1
|
Irie N, Lee SM, Lorenzi V, Xu H, Chen J, Inoue M, Kobayashi T, Sancho-Serra C, Drousioti E, Dietmann S, Vento-Tormo R, Song CX, Surani MA. DMRT1 regulates human germline commitment. Nat Cell Biol 2023; 25:1439-1452. [PMID: 37709822 PMCID: PMC10567552 DOI: 10.1038/s41556-023-01224-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
Germline commitment following primordial germ cell (PGC) specification during early human development establishes an epigenetic programme and competence for gametogenesis. Here we follow the progression of nascent PGC-like cells derived from human embryonic stem cells in vitro. We show that switching from BMP signalling for PGC specification to Activin A and retinoic acid resulted in DMRT1 and CDH5 expression, the indicators of migratory PGCs in vivo. Moreover, the induction of DMRT1 and SOX17 in PGC-like cells promoted epigenetic resetting with striking global enrichment of 5-hydroxymethylcytosine and locus-specific loss of 5-methylcytosine at DMRT1 binding sites and the expression of DAZL representing DNA methylation-sensitive genes, a hallmark of the germline commitment programme. We provide insight into the unique role of DMRT1 in germline development for advances in human germ cell biology and in vitro gametogenesis.
Collapse
Affiliation(s)
- Naoko Irie
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK.
- Metabolic Systems Laboratory, Live Imaging Center, Central Institute for Experimental Animals, Kanagawa, Japan.
| | - Sun-Min Lee
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK
- Department of Physics, Konkuk University, Seoul, Republic of Korea
| | - Valentina Lorenzi
- Wellcome Sanger Institute, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | - Haiqi Xu
- Ludwig Institute for Cancer Research and Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jinfeng Chen
- Ludwig Institute for Cancer Research and Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Masato Inoue
- Ludwig Institute for Cancer Research and Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Toshihiro Kobayashi
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi, Japan
| | | | - Elena Drousioti
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK
| | - Sabine Dietmann
- Department of Developmental Biology and Institute for Informatics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Chun-Xiao Song
- Ludwig Institute for Cancer Research and Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M Azim Surani
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK.
- Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Zhang MF, Wan SC, Chen WB, Yang DH, Liu WQ, Li BL, Aierken A, Du XM, Li YX, Wu WP, Yang XC, Wei YD, Li N, Peng S, Li XL, Li GP, Hua JL. Transcription factor Dmrt1 triggers the SPRY1-NF-κB pathway to maintain testicular immune homeostasis and male fertility. Zool Res 2023; 44:505-521. [PMID: 37070575 PMCID: PMC10236308 DOI: 10.24272/j.issn.2095-8137.2022.440] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
Bacterial or viral infections, such as Brucella, mumps virus, herpes simplex virus, and Zika virus, destroy immune homeostasis of the testes, leading to spermatogenesis disorder and infertility. Of note, recent research shows that SARS-CoV-2 can infect male gonads and destroy Sertoli and Leydig cells, leading to male reproductive dysfunction. Due to the many side effects associated with antibiotic therapy, finding alternative treatments for inflammatory injury remains critical. Here, we found that Dmrt1 plays an important role in regulating testicular immune homeostasis. Knockdown of Dmrt1 in male mice inhibited spermatogenesis with a broad inflammatory response in seminiferous tubules and led to the loss of spermatogenic epithelial cells. Chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) revealed that Dmrt1 positively regulated the expression of Spry1, an inhibitory protein of the receptor tyrosine kinase (RTK) signaling pathway. Furthermore, immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP) analysis indicated that SPRY1 binds to nuclear factor kappa B1 (NF-κB1) to prevent nuclear translocation of p65, inhibit activation of NF-κB signaling, prevent excessive inflammatory reaction in the testis, and protect the integrity of the blood-testis barrier. In view of this newly identified Dmrt1- Spry1-NF-κB axis mechanism in the regulation of testicular immune homeostasis, our study opens new avenues for the prevention and treatment of male reproductive diseases in humans and livestock.
