Whole exome sequencing was performed to identify mutations. Quantitative polymerase chain reaction (qPCR) and flow variant assays were performed to quantify RNA, proteins and phosphorylated proteins. The TOPFLASH reporter was performed to quantify β-catenin activity. Mutations were identified in the FLNB gene (FLNB:p.F964L, FLNB:p.A1577V). These mutations increased binding of FLNB protein to the MAP3K1 and RAC1 signal transduction complex and activated β-catenin and had different effects on phosphorylation of MAP kinase pathway intermediates and SOX9 expression. Direct activation of β-catenin through the FLNB-MAP3K1-RAC1 complex by FLNB mutations is a novel mechanism for causing 46,XY gonadal dysgenesis. The mechanism of action varies from those reported previously for loss of function mutations in SOX9 and gain-of-function mutations in MAP3K1.
Collapse
Affiliation(s)
- K Upadhyay
- Departments of Pathology and Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York
| | - J Loke
- Departments of Pathology and Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York
| | - V O
- Departments of Pathology and Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York
| | - B Taragin
- Departments of Pathology and Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York
| | - H Ostrer
- Departments of Pathology and Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York
| |
Collapse
|
55
|
Rahmoun M, Lavery R, Laurent-Chaballier S, Bellora N, Philip GK, Rossitto M, Symon A, Pailhoux E, Cammas F, Chung J, Bagheri-Fam S, Murphy M, Bardwell V, Zarkower D, Boizet-Bonhoure B, Clair P, Harley VR, Poulat F. In mammalian foetal testes, SOX9 regulates expression of its target genes by binding to genomic regions with conserved signatures. Nucleic Acids Res 2017; 45:7191-7211. [PMID: 28472341 PMCID: PMC5499551 DOI: 10.1093/nar/gkx328] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 04/17/2017] [Indexed: 01/22/2023] Open
Abstract
In mammalian embryonic gonads, SOX9 is required for the determination of Sertoli cells that orchestrate testis morphogenesis. To identify genetic networks directly regulated by SOX9, we combined analysis of SOX9-bound chromatin regions from murine and bovine foetal testes with sequencing of RNA samples from mouse testes lacking Sox9. We found that SOX9 controls a conserved genetic programme that involves most of the sex-determining genes. In foetal testes, SOX9 modulates both transcription and directly or indirectly sex-specific differential splicing of its target genes through binding to genomic regions with sequence motifs that are conserved among mammals and that we called ‘Sertoli Cell Signature’ (SCS). The SCS is characterized by a precise organization of binding motifs for the Sertoli cell reprogramming factors SOX9, GATA4 and DMRT1. As SOX9 biological role in mammalian gonads is to determine Sertoli cells, we correlated this genomic signature with the presence of SOX9 on chromatin in foetal testes, therefore equating this signature to a genomic bar code of the fate of foetal Sertoli cells. Starting from the hypothesis that nuclear factors that bind to genomic regions with SCS could functionally interact with SOX9, we identified TRIM28 as a new SOX9 partner in foetal testes.
Collapse
Affiliation(s)
- Massilva Rahmoun
- Institute of Human Genetics, CNRS-University of Montpellier UMR9002, 34396 Montpellier cedex 5, France
| | - Rowena Lavery
- The Hudson Institute of Medical Research and Department of Anatomy, Monash University, Melbourne, Australia
| | - Sabine Laurent-Chaballier
- Institut de Recherche en Cancérologie de Montpellier, IRCM, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France
| | - Nicolas Bellora
- Instituto Andino Patagónico de Tecnologías Biológicas y Geoambientales (IPATEC), Universidad Nacional del Comahue - CONICET, Bariloche, Argentina
| | - Gayle K Philip
- VLSCI, LAB-14, 700 Swanston Street, Carlton 3053, Victoria, Australia
| | - Moïra Rossitto
- Institute of Human Genetics, CNRS-University of Montpellier UMR9002, 34396 Montpellier cedex 5, France
| | - Aleisha Symon
- The Hudson Institute of Medical Research and Department of Anatomy, Monash University, Melbourne, Australia
| | - Eric Pailhoux
- INRA Biologie du Développement et Reproduction, Domaine de Vilvert, 78352 Jouy-en-Josas Cedex, France
| | - Florence Cammas
- Institut de Recherche en Cancérologie de Montpellier, IRCM, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France
| | - Jessica Chung
- VLSCI, LAB-14, 700 Swanston Street, Carlton 3053, Victoria, Australia
| | - Stefan Bagheri-Fam
- The Hudson Institute of Medical Research and Department of Anatomy, Monash University, Melbourne, Australia
| | - Mark Murphy
- Department of Genetics, Cell Biology and Development, University of Minnesota, 6-160 Jackson hall, 321 Church St, SE, Minneapolis, MN 55455, USA
| | - Vivian Bardwell
- Department of Genetics, Cell Biology and Development, University of Minnesota, 6-160 Jackson hall, 321 Church St, SE, Minneapolis, MN 55455, USA
| | - David Zarkower
- Department of Genetics, Cell Biology and Development, University of Minnesota, 6-160 Jackson hall, 321 Church St, SE, Minneapolis, MN 55455, USA
| | - Brigitte Boizet-Bonhoure
- Institute of Human Genetics, CNRS-University of Montpellier UMR9002, 34396 Montpellier cedex 5, France
| | - Philippe Clair
- University of Montpellier, Montpellier GenomiX, bat 24, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Vincent R Harley
- The Hudson Institute of Medical Research and Department of Anatomy, Monash University, Melbourne, Australia
| | - Francis Poulat
- Institute of Human Genetics, CNRS-University of Montpellier UMR9002, 34396 Montpellier cedex 5, France
| |
Collapse
|
56
|
Maguire M, Larsen MC, Foong YH, Tanumihardjo S, Jefcoate CR. Cyp1b1 deletion and retinol deficiency coordinately suppress mouse liver lipogenic genes and hepcidin expression during post-natal development. Mol Cell Endocrinol 2017; 454:50-68. [PMID: 28583802 PMCID: PMC5985816 DOI: 10.1016/j.mce.2017.05.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/30/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
UNLABELLED Cyp1b1 deletion and gestational vitamin A deficiency (GVAD) redirect adult liver gene expression. A matched sufficient pre- and post-natal diet, which has high carbohydrate and normal iron content (LF12), increased inflammatory gene expression markers in adult livers that were suppressed by GVAD and Cyp1b1 deletion. At birth on the LF12 diet, Cyp1b1 deletion and GVAD each suppress liver expression of the iron suppressor, hepcidin (Hepc), while increasing stellate cell activation markers and suppressing post-natal increases in lipogenesis. Hepc was less suppressed in Cyp1b1-/- pups with a standard breeder diet, but was restored by iron supplementation of the LF12 diet. CONCLUSIONS The LF12 diet delivered low post-natal iron and attenuated Hepc. Hepc decreases in Cyp1b1-/- and GVAD mice resulted in stellate activation and lipogenesis suppression. Endothelial BMP6, a Hepc stimulant, is a potential coordinator and Cyp1b1 target. These neonatal changes in Cyp1b1-/- mice link to diminished adult obesity and liver inflammation.
Collapse
Affiliation(s)
- Meghan Maguire
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI 53705, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Yee Hoon Foong
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Sherry Tanumihardjo
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Colin R Jefcoate
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI 53705, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
57
|
Koopman P, Sinclair A, Lovell-Badge R. Of sex and determination: marking 25 years of Randy, the sex-reversed mouse. Development 2017; 143:1633-7. [PMID: 27190031 DOI: 10.1242/dev.137372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 03/21/2016] [Indexed: 12/31/2022]
Abstract
On Thursday 9 May 1991, the world awoke to front-page news of a breakthrough in biological research. From Washington to Wollongong, newspapers, radio and TV were abuzz with the story of a transgenic mouse in London called Randy. Why was this mouse so special? The mouse in question was a chromosomal female (XX) made male by the presence of a transgene containing the Y chromosome gene Sry This sex-reversal provided clear experimental proof that Sry was the elusive mammalian sex-determining gene. Twenty-five years on, we reflect on what this discovery meant for our understanding of how males and females arise and what remains to be understood.