Collapse
Affiliation(s)
- Meng-Fei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shi-Cheng Wan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Bo Chen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Dong-Hui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Qing Liu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- Center of Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam 1105AZ, Amsterdam, Netherlands
| | - Ba-Lun Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Aili Aierken
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiao-Min Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yun-Xiang Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wen-Ping Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xin-Chun Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yu-Dong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xue-Ling Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Guang-Peng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Jin-Lian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
3
|
Imai A, Matsuda K, Niimi Y, Suzuki A. Loss of Dead end1 induces testicular teratomas from primordial germ cells that failed to undergo sexual differentiation in embryonic testes. Sci Rep 2023; 13:6398. [PMID: 37076592 PMCID: PMC10115811 DOI: 10.1038/s41598-023-33706-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/18/2023] [Indexed: 04/21/2023] Open
Abstract
Spontaneous testicular teratomas (STTs) are tumours comprising a diverse array of cell and tissue types, which are derived from pluripotent stem-like cells called embryonal carcinoma cells (ECCs). Although mouse ECCs originate from primordial germ cells (PGCs) in embryonic testes, the molecular basis underlying ECC development remains unclear. This study shows that the conditional deletion of mouse Dead end1 (Dnd1) from migrating PGCs leads to STT development. In Dnd1-conditional knockout (Dnd1-cKO) embryos, PGCs colonise the embryonic testes but fail to undergo sexual differentiation; subsequently, ECCs develop from a portion of the PGCs. Transcriptomic analyses reveal that PGCs not only fail to undergo sexual differentiation but are also prone to transformation into ECCs by upregulating the expression of marker genes for primed pluripotency in the testes of Dnd1-cKO embryos. Thus, our results clarify the role of Dnd1 in developing STTs and developmental process of ECC from PGC, providing novel insights into pathogenic mechanisms of STTs.
Collapse
Affiliation(s)
- Atsuki Imai
- Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa, Japan
| | - Kazuya Matsuda
- Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa, Japan
| | - Yuki Niimi
- Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa, Japan
- Research & Development Group, Center for Exploratory Research, Hitachi, Ltd., Kobe, Hyogo, Japan
| | - Atsushi Suzuki
- Division of Materials Science and Chemical Engineering, Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa, Japan.
- Division of Materials Science and Chemical Engineering, Faculty of Engineering, Yokohama National University, Yokohama, Kanagawa, Japan.
| |
Collapse
|
4
|
Rossitto M, Déjardin S, Rands CM, Le Gras S, Migale R, Rafiee MR, Neirijnck Y, Pruvost A, Nguyen AL, Bossis G, Cammas F, Le Gallic L, Wilhelm D, Lovell-Badge R, Boizet-Bonhoure B, Nef S, Poulat F. TRIM28-dependent SUMOylation protects the adult ovary from activation of the testicular pathway. Nat Commun 2022; 13:4412. [PMID: 35906245 PMCID: PMC9338040 DOI: 10.1038/s41467-022-32061-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/17/2022] [Indexed: 11/08/2022] Open
Abstract
Gonadal sexual fate in mammals is determined during embryonic development and must be actively maintained in adulthood. In the mouse ovary, oestrogen receptors and FOXL2 protect ovarian granulosa cells from transdifferentiation into Sertoli cells, their testicular counterpart. However, the mechanism underlying their protective effect is unknown. Here, we show that TRIM28 is required to prevent female-to-male sex reversal of the mouse ovary after birth. We found that upon loss of Trim28, ovarian granulosa cells transdifferentiate to Sertoli cells through an intermediate cell type, different from gonadal embryonic progenitors. TRIM28 is recruited on chromatin in the proximity of FOXL2 to maintain the ovarian pathway and to repress testicular-specific genes. The role of TRIM28 in ovarian maintenance depends on its E3-SUMO ligase activity that regulates the sex-specific SUMOylation profile of ovarian-specific genes. Our study identifies TRIM28 as a key factor in protecting the adult ovary from the testicular pathway.
Collapse
Affiliation(s)
- Moïra Rossitto
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Stephanie Déjardin
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
| | - Chris M Rands
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Stephanie Le Gras
- GenomEast platform, IGBMC, 1, rue Laurent Fries, 67404 ILLKIRCH Cedex, Illkirch-Graffenstaden, France
| | - Roberta Migale
- The Francis Crick Institute, 1 Midland Road, London, NW1 2 1AT, UK
| | | | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Alain Pruvost
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 91191, Gif-sur-Yvette, France
| | - Anvi Laetitia Nguyen
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 91191, Gif-sur-Yvette, France
| | - Guillaume Bossis
- Institut de Génétique Moléculaire de Montpellier (IGMM), University of Montpellier, CNRS, Montpellier, 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
| | - Lionel Le Gallic
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | | | | | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France.
| |
Collapse
|
5
|
Kapadia B, Shetty AC, Bollino D, Bhandary B, Lapidus RG, Mahmood K, Mahurkar A, Gartenhaus RB, Eckert RL, Emadi A. Translatome changes in acute myeloid leukemia cells post-exposure to pegcrisantaspase and venetoclax. Exp Hematol 2022; 108:55-63. [PMID: 35104581 DOI: 10.1016/j.exphem.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 11/04/2022]
Abstract
The clinical outcomes of patients with AML treated with available therapy remains unsatisfactory. We recently reported that the BCL-2 inhibitor, venetoclax, synergized with pegcrisantaspase (PegC) and demonstrated remarkable in vivo efficacy in a preclinical model of AML with complex karyotype. Ven-PegC combination blocks synthesis of proteins in AML cells by inhibiting cap-dependent translation of mRNA. To further explore the impact of Ven-PegC on protein translation, we used polysome profiling and high-throughput RNA-seq to characterize Ven-PegC dependent changes to the translatome. Here we report that the translation of five mRNAs, including two microRNAs, one rRNA, and two mitochondrial genes was altered after exposure to all three treatments (Ven, PegC and Ven-PegC). We focused our translatome validation studies on six additional genes related to translational efficiency that were modified by Ven-PegC. Notably, Ven-PegC treatment increased the RNA translation and protein level of Tribbles homolog 3 (TRIB3), eukaryotic translation initiation factor 3 subunit C (eIF3C), doublesex and mab-3 related transcription factor 1 (DMRT1), salt inducible kinase 1 (SIK1). We validated the observed changes in gene/protein expression in vitro and confirmed our cell line-based studies in the bone marrow of an AML PDX model after Ven-PegC treatment. These results support examining alterations in the translatome post-chemotherapy to offer insight into drug mechanism of action and to inform future therapeutic decisions.