Collapse
Affiliation(s)
- Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew Sinclair
- Murdoch Children's Research Institute and Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Melbourne, Victoria 3052, Australia
| | | |
Collapse
|
58
|
Liu G, Chen M, Yu C, Wang W, Yang L, Li Z, Wang W, Chen J. Molecular cloning, characterization and functional analysis of a putative mitogen-activated protein kinase kinase kinase 4 (MEKK4) from blood clam Tegillarca granosa. FISH & SHELLFISH IMMUNOLOGY 2017; 66:372-381. [PMID: 28476674 DOI: 10.1016/j.fsi.2017.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
The mitogen-activated protein kinase (MAPK) cascades stand for one of the most important signaling mechanisms in response to environmental stimuli. In the present study, we cloned and identified for the first time the full-length cDNA of MAPK kinase kinase 4 (TgMEKK4) from Blood clam Tegillarca granosa using rapid amplification of cDNA ends method. The full-length cDNA of TgMEKK4 was of 1605 bp in length, encoding a polypeptide of 364 amino acids with a predicted molecular mass of 41.22 kDa and theoretical isoelectric point of 6.29. The conserved MEKK4-domain was identified in TgMEKK4 by SMART program analysis. Homology analysis of the deduced amino acid sequence of TgMEKK4 with other known sequences revealed that TgMEKK4 shared 58%-80% identity to MEKK4s from other species. TgMEKK4 mRNA transcripts could be detected in all tissues examined with the highest expression level in the gill by qRT-PCR. The mRNA expression of TgMEKK4 was up-regulated significantly in hemocytes after Vibrio parahaemolyticus, Vibrio alginolyticus and Lipopolysaccharide (LPS) challenges. Overexpression of TgMEKK4 in HEK 293T cells resulted in the activation of JNK and ERK, but not p38. Consistently, In vivo study indicated that LPS stimulation enhanced JNK, ERK and p38 phosphorylation in blood clams. These results suggest that TgMEKK4 is a powerful factor in the regulation of genes that may be involved in innate immune response of blood clam.
Collapse
Affiliation(s)
- Guosheng Liu
- School of Marine Sciences, Ningbo University, Ningbo, 315211 Zhejiang, China; State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Mingliang Chen
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China.
| | - Chen Yu
- School of Marine Sciences, Ningbo University, Ningbo, 315211 Zhejiang, China; State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Wei Wang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Lirong Yang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Zengpeng Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Weiyi Wang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China
| | - Jianming Chen
- State Key Laboratory Breeding Base of Marine Genetic Resources, Fujian Key Laboratory of Marine Genetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005 Fujian, China.
| |
Collapse
|
59
|
Radhakrishnan S, Literman R, Mizoguchi B, Valenzuela N. MeDIP-seq and nCpG analyses illuminate sexually dimorphic methylation of gonadal development genes with high historic methylation in turtle hatchlings with temperature-dependent sex determination. Epigenetics Chromatin 2017; 10:28. [PMID: 28533820 PMCID: PMC5438563 DOI: 10.1186/s13072-017-0136-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/12/2017] [Indexed: 12/15/2022] Open
Abstract
Background DNA methylation alters gene expression but not DNA sequence and mediates some cases of phenotypic plasticity. Temperature-dependent sex determination (TSD) epitomizes phenotypic plasticity where environmental temperature drives embryonic sexual fate, as occurs commonly in turtles. Importantly, the temperature-specific transcription of two genes underlying gonadal differentiation is known to be induced by differential methylation in TSD fish, turtle and alligator. Yet, how extensive is the link between DNA methylation and TSD remains unclear. Here we test for broad differences in genome-wide DNA methylation between male and female hatchling gonads of the TSD painted turtle Chrysemys picta using methyl DNA immunoprecipitation sequencing, to identify differentially methylated candidates for future study. We also examine the genome-wide nCpG distribution (which affects DNA methylation) in painted turtles and test for historic methylation in genes regulating vertebrate gonadogenesis. Results Turtle global methylation was consistent with other vertebrates (57% of the genome, 78% of all CpG dinucleotides). Numerous genes predicted to regulate turtle gonadogenesis exhibited sex-specific methylation and were proximal to methylated repeats. nCpG distribution predicted actual turtle DNA methylation and was bimodal in gene promoters (as other vertebrates) and introns (unlike other vertebrates). Differentially methylated genes, including regulators of sexual development, had lower nCpG content indicative of higher historic methylation. Conclusions Ours is the first evidence suggesting that sexually dimorphic DNA methylation is pervasive in turtle gonads (perhaps mediated by repeat methylation) and that it targets numerous regulators of gonadal development, consistent with the hypothesis that it may regulate thermosensitive transcription in TSD vertebrates. However, further research during embryogenesis will help test this hypothesis and the alternative that instead, most differential methylation observed in hatchlings is the by-product of sexual differentiation and not its cause. Electronic supplementary material The online version of this article (doi:10.1186/s13072-017-0136-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Srihari Radhakrishnan
- Bioinformatics and Computational Biology Program, Iowa State University, Ames, IA 50011 USA.,Department of Ecology, Evolution and Organismal Biology, Iowa State University, 251 Bessey Hall, Ames, IA 50011 USA
| | - Robert Literman
- Ecology and Evolutionary Biology Program, Iowa State University, Ames, IA 50011 USA.,Department of Ecology, Evolution and Organismal Biology, Iowa State University, 251 Bessey Hall, Ames, IA 50011 USA
| | - Beatriz Mizoguchi
- Interdepartmental Genetics and Genomics Program, Iowa State University, Ames, IA 50011 USA.,Department of Ecology, Evolution and Organismal Biology, Iowa State University, 251 Bessey Hall, Ames, IA 50011 USA
| | - Nicole Valenzuela
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, 251 Bessey Hall, Ames, IA 50011 USA
| |
Collapse
|
60
|
Radhakrishnan S, Literman R, Neuwald J, Severin A, Valenzuela N. Transcriptomic responses to environmental temperature by turtles with temperature-dependent and genotypic sex determination assessed by RNAseq inform the genetic architecture of embryonic gonadal development. PLoS One 2017; 12:e0172044. [PMID: 28296881 PMCID: PMC5352168 DOI: 10.1371/journal.pone.0172044] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/30/2017] [Indexed: 12/24/2022] Open
Abstract
Vertebrate sexual fate is decided primarily by the individual's genotype (GSD), by the environmental temperature during development (TSD), or both. Turtles exhibit TSD and GSD, making them ideal to study the evolution of sex determination. Here we analyze temperature-specific gonadal transcriptomes (RNA-sequencing validated by qPCR) of painted turtles (Chrysemys picta TSD) before and during the thermosensitive period, and at equivalent stages in soft-shell turtles (Apalone spinifera-GSD), to test whether TSD's and GSD's transcriptional circuitry is identical but deployed differently between mechanisms. Our data show that most elements of the mammalian urogenital network are active during turtle gonadogenesis, but their transcription is generally more thermoresponsive in TSD than GSD, and concordant with their sex-specific function in mammals [e.g., upregulation of Amh, Ar, Esr1, Fog2, Gata4, Igf1r, Insr, and Lhx9 at male-producing temperature, and of β-catenin, Foxl2, Aromatase (Cyp19a1), Fst, Nf-kb, Crabp2 at female-producing temperature in Chrysemys]. Notably, antagonistic elements in gonadogenesis (e.g., β-catenin and Insr) were thermosensitive only in TSD early-embryos. Cirbp showed warm-temperature upregulation in both turtles disputing its purported key TSD role. Genes that may convert thermal inputs into sex-specific development (e.g., signaling and hormonal pathways, RNA-binding and heat-shock) were differentially regulated. Jak-Stat, Nf-κB, retinoic-acid, Wnt, and Mapk-signaling (not Akt and Ras-signaling) potentially mediate TSD thermosensitivity. Numerous species-specific ncRNAs (including Xist) were differentially-expressed, mostly upregulated at colder temperatures, as were unannotated loci that constitute novel TSD candidates. Cirbp showed warm-temperature upregulation in both turtles. Consistent transcription between turtles and alligator revealed putatively-critical reptilian TSD elements for male (Sf1, Amh, Amhr2) and female (Crabp2 and Hspb1) gonadogenesis. In conclusion, while preliminary, our data helps illuminate the regulation and evolution of vertebrate sex determination, and contribute genomic resources to guide further research into this fundamental biological process.
Collapse
Affiliation(s)
- Srihari Radhakrishnan
- Bioinformatics and Computational Biology Program, Iowa State University, Ames, IA, United States of America
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America
| | - Robert Literman
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America
- Ecology and Evolutionary Biology Program, Iowa State University, Ames, IA, United States of America
| | - Jennifer Neuwald
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America
| | - Andrew Severin
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America
- Genome Informatics Facility, Iowa State University, Ames, IA, United States of America
| | - Nicole Valenzuela
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America
| |
Collapse
|
61
|
Bashamboo A, Eozenou C, Rojo S, McElreavey K. Anomalies in human sex determination provide unique insights into the complex genetic interactions of early gonad development. Clin Genet 2017; 91:143-156. [DOI: 10.1111/cge.12932] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 12/19/2022]
Affiliation(s)
- A. Bashamboo
- Human Developmental Genetics Unit; Institut Pasteur; Paris France
| | - C. Eozenou
- Human Developmental Genetics Unit; Institut Pasteur; Paris France
| | - S. Rojo
- Human Developmental Genetics Unit; Institut Pasteur; Paris France
| | - K. McElreavey
- Human Developmental Genetics Unit; Institut Pasteur; Paris France
| |
Collapse
|
62
|
Maatouk DM, Natarajan A, Shibata Y, Song L, Crawford GE, Ohler U, Capel B. Genome-wide identification of regulatory elements in Sertoli cells. Development 2017; 144:720-730. [PMID: 28087634 DOI: 10.1242/dev.142554] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/30/2016] [Indexed: 01/22/2023]
Abstract
A current goal of molecular biology is to identify transcriptional networks that regulate cell differentiation. However, identifying functional gene regulatory elements has been challenging in the context of developing tissues where material is limited and cell types are mixed. To identify regulatory sites during sex determination, we subjected Sertoli cells from mouse fetal testes to DNaseI-seq and ChIP-seq for H3K27ac. DNaseI-seq identified putative regulatory sites around genes enriched in Sertoli and pregranulosa cells; however, active enhancers marked by H3K27ac were enriched proximal to only Sertoli-enriched genes. Sequence analysis identified putative binding sites of known and novel transcription factors likely controlling Sertoli cell differentiation. As a validation of this approach, we identified a novel Sertoli cell enhancer upstream of Wt1, and used it to drive expression of a transgenic reporter in Sertoli cells. This work furthers our understanding of the complex genetic network that underlies sex determination and identifies regions that potentially harbor non-coding mutations underlying disorders of sexual development.
Collapse
Affiliation(s)
- Danielle M Maatouk
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.,Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL 60611, USA
| | - Anirudh Natarajan
- Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA
| | - Yoichiro Shibata
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Lingyun Song
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA.,Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27708, USA
| | - Uwe Ohler
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708, USA.,Max Delbruck Center for Molecular Medicine, Berlin 13125, Germany
| | - Blanche Capel
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
63
|
Bashamboo A, McElreavey K. Mechanism of Sex Determination in Humans: Insights from Disorders of Sex Development. Sex Dev 2016; 10:313-325. [DOI: 10.1159/000452637] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2016] [Indexed: 12/13/2022] Open
|
64
|
Saba-El-Leil MK, Frémin C, Meloche S. Redundancy in the World of MAP Kinases: All for One. Front Cell Dev Biol 2016; 4:67. [PMID: 27446918 PMCID: PMC4921452 DOI: 10.3389/fcell.2016.00067] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
The protein kinases ERK1 and ERK2 are the effector components of the prototypical ERK1/2 mitogen-activated protein (MAP) kinase pathway. This signaling pathway regulates cell proliferation, differentiation and survival, and is essential for embryonic development and cellular homeostasis. ERK1 and ERK2 homologs share similar biochemical properties but whether they exert specific physiological functions or act redundantly has been a matter of controversy. However, recent studies now provide compelling evidence in support of functionally redundant roles of ERK1 and ERK2 in embryonic development and physiology. In this review, we present a critical assessment of the evidence for the functional specificity or redundancy of MAP kinase isoforms. We focus on the ERK1/ERK2 pathway but also discuss the case of JNK and p38 isoforms.
Collapse
Affiliation(s)
- Marc K Saba-El-Leil
- Institute for Research in Immunology and Cancer, Université de Montréal Montréal, QC, Canada
| | - Christophe Frémin
- Institute for Research in Immunology and Cancer, Université de MontréalMontréal, QC, Canada; Institute for Research in Cancer of MontpellierMontpellier, France
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Université de MontréalMontréal, QC, Canada; Molecular Biology Program, Université de MontréalMontréal, QC, Canada; Department of Pharmacology, Université de MontréalMontréal, QC, Canada
| |
Collapse
|
65
|
Warr N, Siggers P, Carré GA, Wells S, Greenfield A. Genetic Analyses Reveal Functions for MAP2K3 and MAP2K6 in Mouse Testis Determination. Biol Reprod 2016; 94:103. [PMID: 27009039 PMCID: PMC5842889 DOI: 10.1095/biolreprod.115.138057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/08/2016] [Indexed: 01/27/2023] Open
Abstract
Testis determination in mammals is initiated by expression of SRY in somatic cells of the embryonic gonad. Genetic analyses in the mouse have revealed a requirement for mitogen-activated protein kinase (MAPK) signaling in testis determination: targeted loss of the kinases MAP3K4 and p38 MAPK causes complete XY embryonic gonadal sex reversal. These kinases occupy positions at the top and bottom level, respectively, in the canonical threetier MAPK-signaling cascade: MAP3K, MAP2K, MAPK. To date, no role in sex determination has been attributed to a MAP2K, although such a function is predicted to exist. Here, we report roles for the kinases MAP2K3 and MAP2K6 in testis determination. C57BL/6J (B6) embryos lacking MAP2K3 exhibited no significant abnormalities of testis development, whilst those lacking MAP2K6 exhibited a minor delay in testis determination. Compound mutants lacking three out of four functional alleles at the two loci also exhibited delayed testis determination and transient ovotestis formation as a consequence, suggestive of partially redundant roles for these kinases in testis determination. Early lethality of double-knockout embryos precludes analysis of sexual development. To reveal their roles in testis determination more clearly, we generated Map2k mutant B6 embryos using a weaker Sry allele (SryAKR). Loss of Map2k3 on this highly sensitized background exacerbates ovotestis development, whilst loss of Map2k6 results in complete XY gonadal sex reversal associated with reduction of Sry expression at 11.25 days postcoitum. Our data suggest that MAP2K6 functions in mouse testis determination, via positive effects on Sry, and also indicate a minor role for MAP2K3.
Collapse
Affiliation(s)
- Nick Warr
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Sara Wells
- The Mary Lyon Centre, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| |
Collapse
|
66
|
Windley SP, Wilhelm D. Signaling Pathways Involved in Mammalian Sex Determination and Gonad Development. Sex Dev 2016; 9:297-315. [PMID: 26905731 DOI: 10.1159/000444065] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 11/19/2022] Open
Abstract
The development of any organ system requires a complex interplay of cellular signals to initiate the differentiation and development of the heterogeneous cell and tissue types required to carry out the organs' functions. In this way, an extracellular stimulus is transmitted to an intracellular target through an array of interacting protein intermediaries, ultimately enabling the target cell to elicit a response. Surprisingly, only a small number of signaling pathways are implicated throughout embryogenesis and are used over and over again. Gonadogenesis is a unique process in that 2 morphologically distinct organs, the testes and ovaries, arise from a common precursor, the bipotential genital ridge. Accordingly, most of the signaling pathways observed throughout embryogenesis also have been shown to be important for mammalian sex determination and gonad development. Here, we review the mechanisms of signal transduction within these pathways and the importance of these pathways throughout mammalian gonad development, mainly concentrating on data obtained in mouse but including other species where appropriate.
Collapse
Affiliation(s)
- Simon P Windley
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Vic., Australia
| | | |
Collapse
|
67
|
Abstract
In the female gonad, distinct signalling pathways activate ovarian differentiation while repressing the formation of testes. Human disorders of sex development (DSDs), such as 46,XX DSDs, can arise when this signalling is aberrant. Here we review the current understanding of the genetic mechanisms that control gonadal development, with particular emphasis on those that drive or inhibit ovarian differentiation. We discuss how disruption to these molecular pathways can lead to 46,XX disorders of ovarian development. Finally, we look at recently characterized novel genes and pathways that contribute and speculate how advances in technology will aid in further characterization of normal and disrupted human ovarian development.
Collapse
|
68
|
Carré GA, Greenfield A. The Gonadal Supporting Cell Lineage and Mammalian Sex Determination: The Differentiation of Sertoli and Granulosa Cells. Results Probl Cell Differ 2016; 58:47-66. [PMID: 27300175 DOI: 10.1007/978-3-319-31973-5_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The supporting cell lineage plays a crucial role in nurturing the development of germ cells in the adult gonad. Sertoli cells in the testis support the progression of spermatogonial stem cells through meiosis to the production of motile spermatozoa. Granulosa cells, meanwhile, are a critical component of the ovarian follicle that produces the mature oocyte. It is a distinctive feature of the embryonic gonad that at least some of the supporting cells are derived from a single sexually bipotential precursor lineage. It is the commitment of this somatic lineage to either the Sertoli or granulosa cell fate that defines sex determination. In this chapter we review what is known about the key molecules responsible for this lineage decision in the developing mammalian gonads, relying primarily on data from studies of mice and humans. We focus on recent advances in our understanding of the mutually antagonistic interactions of testis- and ovary-determining pathways and their complexity as revealed by genetic analyses. For the sake of simplicity, we will deal with supporting cells in testis and ovary development in separate sections, but numerous points of contact exist between these accounts of gonadogenesis in male and female embryos, primarily due to the aforementioned mutual antagonisms. The final section will offer a brief synthesis of these organ-specific overviews and a summary of the key themes that emerge in this review of supporting cell differentiation in mammalian sex determination.
Collapse
Affiliation(s)
- Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, OX11 0RD, UK
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
69
|
Chojnacka K, Zarzycka M, Mruk DD. Biology of the Sertoli Cell in the Fetal, Pubertal, and Adult Mammalian Testis. Results Probl Cell Differ 2016; 58:225-251. [PMID: 27300181 DOI: 10.1007/978-3-319-31973-5_9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A healthy man typically produces between 50 × 10(6) and 200 × 10(6) spermatozoa per day by spermatogenesis; in the absence of Sertoli cells in the male gonad, this individual would be infertile. In the adult testis, Sertoli cells are sustentacular cells that support germ cell development by secreting proteins and other important biomolecules that are essential for germ cell survival and maturation, establishing the blood-testis barrier, and facilitating spermatozoa detachment at spermiation. In the fetal testis, on the other hand, pre-Sertoli cells form the testis cords, the future seminiferous tubules. However, the role of pre-Sertoli cells in this process is much less clear than the function of Sertoli cells in the adult testis. Within this framework, we provide an overview of the biology of the fetal, pubertal, and adult Sertoli cell, highlighting relevant cell biology studies that have expanded our understanding of mammalian spermatogenesis.