Collapse
Affiliation(s)
- Bandish Kapadia
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Amol C Shetty
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland Institute of Genome Sciences, Baltimore, MD
| | - Dominique Bollino
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland School of Medicine Department of Medicine, Baltimore, MD
| | - Binny Bhandary
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Rena G Lapidus
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland School of Medicine Department of Medicine, Baltimore, MD
| | - Kanwal Mahmood
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Anup Mahurkar
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland Institute of Genome Sciences, Baltimore, MD
| | - Ronald B Gartenhaus
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland Institute of Genome Sciences, Baltimore, MD
| | - Richard L Eckert
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland School of Medicine Department of Biochemistry and Molecular Biology
| | - Ashkan Emadi
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD; University of Maryland School of Medicine Department of Medicine, Baltimore, MD; University of Maryland School of Medicine Department of Pharmacology, Baltimore, MD.
| |
Collapse
|
6
|
Poulat F. Non-Coding Genome, Transcription Factors, and Sex Determination. Sex Dev 2021; 15:295-307. [PMID: 34727549 DOI: 10.1159/000519725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/15/2021] [Indexed: 11/19/2022] Open
Abstract
In vertebrates, gonadal sex determination is the process by which transcription factors drive the choice between the testicular and ovarian identity of undifferentiated somatic progenitors through activation of 2 different transcriptional programs. Studies in animal models suggest that sex determination always involves sex-specific transcription factors that activate or repress sex-specific genes. These transcription factors control their target genes by recognizing their regulatory elements in the non-coding genome and their binding motifs within their DNA sequence. In the last 20 years, the development of genomic approaches that allow identifying all the genomic targets of a transcription factor in eukaryotic cells gave the opportunity to globally understand the function of the nuclear proteins that control complex genetic programs. Here, the major transcription factors involved in male and female vertebrate sex determination and the genomic profiling data of mouse gonads that contributed to deciphering their transcriptional regulation role will be reviewed.
Collapse
Affiliation(s)
- Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, Montpellier, France
| |
Collapse
|
7
|
Taguchi J, Shibata H, Kabata M, Kato M, Fukuda K, Tanaka A, Ohta S, Ukai T, Mitsunaga K, Yamada Y, Nagaoka SI, Yamazawa S, Ohnishi K, Woltjen K, Ushiku T, Ozawa M, Saitou M, Shinkai Y, Yamamoto T, Yamada Y. DMRT1-mediated reprogramming drives development of cancer resembling human germ cell tumors with features of totipotency. Nat Commun 2021; 12:5041. [PMID: 34413299 PMCID: PMC8377058 DOI: 10.1038/s41467-021-25249-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
In vivo reprogramming provokes a wide range of cell fate conversion. Here, we discover that in vivo induction of higher levels of OSKM in mouse somatic cells leads to increased expression of primordial germ cell (PGC)-related genes and provokes genome-wide erasure of genomic imprinting, which takes place exclusively in PGCs. Moreover, the in vivo OSKM reprogramming results in development of cancer that resembles human germ cell tumors. Like a subgroup of germ cell tumors, propagated tumor cells can differentiate into trophoblasts. Moreover, these tumor cells give rise to induced pluripotent stem cells (iPSCs) with expanded differentiation potential into trophoblasts. Remarkably, the tumor-derived iPSCs are able to contribute to non-neoplastic somatic cells in adult mice. Mechanistically, DMRT1, which is expressed in PGCs, drives the reprogramming and propagation of the tumor cells in vivo. Furthermore, the DMRT1-related epigenetic landscape is associated with trophoblast competence of the reprogrammed cells and provides a therapeutic target for germ cell tumors. These results reveal an unappreciated route for somatic cell reprogramming and underscore the impact of reprogramming in development of germ cell tumors.