Collapse
Affiliation(s)
- Katarzyna Chojnacka
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY, 10065, USA
| | - Marta Zarzycka
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Dolores D Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
70
|
Understanding sex determination in the mouse: genetics, epigenetics and the story of mutual antagonisms. J Genet 2015; 94:585-90. [DOI: 10.1007/s12041-015-0565-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
71
|
Lan CW, Chen MJ, Tai KY, Yu DC, Yang YC, Jan PS, Yang YS, Chen HF, Ho HN. Functional microarray analysis of differentially expressed genes in granulosa cells from women with polycystic ovary syndrome related to MAPK/ERK signaling. Sci Rep 2015; 5:14994. [PMID: 26459919 PMCID: PMC4602237 DOI: 10.1038/srep14994] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 09/14/2015] [Indexed: 11/26/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Although its aetiology and pathogenesis remain unclear, recent studies suggest that the dysfunction of granulosa cells may partly be responsible. This study aimed to use cDNA microarray technology to compare granulosa cell gene expression profiles in women with and without PCOS to identify genes that may be aetiologically implicated in the pathogenesis of PCOS. The study cohort included 12 women undergoing in vitro fertilization, six with PCOS and six without PCOS. Differential gene expression profiles were classified by post-analyses of microarray data, followed by western blot analyses to confirm the microarray data of selected genes. In total, 243 genes were differentially expressed (125 upregulated and 118 downregulated) between the PCOS and non-PCOS granulosa cells. These genes are involved in reproductive system development, amino acid metabolism and cellular development and proliferation. Comparative analysis revealed genes involved in the mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK) signaling pathways. Western blot analyses confirmed that mitogen-activated protein kinase kinase kinase 4 and phospho-ERK1/2 were decreased in PCOS granulosa cells. This study identified candidate genes involved in MAPK/ERK signaling pathways that may influence the function of granulosa cells in PCOS.
Collapse
Affiliation(s)
- Chen-Wei Lan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University Taipei, Taiwan.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Mei-Jou Chen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Kang-Yu Tai
- Genome and Systems Biology Degree Program, National Taiwan University Taipei, Taiwan
| | - Danny Cw Yu
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Yu-Chieh Yang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Pey-Shynan Jan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Yu-Shih Yang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan
| | - Hsin-Fu Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University Taipei, Taiwan.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan university Taipei, Taiwan
| | - Hong-Nerng Ho
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University Taipei, Taiwan.,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, College of Medicine and the Hospital, National Taiwan University Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan university Taipei, Taiwan
| |
Collapse
|
72
|
Wang Y, Liu W, Yang Q, Yu M, Zhang Z. Di (2-ethylhexyl) phthalate exposure during pregnancy disturbs temporal sex determination regulation in mice offspring. Toxicology 2015. [DOI: 10.1016/j.tox.2015.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
73
|
Tachibana M. Epigenetics of sex determination in mammals. Reprod Med Biol 2015; 15:59-67. [PMID: 29259422 DOI: 10.1007/s12522-015-0223-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 09/02/2015] [Indexed: 10/23/2022] Open
Abstract
Epigenetics is the study of changes in gene function that cannot be explained by changes in DNA sequence. A mammalian body contains more than two hundred types of cells. Since all of them are derived from a single fertilized egg, their genotypes are identical. However, the gene expression patterns are different between the cell types, indicating that each cell type has unique own "epigenotype". Epigenetic gene regulation mechanisms essentially contribute to various processes of mammalian development. The essence of epigenetic regulation is the structural change of chromatin to modulate gene activity in a spatiotemporal manner. DNA methylation and histone modifications are the major epigenetic mechanisms. Sex determination is the process for gender establishment. There are two types of sex-determining mechanisms in animals, environmental sex determination (ESD) and genotypic sex determination (GSD). Recent studies have provided some evidence that epigenetic mechanisms play indispensable roles in ESD and GSD. Some fishes undergo ESD, in which DNA methylation is essentially involved. GSD is employed in therian mammals, where Sry (sex-determining region on the Y chromosome) triggers testis differentiation from undifferentiated gonads. Sry expression is tightly regulated in a spatiotemporal manner. A recent study demonstrated that histone modification is involved in Sry regulation. In this review, we discuss the role of epigenetic mechanisms for sex determination in mammals and other vertebrates.
Collapse
Affiliation(s)
- Makoto Tachibana
- Department of Enzyme Chemistry, Institute for Enzyme Research Tokushima University 18-15-3 Kuramoto-cho 770-8503 Tokushima Japan
| |
Collapse
|
74
|
Ayers KL, Lambeth LS, Davidson NM, Sinclair AH, Oshlack A, Smith CA. Identification of candidate gonadal sex differentiation genes in the chicken embryo using RNA-seq. BMC Genomics 2015; 16:704. [PMID: 26377738 PMCID: PMC4574023 DOI: 10.1186/s12864-015-1886-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/27/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite some advances in recent years, the genetic control of gonadal sex differentiation during embryogenesis is still not completely understood. To identify new candidate genes involved in ovary and testis development, RNA-seq was used to define the transcriptome of embryonic chicken gonads at the onset of sexual differentiation (day 6.0/stage 29). RESULTS RNA-seq revealed more than 1000 genes that were transcribed in a sex-biased manner at this early stage of gonadal differentiation. Comparison with undifferentiated gonads revealed that sex biased expression was derived primarily from autosomal rather than sex-linked genes. Gene ontology and pathway analysis indicated that many of these genes encoded proteins involved in extracellular matrix function and cytoskeletal remodelling, as well as tubulogenesis. Several of these genes are novel candidate regulators of gonadal sex differentiation, based on sex-biased expression profiles that are altered following experimental sex reversal. We further characterised three female-biased (ovarian) genes; calpain-5 (CAPN5), G-protein coupled receptor 56 (GPR56), and FGFR3 (fibroblast growth factor receptor 3). Protein expression of these candidates in the developing ovaries suggests that they play an important role in this tissue. CONCLUSIONS This study provides insight into the earliest steps of vertebrate gonad sex differentiation, and identifies novel candidate genes for ovarian and testicular development.
Collapse
Affiliation(s)
- Katie L Ayers
- Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, 3052, Parkville, VIC, Australia. .,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.
| | - Luke S Lambeth
- Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, 3052, Parkville, VIC, Australia.
| | - Nadia M Davidson
- Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, 3052, Parkville, VIC, Australia.
| | - Andrew H Sinclair
- Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, 3052, Parkville, VIC, Australia. .,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.
| | - Alicia Oshlack
- Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, 3052, Parkville, VIC, Australia.
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
75
|
Bashamboo A, McElreavey K. Human sex-determination and disorders of sex-development (DSD). Semin Cell Dev Biol 2015; 45:77-83. [DOI: 10.1016/j.semcdb.2015.10.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 11/28/2022]
|
76
|
Loke H, Harley V, Lee J. Biological factors underlying sex differences in neurological disorders. Int J Biochem Cell Biol 2015; 65:139-50. [PMID: 26028290 DOI: 10.1016/j.biocel.2015.05.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 11/28/2022]
Abstract
The prevalence, age of onset, pathophysiology, and symptomatology of many neurological and neuropsychiatric conditions differ significantly between males and females. Females suffer more from mood disorders such as depression and anxiety, whereas males are more susceptible to deficits in the dopamine system including Parkinson's disease (PD), attention-deficit hyperactivity disorder (ADHD), schizophrenia, and autism spectrum disorders (ASD). Until recently, these sex differences have been explained solely by the neuroprotective actions of sex hormones in females. Emerging evidence however indicates that the sex chromosome genes (i.e. X- and Y-linked genes) also contribute to brain sex differences. In particular, the Y-chromosome gene, SRY (Sex-determining Region on the Y chromosome) is an interesting candidate as it is expressed in dopamine-abundant brain regions, where it regulates dopamine biosynthesis and dopamine-mediated functions such as voluntary movement in males. Furthermore, SRY expression is dysregulated in a toxin-induced model of PD, suggesting a role for SRY in the pathogenesis of dopamine cells. Taken together, these studies highlight the importance of understanding the interplay between sex-specific hormones and sex-specific genes in healthy and diseased brain. In particular, better understanding of regulation and function of SRY in the male brain could provide entirely novel and important insights into genetic factors involved in the susceptibility of men to neurological disorders, as well as development of novel sex-specific therapies.