Collapse
Affiliation(s)
- Jumpei Taguchi
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minoto-ku, Tokyo, Japan
| | - Hirofumi Shibata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Otolaryngology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Mio Kabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Masaki Kato
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, Japan
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kei Fukuda
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, Japan
| | - Akito Tanaka
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Sho Ohta
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minoto-ku, Tokyo, Japan
| | - Tomoyo Ukai
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minoto-ku, Tokyo, Japan
| | - Kanae Mitsunaga
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yosuke Yamada
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - So I Nagaoka
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
- Department of Embryology, Nara Medical University, Nara, Japan
| | - Sho Yamazawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kotaro Ohnishi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Gastroenterology/Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Manabu Ozawa
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mitinori Saitou
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Yoichi Shinkai
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, Wako-shi, Saitama, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
- AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo, Japan
| | - Yasuhiro Yamada
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Minoto-ku, Tokyo, Japan.
- AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo, Japan.
| |
Collapse
|
8
|
Mori Y, Takashima S, Kanatsu-Shinohara M, Yi Z, Shinohara T. Cdc42 is required for male germline niche development in mice. Cell Rep 2021; 36:109550. [PMID: 34407418 DOI: 10.1016/j.celrep.2021.109550] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are maintained in a special microenvironment called a niche. However, much is unknown about components that constitute the niche. Here, we report that Cdc42 is essential for germline niche development. Sertoli cell-specific Cdc42-deficient mice showed normal premeiotic spermatogenesis. However, germ cells gradually disappeared during haploid cell formation and few germ cells remained in the mature testes. Spermatogonial transplantation experiments revealed a significant loss of SSCs in Cdc42-deficient testes. Moreover, Cdc42 deficiency in Sertoli cells downregulated GDNF, a critical factor for SSC maintenance. Cdc42-deficient Sertoli cells also exhibited lower nuclear MAPK1/3 staining. Inhibition of MAP2K1 or depletion of Pea15a scaffold protein downregulated GDNF expression. A screen of transcription factors revealed that Cdc42-deficient Sertoli cells downregulate DMRT1 and SOX9, both of which are critical for Sertoli cell development. These results indicate that Cdc42 is essential for niche function via MAPK1/3-dependent GDNF secretion.
Collapse
Affiliation(s)
- Yoshifumi Mori
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Seiji Takashima
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Zheng Yi
- Division of Experimental Hematology, Molecular Developmental Biology Graduate Program, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
9
|
Pluta J, Pyle LC, Nead KT, Wilf R, Li M, Mitra N, Weathers B, D'Andrea K, Almstrup K, Anson-Cartwright L, Benitez J, Brown CD, Chanock S, Chen C, Cortessis VK, Ferlin A, Foresta C, Gamulin M, Gietema JA, Grasso C, Greene MH, Grotmol T, Hamilton RJ, Haugen TB, Hauser R, Hildebrandt MAT, Johnson ME, Karlsson R, Kiemeney LA, Lessel D, Lothe RA, Loud JT, Loveday C, Martin-Gimeno P, Meijer C, Nsengimana J, Quinn DI, Rafnar T, Ramdas S, Richiardi L, Skotheim RI, Stefansson K, Turnbull C, Vaughn DJ, Wiklund F, Wu X, Yang D, Zheng T, Wells AD, Grant SFA, Rajpert-De Meyts E, Schwartz SM, Bishop DT, McGlynn KA, Kanetsky PA, Nathanson KL. Identification of 22 susceptibility loci associated with testicular germ cell tumors. Nat Commun 2021; 12:4487. [PMID: 34301922 PMCID: PMC8302763 DOI: 10.1038/s41467-021-24334-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Testicular germ cell tumors (TGCT) are the most common tumor in young white men and have a high heritability. In this study, the international Testicular Cancer Consortium assemble 10,156 and 179,683 men with and without TGCT, respectively, for a genome-wide association study. This meta-analysis identifies 22 TGCT susceptibility loci, bringing the total to 78, which account for 44% of disease heritability. Men with a polygenic risk score (PRS) in the 95th percentile have a 6.8-fold increased risk of TGCT compared to men with median scores. Among men with independent TGCT risk factors such as cryptorchidism, the PRS may guide screening decisions with the goal of reducing treatment-related complications causing long-term morbidity in survivors. These findings emphasize the interconnected nature of two known pathways that promote TGCT susceptibility: male germ cell development within its somatic niche and regulation of chromosomal division and structure, and implicate an additional biological pathway, mRNA translation.