Collapse
Affiliation(s)
- Hannah Loke
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Vincent Harley
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.
| | - Joohyung Lee
- Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
77
|
Wu Q, Fukuda K, Weinstein M, Graff JM, Saga Y. SMAD2 and p38 signaling pathways act in concert to determine XY primordial germ cell fate in mice. Development 2015; 142:575-86. [PMID: 25605784 DOI: 10.1242/dev.119446] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The sex of primordial germ cells (PGCs) is determined in developing gonads on the basis of cues from somatic cells. In XY gonads, sex-determining region Y (SRY) triggers fibroblast growth factor 9 (FGF9) expression in somatic cells. FGF signaling, together with downstream nodal/activin signaling, promotes male differentiation in XY germ cells by suppressing retinoic acid (RA)-dependent meiotic entry and inducing male-specific genes. However, the mechanism by which nodal/activin signaling regulates XY PGC fate is unknown. We uncovered the roles of SMAD2/3 and p38 MAPK, the putative downstream factors of nodal/activin signaling, in PGC sexual fate decision. We found that conditional deletion of Smad2, but not Smad3, from XY PGCs led to a loss of male-specific gene expression. Moreover, suppression of RA signaling did not rescue male-specific gene expression in Smad2-mutant testes, indicating that SMAD2 signaling promotes male differentiation in a RA-independent manner. By contrast, we found that p38 signaling has an important role in the suppression of RA signaling. The Smad2 deletion did not disrupt the p38 signaling pathway even though Nodal expression was significantly reduced, suggesting that p38 was not regulated by nodal signaling in XY PGCs. Additionally, the inhibition of p38 signaling in the Smad2-mutant testes severely impeded XY PGC differentiation and induced meiosis. In conclusion, we propose a model in which p38 and SMAD2 signaling coordinate to determine the sexual fate of XY PGCs.
Collapse
Affiliation(s)
- Quan Wu
- Department of Genetics, SOKENDAI, Yata 1111, Mishima 411-8540, Japan Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan
| | - Kurumi Fukuda
- Department of Genetics, SOKENDAI, Yata 1111, Mishima 411-8540, Japan Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan
| | - Michael Weinstein
- Department of Molecular Genetics and Division of Human Cancer Genetics, Ohio State University, 484 W. 12th Avenue, Columbus, OH 43210, USA
| | - Jonathan M Graff
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, NB5.118, Dallas, TX 75390, USA
| | - Yumiko Saga
- Department of Genetics, SOKENDAI, Yata 1111, Mishima 411-8540, Japan Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan
| |
Collapse
|
78
|
Matsunaga E, Nambu S, Oka M, Iriki A. Comparative analysis of developmentally regulated expressions of Gadd45a, Gadd45b, and Gadd45g in the mouse and marmoset cerebral cortex. Neuroscience 2015; 284:566-580. [PMID: 25450958 DOI: 10.1016/j.neuroscience.2014.10.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/23/2014] [Accepted: 10/20/2014] [Indexed: 11/18/2022]
Abstract
The cerebral cortex is an indispensable region that is involved in higher cognitive function in the mammalian brain, and is particularly evolved in the primate brain. It has been demonstrated that cortical areas are formed by both innate and activity-dependent mechanisms. However, it remains unknown what molecular changes induce cortical expansion and complexity during primate evolution. Active DNA methylation/demethylation is one of the epigenetic mechanisms that can modify gene expression via the methylation/demethylation of promoter regions. Three growth arrest and DNA damage-inducible small nuclear proteins, Gadd45 alpha, beta, and gamma, have been identified as regulators of methylation status. To understand the involvement of epigenetic factors in primate cortical evolution, we started by analyzing expression of these demethylation genes in the developing common marmoset (Callithrix jacchus) and mouse (Mus musculus) brain. In the marmoset brain, we found that cortical expression levels of Gadd45 alpha and gamma were reduced during development, whereas there was high expression of Gadd45 beta in some areas of the adult brain, including the prefrontal, temporal, posterior parietal and insula cortices, which are particularly expanded in greater primates and humans. Compared to the marmoset brain, there were no clear regional differences and constant or reduced Gadd45 expression was seen between juvenile and adult mouse brain. Double staining with a neuronal marker revealed that most Gadd45-expressing cells were NeuN-positive neurons. Thus, these results suggest the possibility that differential Gadd45 expression affects neurons, contributing cortical evolution and diversity.
Collapse
Affiliation(s)
- E Matsunaga
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako 351-0198, Japan.
| | - S Nambu
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako 351-0198, Japan
| | - M Oka
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako 351-0198, Japan
| | - A Iriki
- Laboratory for Symbolic Cognitive Development, RIKEN Brain Science Institute, Hirosawa 2-1, Wako 351-0198, Japan
| |
Collapse
|
79
|
Abstract
Epigenetics is the study of changes in gene function that cannot be explained by changes in DNA sequence. A mammalian body contains more than two-hundred different types of cells, all derived from a single fertilized egg. Epigenetic gene regulation mechanisms essentially contribute to various processes of mammalian development. The essence of epigenetic regulation is the modulation of gene activity through changes in chromatin structure. DNA methylation and histone modifications are the major epigenetic mechanisms. Sex determination is the process of establishing a gender. Sry, the sex-determining gene in therian mammals, initiates testis differentiation. Recent studies have provided evidence that epigenetic mechanisms contribute to Sry regulation.
Collapse
Affiliation(s)
- Makoto Tachibana
- Department of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University
| |
Collapse
|
80
|
Liu C, Zhang W, Yang D, Liu Y. Molecular Characterization, Polymorphism, and Association of Porcine GADD45G Gene. Anim Biotechnol 2015; 26:230-6. [PMID: 25927170 DOI: 10.1080/10495398.2015.1005216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Growth arrest and DNA-damage-inducible gamma (GADD45G) is a reproduction related gene. In this study, the full-length cDNA sequence of porcine GADD45G gene was cloned through rapid amplification of cDNA ends (RACE) method. The porcine GADD45G gene encodes a protein of 159 amino acids that shares high homology with the GADD45G of nine species: chimpanzee (97%), sumatran orangutan (97%), white-tufted-ear marmoset (97%), northern white-cheeked gibbon (97%), cattle (97%), human (97%), rhesus monkey (97%), rat (96%), and mouse (95%). This novel porcine gene was assigned to GeneID: 100152997. Phylogenetic analysis revealed that the porcine GADD45G gene has a closer genetic relationship with the GADD45G gene of cattle. Computer-assisted analysis indicated that porcine GADD45G gene is structured in four exons and three introns. PCR-Rsa I-RFLP was established to detect an A/G mutation on the position of 294-bp of coding sequence and eight pig breeds display obvious genotype and allele frequency differences at this mutation locus. Association of this SNP with litter size traits was assessed in Large White (n = 100) and Landrace (n = 100) pig populations, and result demonstrated that this polymorphic locus was significantly associated with the litter size of all parities in Large White and Landrace sows (P < 0.01). Therefore, porcine GADD45G gene could be a useful candidate gene in selection for increasing the litter size. These data serve as a foundation for further insight into this novel porcine gene.