Collapse
Affiliation(s)
- John Pluta
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Louise C Pyle
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kevin T Nead
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rona Wilf
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benita Weathers
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt D'Andrea
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristian Almstrup
- Department of Growth and Reproduction, Rigshospitalet, Copenhagen, Denmark
| | - Lynn Anson-Cartwright
- Department of Surgery (Urology), University of Toronto and The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Javier Benitez
- Human Genetics Group, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Christopher D Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Chu Chen
- Program in Epidemiology, Fred Hutchinson Cancer Research Center; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Victoria K Cortessis
- Departments of Preventive Medicine and Obstetrics and Gynecology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Alberto Ferlin
- Unit of Endocrinology and Metabolism, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Carlo Foresta
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Marija Gamulin
- Department of Oncology, Division of Medical Oncology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Chiara Grasso
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin and CPO-Piemonte, Turin, Italy
| | - Mark H Greene
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Tom Grotmol
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Robert J Hamilton
- Department of Surgery (Urology), University of Toronto and The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Trine B Haugen
- Faculty of Health Sciences, OsloMet-Oslo Metropolitan University, Oslo, Norway
| | - Russ Hauser
- Department of Environmental Health, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Matthew E Johnson
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jennifer T Loud
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Chey Loveday
- Division of Genetics & Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Coby Meijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jérémie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - David I Quinn
- Division of Oncology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | | | - Shweta Ramdas
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenzo Richiardi
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin and CPO-Piemonte, Turin, Italy
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | | | - Clare Turnbull
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- William Harvey Research Institute, Queen Mary University, London, UK
| | - David J Vaughn
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Xifeng Wu
- School of Public Health, Zhejiang University, Zhejiang, China
| | - Daphne Yang
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tongzhang Zheng
- Department of Epidemiology, Brown School of Public Health, Brown University, Providence, RI, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F A Grant
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Stephen M Schwartz
- Program in Epidemiology, Fred Hutchinson Cancer Research Center; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - D Timothy Bishop
- Department of Haematology and Immunology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, Clinical Genetics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Katherine L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Lindeman RE, Murphy MW, Agrimson KS, Gewiss R, Bardwell V, Gearhart M, Zarkower D. The conserved sex regulator DMRT1 recruits SOX9 in sexual cell fate reprogramming. Nucleic Acids Res 2021; 49:6144-6164. [PMID: 34096593 PMCID: PMC8216462 DOI: 10.1093/nar/gkab448] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/30/2021] [Accepted: 05/17/2021] [Indexed: 01/16/2023] Open
Abstract
Mammalian sexual development commences when fetal bipotential progenitor cells adopt male Sertoli (in XY) or female granulosa (in XX) gonadal cell fates. Differentiation of these cells involves extensive divergence in chromatin state and gene expression, reflecting distinct roles in sexual differentiation and gametogenesis. Surprisingly, differentiated gonadal cell fates require active maintenance through postnatal life to prevent sexual transdifferentiation and female cell fate can be reprogrammed by ectopic expression of the sex regulator DMRT1. Here we examine how DMRT1 reprograms granulosa cells to Sertoli-like cells in vivo and in culture. We define postnatal sex-biased gene expression programs and identify three-dimensional chromatin contacts and differentially accessible chromatin regions (DARs) associated with differentially expressed genes. Using a conditional transgene we find DMRT1 only partially reprograms the ovarian transcriptome in the absence of SOX9 and its paralog SOX8, indicating that these factors functionally cooperate with DMRT1. ATAC-seq and ChIP-seq show that DMRT1 induces formation of many DARs that it binds with SOX9, and DMRT1 is required for binding of SOX9 at most of these. We suggest that DMRT1 can act as a pioneer factor to open chromatin and allow binding of SOX9, which then cooperates with DMRT1 to reprogram sexual cell fate.
Collapse
Affiliation(s)
- Robin E Lindeman
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Mark W Murphy
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Kellie S Agrimson
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Rachel L Gewiss
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Vivian J Bardwell
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Micah D Gearhart
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - David Zarkower
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| |
Collapse
|
11
|
Naseem M, Cao S, Yang D, Millstein J, Puccini A, Loupakis F, Stintzing S, Cremolini C, Tokunaga R, Battaglin F, Soni S, Berger MD, Barzi A, Zhang W, Falcone A, Heinemann V, Lenz HJ. Random survival forests identify pathways with polymorphisms predictive of survival in KRAS mutant and KRAS wild-type metastatic colorectal cancer patients. Sci Rep 2021; 11:12191. [PMID: 34108518 PMCID: PMC8190302 DOI: 10.1038/s41598-021-91330-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/20/2021] [Indexed: 12/22/2022] Open
Abstract
KRAS status serves as a predictive biomarker of response to treatment in metastatic colorectal cancer (mCRC). We hypothesize that complex interactions between multiple pathways contribute to prognostic differences between KRAS wild-type and KRAS mutant patients with mCRC, and aim to identify polymorphisms predictive of clinical outcomes in this subpopulation. Most pathway association studies are limited in assessing gene–gene interactions and are restricted to an individual pathway. In this study, we use a random survival forests (RSF) method for identifying predictive markers of overall survival (OS) and progression-free survival (PFS) in mCRC patients treated with FOLFIRI/bevacizumab. A total of 486 mCRC patients treated with FOLFIRI/bevacizumab from two randomized phase III trials, TRIBE and FIRE-3, were included in the current study. Two RSF approaches were used, namely variable importance and minimal depth. We discovered that Wnt/β-catenin and tumor associated macrophage pathway SNPs are strong predictors of OS and PFS in mCRC patients treated with FOLFIRI/bevacizumab independent of KRAS status, whereas a SNP in the sex-differentiation pathway gene, DMRT1, is strongly predictive of OS and PFS in KRAS mutant mCRC patients. Our results highlight RSF as a useful method for identifying predictive SNPs in multiple pathways.
Collapse
Affiliation(s)
- Madiha Naseem
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Shu Cao
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dongyun Yang
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joshua Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alberto Puccini
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Fotios Loupakis
- Oncologia Medica 1, Istituto Oncologico Veneto, Istituto Di Ricovero E Cura a Carattere Scientifico, Via Gattamelata, Padua, Italy
| | - Sebastian Stintzing
- Medical Department, Division of Hematology, Oncology and Hematology, Tumor Immunology (CCM), Charité-Universitätsmedizin, Berlin, Germany
| | - Chiara Cremolini
- Oncologia Medica, Azienda Ospedaliero-Universitaria Pisana, Istituto Toscano Tumori, Via Roma, Pisa, Italy
| | - Ryuma Tokunaga
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Francesca Battaglin
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA.,Oncologia Medica 1, Istituto Oncologico Veneto, Istituto Di Ricovero E Cura a Carattere Scientifico, Via Gattamelata, Padua, Italy
| | - Shivani Soni
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Martin D Berger
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Afsaneh Barzi
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Wu Zhang
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Alfredo Falcone
- Oncologia Medica, Azienda Ospedaliero-Universitaria Pisana, Istituto Toscano Tumori, Via Roma, Pisa, Italy
| | - Volker Heinemann
- Department of Medicine and Comprehensive Cancer Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Sharon Carpenter Laboratory, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA.
| |
Collapse
|
12
|
Nicholls PK, Page DC. Germ cell determination and the developmental origin of germ cell tumors. Development 2021; 148:239824. [PMID: 33913479 DOI: 10.1242/dev.198150] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In each generation, the germline is tasked with producing somatic lineages that form the body, and segregating a population of cells for gametogenesis. During animal development, when do cells of the germline irreversibly commit to producing gametes? Integrating findings from diverse species, we conclude that the final commitment of the germline to gametogenesis - the process of germ cell determination - occurs after primordial germ cells (PGCs) colonize the gonads. Combining this understanding with medical findings, we present a model whereby germ cell tumors arise from cells that failed to undertake germ cell determination, regardless of their having colonized the gonads. We propose that the diversity of cell types present in these tumors reflects the broad developmental potential of migratory PGCs.
Collapse
Affiliation(s)
- Peter K Nicholls
- Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA.,Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - David C Page
- Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| |
Collapse
|
13
|
Adolfi MC, Herpin A, Martinez-Bengochea A, Kneitz S, Regensburger M, Grunwald DJ, Schartl M. Crosstalk Between Retinoic Acid and Sex-Related Genes Controls Germ Cell Fate and Gametogenesis in Medaka. Front Cell Dev Biol 2021; 8:613497. [PMID: 33537305 PMCID: PMC7848095 DOI: 10.3389/fcell.2020.613497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Sex determination (SD) is a highly diverse and complex mechanism. In vertebrates, one of the first morphological differences between the sexes is the timing of initiation of the first meiosis, where its initiation occurs first in female and later in male. Thus, SD is intimately related to the responsiveness of the germ cells to undergo meiosis in a sex-specific manner. In some vertebrates, it has been reported that the timing for meiosis entry would be under control of retinoic acid (RA), through activation of Stra8. In this study, we used a fish model species for sex determination and lacking the stra8 gene, the Japanese medaka (Oryzias latipes), to investigate the connection between RA and the sex determination pathway. Exogenous RA treatments act as a stress factor inhibiting germ cell differentiation probably by activation of dmrt1a and amh. Disruption of the RA degrading enzyme gene cyp26a1 induced precocious meiosis and oogenesis in embryos/hatchlings of female and even some males. Transcriptome analyzes of cyp26a1–/–adult gonads revealed upregulation of genes related to germ cell differentiation and meiosis, in both ovaries and testes. Our findings show that germ cells respond to RA in a stra8 independent model species. The responsiveness to RA is conferred by sex-related genes, restricting its action to the sex differentiation period in both sexes.