Collapse
Affiliation(s)
- Chong Liu
- a Key Laboratory for Fertility Regulation and Eugenics of Minority Research of Yunnan Province , Kunming , China
| | | | | | | |
Collapse
|
81
|
Zhao L, Gu H, Chang J, Wu J, Wang D, Chen S, Yang X, Qian B. MicroRNA-383 regulates the apoptosis of tumor cells through targeting Gadd45g. PLoS One 2014; 9:e110472. [PMID: 25415264 PMCID: PMC4240536 DOI: 10.1371/journal.pone.0110472] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 09/09/2014] [Indexed: 11/18/2022] Open
Abstract
Background MicroRNAs (miRNAs) are a class of small non-coding single-stranded RNA molecules that inhibit gene expression at post-transcriptional level. Gadd45g (growth arrest and DNA-damage-inducible 45 gamma) is a stress-response protein, which has been implicated in several biological processes, including DNA repair, the cell cycle and cell differentiation. Results In this work, we found that miR-383 is a negative regulator of Gadd45g. Forced expression of miR-383 decreased the expression of Gadd45g through binding to the 3′ untranslated region (3′-UTR), whereas inhibition of miR-383 increased Gadd45g expression. The presence of miR-383 increased the cellular sensitivity to DNA damage in breast cancer cells, which was rescued by ectopic expression of Gadd45g without the 3′-UTR. miR-383 also regulates the expression of Gadd45g in embryonic stem (ES) cells, but not their apoptosis under genotoxic stress. miR-383 was further showed to negatively regulate ES cell differentiation via targeting Gadd45g, which subsequently modulates the pluripotency-associated genes. Taken together, our study demonstrates that miR-383 is a negative regulator of Gadd45g in both tumor cells and ES cells, however, has distinct function in regulating cell apoptosis. miR-383 may be used as antineoplastic agents in cancer chemotherapy. Conclusion We demonstrate for the first time that miR-383 can specifically regulates the expression of Gadd45g by directly targeting to the 3-UTR region of Gadd45g mRNA, a regulatory process conserved in human tumor cells and mouse embryonic stem cells. These two compotents can be potentially used as antineoplastic agents in cancer chemotherapy.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Epigenetics and Cancer Research, Medical Science Building C-315, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haihui Gu
- Department of Transfusion Medicine, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jianfeng Chang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road/1239 Siping Road, Shanghai 200120/200092, China
| | - Junyu Wu
- Institute of Epigenetics and Cancer Research, Medical Science Building C-315, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Daliang Wang
- Institute of Epigenetics and Cancer Research, Medical Science Building C-315, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Su Chen
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road/1239 Siping Road, Shanghai 200120/200092, China
| | - Xiaomei Yang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road/1239 Siping Road, Shanghai 200120/200092, China
- * E-mail: (XY); (BQ)
| | - Baohua Qian
- Department of Transfusion Medicine, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
- * E-mail: (XY); (BQ)
| |
Collapse
|
82
|
Liu WJ, Zhang LY, Shao CW, Wang N, Liu K, Wen HS, Zhang N, Dong ZD, Zhang JJ, Chen SL. Molecular characterization and functional divergence of two Gadd45g homologs in sex determination in half-smooth tongue sole (Cynoglossus semilaevis). Comp Biochem Physiol B Biochem Mol Biol 2014; 177-178:56-64. [PMID: 25220289 DOI: 10.1016/j.cbpb.2014.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 05/01/2014] [Accepted: 09/01/2014] [Indexed: 12/29/2022]
Abstract
The growth arrest and DNA-damage-inducible protein 45 gamma (Gadd45g) is known to play a major role in embryonic development and sex determination. In this study, two Gadd45g genes were isolated from half-smooth tongue sole (Cynoglossus semilaevis). Using chromosomal fluorescence in situ hybridization (FISH), Gadd45g1 and Gadd45g2 were located on the W and Z chromosomes, respectively. The full-length cDNA sequences of Gadd45g1 (1270bp) and Gadd45g2 (1181bp) were predicted to contain a 480-bp coding sequence that could encode a protein of 159 amino acids residues. A phylogenetic tree showed that the predicted Gadd45g1 and Gadd45g2 amino acid sequences clustered closely in one branch. It is proposed that Gadd45g1 and Gadd45g2 are paralogous genes derived from the divergence of the sex chromosome. Ka/Ks ratios indicated that Gadd45g1 and Gadd45g2 may have undergone a high number of mutations and have a divergence time of only about 68,000years, although Gadd45g homologs are highly conserved. The qRT-PCR demonstrated that Gadd45g1 and Gadd45g2 were highly expressed in ovary, and negligibly expressed in testis of male and neo-male. During development of the ovary (from 80 to 150days), the expression levels of both genes reached high levels. Gadd45g1 was also highly expressed at 50days, the stage just before gonad differentiation in C. semilaevis. All these findings imply functional divergence of the two Gadd45g homologs; Gadd45g1 may be necessary for sex differentiation in the early stage of gonad development, and then Gadd45g1 and Gadd45g2 maintain ovary development and the female character of half-smooth tongue sole.
Collapse
Affiliation(s)
- Wan-Jun Liu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China; Fisheries College, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Li-Yan Zhang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Chang-Wei Shao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Na Wang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Kun Liu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Hai-Shen Wen
- Fisheries College, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ning Zhang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Zhong-Dian Dong
- Fisheries College, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jun-Jie Zhang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China
| | - Song-Lin Chen
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fisheries Sciences, Qingdao 266071, China.
| |
Collapse
|
83
|
Abstract
Sex-specific gonadal development starts with formation of the bipotential gonad, which then differentiates into either a mature testis or an ovary. This process is dependent on activation of either the testis-specific or the ovary-specific pathway while the opposite pathway is continuously repressed. A network of transcription factors tightly regulates initiation and maintenance of these distinct pathways; disruption of these networks can lead to disorders of sex development in humans and male-to-female or female-to-male sex reversal in mice. Sry is the Y-linked master switch that is both required and sufficient to drive the testis-determining pathway. Another key component of the testis pathway is Sox9, which acts immediately downstream of Sry. In contrast to the testis pathway, no single sex-determining factor has been identified in the ovary pathway; however, multiple genes, such as Foxl2, Rspo1, Ctnnb1, and Wnt4, seem to work synergistically and in parallel to ensure proper ovary development. Our understanding of the regulatory networks that underpin testis and ovary development has grown substantially over the past two decades.
Collapse
Affiliation(s)
- Stefanie Eggers
- Murdoch Childrens Research Institute, Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital, 50 Flemington Road, Melbourne, VIC 3052, Australia
| | - Thomas Ohnesorg
- Murdoch Childrens Research Institute, Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital, 50 Flemington Road, Melbourne, VIC 3052, Australia
| | - Andrew Sinclair
- Murdoch Childrens Research Institute, Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital, 50 Flemington Road, Melbourne, VIC 3052, Australia
| |
Collapse
|
84
|
Abstract
Mammalian sex determination is the unique process whereby a single organ, the bipotential gonad, undergoes a developmental switch that promotes its differentiation into either a testis or an ovary. Disruptions of this complex genetic process during human development can manifest as disorders of sex development (DSDs). Sex development can be divided into two distinct processes: sex determination, in which the bipotential gonads form either testes or ovaries, and sex differentiation, in which the fully formed testes or ovaries secrete local and hormonal factors to drive differentiation of internal and external genitals, as well as extragonadal tissues such as the brain. DSDs can arise from a number of genetic lesions, which manifest as a spectrum of gonadal (gonadal dysgenesis to ovotestis) and genital (mild hypospadias or clitoromegaly to ambiguous genitalia) phenotypes. The physical attributes and medical implications associated with DSDs confront families of affected newborns with decisions, such as gender of rearing or genital surgery, and additional concerns, such as uncertainty over the child's psychosexual development and personal wishes later in life. In this Review, we discuss the underlying genetics of human sex determination and focus on emerging data, genetic classification of DSDs and other considerations that surround gender development and identity in individuals with DSDs.
Collapse
Affiliation(s)
- Valerie A Arboleda
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095-7088, USA
| | - David E Sandberg
- Department of Pediatrics, Division of Child Behavioral Health and Child Health Evaluation &Research (CHEAR) Unit, University of Michigan, 300 North Ingalls Street, Ann Arbor, MI 48109-5456, USA
| | - Eric Vilain
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095-7088, USA
| |
Collapse
|
85
|
Chassot AA, Gillot I, Chaboissier MC. R-spondin1, WNT4, and the CTNNB1 signaling pathway: strict control over ovarian differentiation. Reproduction 2014; 148:R97-110. [PMID: 25187620 DOI: 10.1530/rep-14-0177] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Sex differentiation is a unique developmental process. Starting from a bipotential gonad, it gives rise to the ovary and the testis, two highly specialized organs that differ morphologically and physiologically despite sharing common reproductive and endocrine functions. This highlights the specific plasticity of the gonadal precursors and the existence of complex antagonistic genetic regulation. Mammalian sex determination is controlled by paternal transmission of the Y-linked gene, sex-determining region Y (SRY). Using mouse models, it has been shown that the main role of Sry is to activate the expression of the transcription factor Sox9; either one of these two genes is necessary and sufficient to allow testicular development through Sertoli cell differentiation. Thus, defects in SRY/Sry and/or SOX9/Sox9 expression result in male-to-female sex reversal of XY individuals. Molecular mechanisms governing ovarian differentiation remained unknown for a long time, until the discovery of the roles of R-spondin1 (RSPO1) and WNT4. In XX individuals, activation of the β-catenin signaling pathway by the secreted proteins RSPO1 and WNT4 is required to allow granulosa cell differentiation and, in turn, ovarian differentiation. Thus, mutations in RSPO1 result in female-to-male sex reversal of XX patients, and mouse models have allowed the identification of genetic cascades activated by RSPO1 and WNT4 to regulate ovarian development. In this review, we will discuss the respective roles of RSPO1, WNT4, and the β-catenin signaling pathway during ovarian differentiation in mice.
Collapse
Affiliation(s)
- Anne-Amandine Chassot
- University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France
| | - Isabelle Gillot
- University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France
| | - Marie-Christine Chaboissier
- University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France University of Nice-Sophia AntipolisParc Valrose, F-06108 Nice, FranceUMR-INSERM1091IBV, F-06108 Nice, France
| |
Collapse
|
86
|
Regulation of male sex determination: genital ridge formation and Sry activation in mice. Cell Mol Life Sci 2014; 71:4781-802. [PMID: 25139092 PMCID: PMC4233110 DOI: 10.1007/s00018-014-1703-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 11/27/2022]
Abstract
Sex determination is essential for the sexual reproduction to generate the next generation by the formation of functional male or female gametes. In mammals, primary sex determination is commenced by the presence or absence of the Y chromosome, which controls the fate of the gonadal primordium. The somatic precursor of gonads, the genital ridge is formed at the mid-gestation stage and gives rise to one of two organs, a testis or an ovary. The fate of the genital ridge, which is governed by the differentiation of somatic cells into Sertoli cells in the testes or granulosa cells in the ovaries, further determines the sex of an individual and their germ cells. Mutation studies in human patients with disorders of sex development and mouse models have revealed factors that are involved in mammalian sex determination. In most of mammals, a single genetic trigger, the Y-linked gene Sry (sex determination region on Y chromosome), regulates testicular differentiation. Despite identification of Sry in 1990, precise mechanisms underlying the sex determination of bipotential genital ridges are still largely unknown. Here, we review the recent progress that has provided new insights into the mechanisms underlying genital ridge formation as well as the regulation of Sry expression and its functions in male sex determination of mice.