Collapse
Affiliation(s)
- Mateus C Adolfi
- University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany
| | - Amaury Herpin
- INRA, UR1037, Fish Physiology and Genomics, Rennes, France.,State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Anabel Martinez-Bengochea
- University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany.,Reproductive and Molecular Biology Group, Department of Morphology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, Brazil
| | - Susanne Kneitz
- University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany
| | - Martina Regensburger
- University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany
| | - David J Grunwald
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
| | - Manfred Schartl
- University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany.,University of Wuerzburg, Developmental Biochemistry, Biocenter, Wuerzburg, Germany
| |
Collapse
|
14
|
Yin H, Zhou C, Shi S, Fang L, Liu J, Sun D, Jiang L, Zhang S. Weighted Single-Step Genome-Wide Association Study of Semen Traits in Holstein Bulls of China. Front Genet 2019; 10:1053. [PMID: 31749837 PMCID: PMC6842931 DOI: 10.3389/fgene.2019.01053] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Efficient production of high-quality semen is a crucial trait in the dairy cattle breeding due to the widespread use of artificial insemination. However, the genetic architecture (e.g., distributions of causal variants and their corresponding effects) underlying such semen quality traits remains unclear. In this study, we performed genome-wide association studies to identify genes associated with five semen quality traits in Chinese Holstein population, including ejaculate volume, progressive sperm motility, sperm concentration, number of sperm, and number of progressive motile sperm. Our dataset consisted of 2,218 Holstein bulls in China with full pedigree information, representing 12 artificial insemination centers, with 1,508 genotyped using the Illumina BovineSNP50 BeadChip. We used a weighted single-step genome-wide association method with 10 adjacent Single nucleotide polymorphisms (SNPs) as sliding windows, which can make use of individuals without genotypes. We considered the top 10 genomic regions in terms of their explained genomic variants as candidate window regions for each trait. In total, we detected 36 window regions related to one or multiple semen traits across 19 chromosomes. Promising candidate genes of PSMB5, PRMT5, ACTB, PDE3A, NPC1, FSCN1, NR5A2, IQCG, LHX8, and DMRT1 were identified in these window regions for these five semen traits. Our findings provided a solid basis for further research into genetic mechanisms underlying semen quality traits, which may contribute to their accurate genomic prediction in Chinese Holstein population.
Collapse
Affiliation(s)
- Hongwei Yin
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chenghao Zhou
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shaolei Shi
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lingzhao Fang
- Department of Animal and Avian Sciences, University of Maryland, College Park, College Park, MD, United States
| | - Jianfeng Liu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dongxiao Sun
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Li Jiang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shengli Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Andries V, De Keuckelaere E, Staes K, Hochepied T, Taminau J, Lemeire K, Birembaut P, Berx G, van Roy F. A new mouse model to study the role of ectopic Nanos3 expression in cancer. BMC Cancer 2019; 19:598. [PMID: 31208373 PMCID: PMC6580527 DOI: 10.1186/s12885-019-5807-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND NANOS3 is a gene conserved throughout evolution. Despite the quite low conservation of Nanos sequences between different organisms and even between Nanos paralogs, their role in germ cell development is remarkably universal. Human Nanos3 expression is normally restricted to the gonads and the brain. However, ectopic activation of this gene has been detected in various human cancers. Until now, Nanos3 and other Nanos proteins have been studied almost exclusively in germ cell development. METHODS Transgenic mice were generated by targeted insertion of a human Nanos3 cDNA into the ROSA26 locus. The transgene could be spatiotemporally induced by Cre recombinase activity removing an upstream floxed STOP cassette. A lung tumor model with ectopic Nanos3 expression was based on the lung-specific activation of the reverse tetracycline transactivator gene, in combination with a tetO-CMV promoter controlling Cre expression. When doxycycline was provided to the mice, Cre was activated leading to deletion of TP53 alleles and activation of both oncogenic KRasG12D and Nanos3. Appropriate controls were foreseen. Tumors and tumor-derived cell cultures were analyzed in various ways. RESULTS We describe the successful generation of Nanos3LSL/- and Nanos3LSL/LSL mice in which an exogenous human NANOS3 gene can be activated in vivo upon Cre expression. These mice, in combination with different conditional and doxycycline-inducible Cre lines, allow the study of the role of ectopic Nanos3 expression in several cancer types. The Nanos3LSL mice were crossed with a non-small cell lung cancer (NSCLC) mouse model based on conditional expression of oncogenic KRas and homozygous loss of p53. This experiment demonstrated that ectopic expression of Nanos3 in the lungs has a significant negative effect on survival. Enhanced bronchiolar dysplasia was observed when Nanos3-expressing NSCLC mice were compared with control NSCLC mice. An allograft experiment, performed with cell cultures derived from primary lung tumors of control and Nanos3-expressing NSCLC mice, revealed lymph node metastasis in mice injected with Nanos3-expressing NSCLC cells. CONCLUSIONS A new mouse model was generated allowing examination of Nanos3-associated pathways and investigation of the influence of ectopic Nanos3 expression in various cancer types. This model might identify Nanos3 as an interesting target in cancer therapeutics.