Collapse
|
87
|
Kuno J, Poueymirou WT, Gong G, Siao CJ, Clarke G, Esau L, Kojak N, Posca J, Atanasio A, Strein J, Yancopoulos GD, Lai KMV, DeChiara TM, Frendewey D, Auerbach W, Valenzuela DM. Generation of fertile and fecund F0 XY female mice from XY ES cells. Transgenic Res 2014; 24:19-29. [PMID: 25087174 DOI: 10.1007/s11248-014-9815-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 07/02/2014] [Indexed: 12/13/2022]
Abstract
Known examples of male to female sex reversal in mice are caused by either strain incompatibilities or mutations in genes required for male sex determination. The resultant XY females are often sterile or exhibit very poor fertility. We describe here embryonic stem (ES) cell growth conditions that promote the production of healthy, anatomically normal fertile and fecund female F0 generation mice completely derived from gene-targeted XY male ES cells. The sex reversal is a transient trait that is not transmitted to the F1 progeny. Growth media with low osmolality and reduced sodium bicarbonate, maintained throughout the gene targeting process, enhance the yield of XY females. As a practical application of the induced sex reversal, we demonstrate the generation of homozygous mutant mice ready for phenotypic studies by the breeding of F0 XY females with their isogenic XY male clonal siblings, thereby eliminating one generation of breeding and the associated costs.
Collapse
Affiliation(s)
- Junko Kuno
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
Sex determination refers to the developmental decision that directs the bipotential genital ridge to develop as a testis or an ovary. Genetic studies on mice and humans have led to crucial advances in understanding the molecular fundamentals of sex determination and the mutually antagonistic signaling pathway. In this review, we summarize the current molecular mechanisms of sex determination by focusing on the known critical sex determining genes and their related signaling pathways in mammalian vertebrates from mice to humans. We also discuss the underlying delicate balance between testis and ovary sex determination pathways, concentrating on the antagonisms between major sex determining genes.
Collapse
Affiliation(s)
- Zhen-Yu She
- The Sperm LaboratoryCollege of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm LaboratoryCollege of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| |
Collapse
|
89
|
Abstract
The coactivator peroxisome proliferator-activated receptor-gamma coactivator 1 α (PGC-1α) is widely considered a central transcriptional regulator of adaptive thermogenesis in brown adipose tissue (BAT). However, mice lacking PGC-1α specifically in adipose tissue have only mild thermogenic defects, suggesting the presence of additional regulators. Using the activity of estrogen-related receptors (ERRs), downstream effectors of PGC-1α, as read-out in a high-throughput genome-wide cDNA screen, we identify here growth arrest and DNA-damage-inducible protein 45 γ (GADD45γ) as a cold-induced activator of uncoupling protein 1 (UCP1) and oxidative capacity in BAT. Mice lacking Gadd45γ have defects in Ucp1 induction and the thermogenic response to cold. GADD45γ works by activating MAPK p38, which is a potent activator of ERRβ and ERRγ transcriptional function. GADD45γ activates ERRγ independently of PGC-1 coactivators, yet synergizes with PGC-1α to induce the thermogenic program. Our findings elucidate a previously unidentified GADD45γ/p38/ERRγ pathway that regulates BAT thermogenesis and may enable new approaches for the stimulation of energy expenditure. Our study also implicates GADD45 proteins as general metabolic regulators.
Collapse
|
90
|
Larney C, Bailey TL, Koopman P. Switching on sex: transcriptional regulation of the testis-determining gene Sry. Development 2014; 141:2195-205. [PMID: 24866114 DOI: 10.1242/dev.107052] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mammalian sex determination hinges on the development of ovaries or testes, with testis fate being triggered by the expression of the transcription factor sex-determining region Y (Sry). Reduced or delayed Sry expression impairs testis development, highlighting the importance of its accurate spatiotemporal regulation and implying a potential role for SRY dysregulation in human intersex disorders. Several epigenetic modifiers, transcription factors and kinases are implicated in regulating Sry transcription, but it remains unclear whether or how this farrago of factors acts co-ordinately. Here we review our current understanding of Sry regulation and provide a model that assembles all known regulators into three modules, each converging on a single transcription factor that binds to the Sry promoter. We also discuss potential future avenues for discovering the cis-elements and trans-factors required for Sry regulation.
Collapse
Affiliation(s)
- Christian Larney
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Timothy L Bailey
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
91
|
Czech DP, Lee J, Correia J, Loke H, Möller EK, Harley VR. Transient neuroprotection by SRY upregulation in dopamine cells following injury in males. Endocrinology 2014; 155:2602-12. [PMID: 24708242 DOI: 10.1210/en.2013-2158] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Emerging evidence suggest sex-specific regulation of dopamine neurons may underlie susceptibility of males to disorders such as Parkinson's disease (PD). In healthy male dopamine neurons, the Y-chromosome gene product, the sex-determining region on the Y chromosome (SRY) modulates dopamine biosynthesis and motor function. We investigated the regulation and function of SRY in a model of dopamine cell injury. Treatment with the dopaminergic toxin, 6-hydroxydopamine (6-OHDA), significantly elevated SRY mRNA expression (9-fold) in human male dopamine M17 cells. SRY up-regulation occurred via the p-quinone pathway, associated with a 3.5-fold increase in expression of GADD45γ, a DNA damage inducible factor gene and known SRY regulator. In turn, a signaling cascade involving GADD45γ/p38-MAPK/GATA activated the SRY promoter. Knockdown of SRY mRNA in 6-OHDA-treated M17 cells was deleterious, increasing levels of reactive oxygen species (ROS), pro-apoptotic marker PUMA mRNA, and cell injury (+25%, +32% and +34%, respectively). Conversely, ectopic over-expression of SRY in 6-OHDA-treated female SH-SY5Y cells was protective, decreasing ROS, PUMA, and cell injury (-40%, -46%, and -30%, respectively). However, the 6-OHDA-induced increase in SRY expression was diminished with higher concentrations of toxins or with chronic exposure to 6-OHDA. We conclude that SRY upregulation after dopamine cell injury is initially a protective response in males, but diminishes with gradual loss in dopamine cells. We speculate that dysregulation of SRY may contribute the susceptibility of males to PD.
Collapse
Affiliation(s)
- Daniel P Czech
- Brain and Gender Laboratory (D.P.C., J.L., J.C., H.L., E.K.M., V.R.H.), MIMR-PHI (formerly Prince Henry's Institute of Medical Research), Department of Biochemistry and Molecular Biology (D.P.C., V.R.H.), and Department of Anatomy and Developmental Biology (J.L., V.R.H), Monash University, Melbourne, 3168, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
92
|
A novel mouse Fgfr2 mutant, hobbyhorse (hob), exhibits complete XY gonadal sex reversal. PLoS One 2014; 9:e100447. [PMID: 24956260 PMCID: PMC4067367 DOI: 10.1371/journal.pone.0100447] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 05/23/2014] [Indexed: 12/13/2022] Open
Abstract
The secreted molecule fibroblast growth factor 9 (FGF9) plays a critical role in testis determination in the mouse. In embryonic gonadal somatic cells it is required for maintenance of SOX9 expression, a key determinant of Sertoli cell fate. Conditional gene targeting studies have identified FGFR2 as the main gonadal receptor for FGF9 during sex determination. However, such studies can be complicated by inefficient and variable deletion of floxed alleles, depending on the choice of Cre deleter strain. Here, we report a novel, constitutive allele of Fgfr2, hobbyhorse (hob), which was identified in an ENU-based forward genetic screen for novel testis-determining loci. Fgr2hob is caused by a C to T mutation in the invariant exon 7, resulting in a polypeptide with a mis-sense mutation at position 263 (Pro263Ser) in the third extracellular immunoglobulin-like domain of FGFR2. Mutant homozygous embryos show severe limb and lung defects and, when on the sensitised C57BL/6J (B6) genetic background, undergo complete XY gonadal sex reversal associated with failure to maintain expression of Sox9. Genetic crosses employing a null mutant of Fgfr2 suggest that Fgr2hob is a hypomorphic allele, affecting both the FGFR2b and FGFR2c splice isoforms of the receptor. We exploited the consistent phenotype of this constitutive mutant by analysing MAPK signalling at the sex-determining stage of gonad development, but no significant abnormalities in mutant embryos were detected.
Collapse
|
93
|
Abstract
To identify novel genomic regions that regulate sex determination, we utilized the powerful C57BL/6J-Y(POS) (B6-Y(POS)) model of XY sex reversal where mice with autosomes from the B6 strain and a Y chromosome from a wild-derived strain, Mus domesticus poschiavinus (Y(POS)), show complete sex reversal. In B6-Y(POS), the presence of a 55-Mb congenic region on chromosome 11 protects from sex reversal in a dose-dependent manner. Using mouse genetic backcross designs and high-density SNP arrays, we narrowed the congenic region to a 1.62-Mb genomic region on chromosome 11 that confers 80% protection from B6-Y(POS) sex reversal when one copy is present and complete protection when two copies are present. It was previously believed that the protective congenic region originated from the 129S1/SviMJ (129) strain. However, genomic analysis revealed that this region is not derived from 129 and most likely is derived from the semi-inbred strain POSA. We show that the small 1.62-Mb congenic region that protects against B6-Y(POS) sex reversal is located within the Sox9 promoter and promotes the expression of Sox9, thereby driving testis development within the B6-Y(POS) background. Through 30 years of backcrossing, this congenic region was maintained, as it promoted male sex determination and fertility despite the female-promoting B6-Y(POS) genetic background. Our findings demonstrate that long-range enhancer regions are critical to developmental processes and can be used to identify the complex interplay between genome variants, epigenetics, and developmental gene regulation.
Collapse
|
94
|
Abstract
CONTEXT Disorders of sex development (DSDs) may arise from genetic defects in testis or ovary determination. Current analytical technologies and improved understanding of major regulatory pathways have cast new insight into the genetic basis for these disorders. EVIDENCE ACQUISITION A PubMed search was performed for the years 2011-13 using the terms "disorder of sex development," "gonadal dysgenesis," "ovarian dysgenesis," "array CGH," and "whole exome sequencing." Only articles from peer-reviewed journals were included. EVIDENCE SYNTHESIS Key themes that emerged included aberrant regulation of SOX9 via the hTES promoter in 46,XY gonadal DSDs, the role of the MAPK pathway in normal and aberrant gonadal development, and the role of new technologies in identification of gonadal DSDs. CONCLUSIONS With the advent of the robust new technologies of array comparative genomic hybridization and genomic sequencing in recent years, many new sex-determining genes have been identified. These genes have been organized into ovarian- and testicular-determining pathways that can block each other's activities. Identification of a mutation in a sex-determining gene in an individual affected with a DSD may warrant more extensive investigation for other phenotypic effects as well as genetic testing of other family members.
Collapse
Affiliation(s)
- Harry Ostrer
- Departments of Pathology, Genetics, and Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
95
|
Quinn A, Kashimada K, Davidson TL, Ng ET, Chawengsaksophak K, Bowles J, Koopman P. A site-specific, single-copy transgenesis strategy to identify 5' regulatory sequences of the mouse testis-determining gene Sry. PLoS One 2014; 9:e94813. [PMID: 24743337 PMCID: PMC3990564 DOI: 10.1371/journal.pone.0094813] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/19/2014] [Indexed: 11/18/2022] Open
Abstract
The Y-chromosomal gene SRY acts as the primary trigger for male sex determination in mammalian embryos. Correct regulation of SRY is critical: aberrant timing or level of Sry expression is known to disrupt testis development in mice and we hypothesize that mutations that affect regulation of human SRY may account for some of the many cases of XY gonadal dysgenesis that currently remain unexplained. However, the cis-sequences involved in regulation of Sry have not been identified, precluding a test of this hypothesis. Here, we used a transgenic mouse approach aimed at identifying mouse Sry 5' flanking regulatory sequences within 8 kb of the Sry transcription start site (TSS). To avoid problems associated with conventional pronuclear injection of transgenes, we used a published strategy designed to yield single-copy transgene integration at a defined, transcriptionally open, autosomal locus, Col1a1. None of the Sry transgenes tested was expressed at levels compatible with activation of Sox9 or XX sex reversal. Our findings indicate either that the Col1a1 locus does not provide an appropriate context for the correct expression of Sry transgenes, or that the cis-sequences required for Sry expression in the developing gonads lie beyond 8 kb 5' of the TSS.
Collapse
Affiliation(s)
- Alexander Quinn
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- * E-mail:
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Tara-Lynne Davidson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ee Ting Ng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | | | - Josephine Bowles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| |
Collapse
|
96
|
Salvador JM, Brown-Clay JD, Fornace AJ. Gadd45 in stress signaling, cell cycle control, and apoptosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 793:1-19. [PMID: 24104470 DOI: 10.1007/978-1-4614-8289-5_1] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The first identified Gadd45 gene, Gadd45a, encodes a ubiquitously expressed protein that is often induced by DNA damage and other stress signals associated with growth arrest and apoptosis. This protein and the other two members of this small gene family, Gadd45b and Gadd45g, have been implicated in a variety of the responses to cell injury including cell cycle checkpoints, apoptosis, and DNA repair. In vivo, many of the prominent roles for the Gadd45 proteins are associated with signaling mediated by p38 mitogen-activated protein kinases (MAPK). Gadd45 proteins can contribute to p38 activation either by activation of upstream kinase(s) or by direct interaction. In vivo, there are important tissue and cell-type-specific differences in the roles for Gadd45 in MAPK signaling. In addition to being p53-regulated, Gadd45a has been found to contribute to p53 activation via p38. Like other stress and signaling proteins, Gadd45 proteins show complex regulation and numerous effectors.
Collapse
Affiliation(s)
- Jesús M Salvador
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, (CNB-CSIC) Lab 417, c/Darwin n 3, Campus Cantoblanco, 28049, Madrid, Spain
| | | | | |
Collapse
|
97
|
Ohnesorg T, Vilain E, Sinclair AH. The genetics of disorders of sex development in humans. Sex Dev 2014; 8:262-72. [PMID: 24504012 DOI: 10.1159/000357956] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
One of the defining events during human embryonic development with the most far-reaching effects for the individual is whether the embryo develops as male or female. The crucial step in this process is the differentiation of the bipotential embryonic gonads into either testes or ovaries. If the embryo inherits X and Y sex chromosomes, the Y-linked SRY (sex determining region in Y) gene initiates a network of genes that results in a functional testis and ultimately a male phenotype. By contrast, in an embryo with 2 X chromosomes, the undifferentiated gonad develops as an ovary resulting in a female phenotype. Perturbation of any of the genes in either the testicular or ovarian developmental pathway can result in individuals with disorders of sex development. In this review, we provide a summary of known components of testicular or ovarian pathways and their antagonistic actions and give a brief overview of new technologies currently used to identify the missing pieces of the sex development network.
Collapse
Affiliation(s)
- Thomas Ohnesorg
- Murdoch Children's Research Institute and Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Melbourne, Vic., Australia
| | | | | |
Collapse
|
98
|
Warr N, Siggers P, Carré GA, Bogani D, Brixey R, Akiyoshi M, Tachibana M, Teboul L, Wells S, Sanderson J, Greenfield A. Transgenic expression of Map3k4 rescues T-associated sex reversal (Tas) in mice. Hum Mol Genet 2014; 23:3035-44. [PMID: 24452333 PMCID: PMC4014197 DOI: 10.1093/hmg/ddu020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Disorders of sex development in the human population range in severity from mild genital defects to gonadal sex reversal. XY female development has been associated with heterozygous mutations in several genes, including SOX9, WT1 and MAP3K1. In contrast, XY sex reversal in mice usually requires complete absence of testis-determining gene products. One exception to this involves T-associated sex reversal (Tas), a phenomenon characterized by the formation of ovotestes or ovaries in XY mice hemizygous for the hairpin-tail (T(hp)) or T-Orleans (T(Orl)) deletions on proximal mouse chromosome 17. We recently reported that mice heterozygous for a null allele of Map3k4, which resides in the T(hp) deletion, exhibit XY ovotestis development and occasional gonadal sex reversal on the sensitized C57BL/6J-Y(AKR) (B6-Y(AKR)) genetic background, reminiscent of the Tas phenotype. However, these experiments did not exclude the possibility that loss of other loci in the T(hp) deletion, or other effects of the deletion itself, might contribute to Tas. Here, we show that disruption to Sry expression underlies XY gonadal defects in B6-Y(AKR) embryos harbouring the T(hp) deletion and that a functional Map3k4 bacterial artificial chromosome rescues these abnormalities by re-establishing a normal Sry expression profile. These data demonstrate that Map3k4 haploinsufficiency is the cause of T-associated sex reversal and that levels of this signalling molecule are a major determinant of the expression profile of Sry.
Collapse
|
99
|
Carré GA, Greenfield A. Characterising Novel Pathways in Testis Determination Using Mouse Genetics. Sex Dev 2014; 8:199-207. [DOI: 10.1159/000358402] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
100
|
Fujimoto Y, Tanaka S, Yamaguchi Y, Kobayashi H, Kuroki S, Tachibana M, Shinomura M, Kanai Y, Morohashi KI, Kawakami K, Nishinakamura R. Homeoproteins Six1 and Six4 Regulate Male Sex Determination and Mouse Gonadal Development. Dev Cell 2013; 26:416-30. [DOI: 10.1016/j.devcel.2013.06.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 06/11/2013] [Accepted: 06/19/2013] [Indexed: 01/11/2023]
|