Collapse
Affiliation(s)
- Vanessa Andries
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Evi De Keuckelaere
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Katrien Staes
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Tino Hochepied
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Joachim Taminau
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kelly Lemeire
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Philippe Birembaut
- INSERM UMRS 1250, Department of Biopathology, CHU Maison-Blanche, University Hospital of Reims & University of Reims Champagne-Ardenne, rue Cognacq-Jay 45, 51092, Reims, France
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Frans van Roy
- VIB-UGent Center for Inflammation Research (IRC), Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
| |
Collapse
|
16
|
Nicol B, Grimm SA, Gruzdev A, Scott GJ, Ray MK, Yao HHC. Genome-wide identification of FOXL2 binding and characterization of FOXL2 feminizing action in the fetal gonads. Hum Mol Genet 2019; 27:4273-4287. [PMID: 30212841 DOI: 10.1093/hmg/ddy312] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
The identity of the gonads is determined by which fate, ovarian granulosa cell or testicular Sertoli cell, the bipotential somatic cell precursors choose to follow. In most vertebrates, the conserved transcription factor FOXL2 contributes to the fate of granulosa cells. To understand FOXL2 functions during gonad differentiation, we performed genome-wide analysis of FOXL2 chromatin occupancy in fetal ovaries and established a genetic mouse model that forces Foxl2 expression in the fetal testis. When FOXL2 was ectopically expressed in the somatic cell precursors in the fetal testis, FOXL2 was sufficient to repress Sertoli cell differentiation, ultimately resulting in partial testis-to-ovary sex-reversal. Combining genome-wide analysis of FOXL2 binding in the fetal ovary with transcriptomic analyses of our Foxl2 gain-of-function and previously published Foxl2 loss-of-function models, we identified potential pathways responsible for the feminizing action of FOXL2. Finally, comparison of FOXL2 genome-wide occupancy in the fetal ovary with testis-determining factor SOX9 genome-wide occupancy in the fetal testis revealed extensive overlaps, implying that antagonistic signals between FOXL2 and SOX9 occur at the chromatin level.
Collapse
Affiliation(s)
- Barbara Nicol
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Greg J Scott
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Manas K Ray
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
17
|
Inoue M, Baba T, Morohashi KI. Recent progress in understanding the mechanisms of Leydig cell differentiation. Mol Cell Endocrinol 2018; 468:39-46. [PMID: 29309805 DOI: 10.1016/j.mce.2017.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/25/2017] [Accepted: 12/26/2017] [Indexed: 01/26/2023]
Abstract
Leydig cells in fetal and adult testes play pivotal roles in eliciting male characteristics by producing androgen. Although numerous studies of Leydig cells have been performed, the mechanisms for differentiation of the two cell types (fetal Leydig and adult Leydig cells), their developmental and functional relationship, and their differential characteristics remain largely unclear. Based on recent technical progress in genome-wide analysis and in vitro investigation, novel and fascinating observations concerning the issues above have been obtained. Focusing on fetal and adult Leydig cells, this review summarizes the recent progress that has advanced our understanding of the cells.
Collapse
Affiliation(s)
- Miki Inoue
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Baba
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-Ichirou Morohashi
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
18
|
De Keuckelaere E, Hulpiau P, Saeys Y, Berx G, van Roy F. Nanos genes and their role in development and beyond. Cell Mol Life Sci 2018; 75:1929-1946. [PMID: 29397397 PMCID: PMC11105394 DOI: 10.1007/s00018-018-2766-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
The hallmark of Nanos proteins is their typical (CCHC)2 zinc finger motif (zf-nanos). Animals have one to four nanos genes. For example, the fruit fly and demosponge have only one nanos gene, zebrafish and humans have three, and Fugu rubripes has four. Nanos genes are mainly known for their evolutionarily preserved role in germ cell survival and pluripotency. Nanos proteins have been reported to bind the C-terminal RNA-binding domain of Pumilio to form a post-transcriptional repressor complex. Several observations point to a link between the miRNA-mediated repression complex and the Nanos/Pumilio complex. Repression of the E2F3 oncogene product is, indeed, mediated by cooperation between the Nanos/Pumilio complex and miRNAs. Another important interaction partner of Nanos is the CCR4-NOT deadenylase complex. Besides the tissue-specific contribution of Nanos proteins to normal development, their ectopic expression has been observed in several cancer cell lines and various human cancers. An inverse correlation between the expression levels of human Nanos1 and Nanos3 and E-cadherin was observed in several cancer cell lines. Loss of E-cadherin, an important cell-cell adhesion protein, contributes to tumor invasion and metastasis. Overexpression of Nanos3 induces epithelial-mesenchymal transition in lung cancer cell lines partly by repressing E-cadherin. Other than some most interesting data from Nanos knockout mice, little is known about mammalian Nanos proteins, and further research is needed. In this review, we summarize the main roles of Nanos proteins and discuss the emerging concept of Nanos proteins as oncofetal antigens.
Collapse
Affiliation(s)
- Evi De Keuckelaere
- VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052, Ghent, Belgium
- Molecular Cell Biology Unit, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Paco Hulpiau
- VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052, Ghent, Belgium
- Molecular Cell Biology Unit, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Yvan Saeys
- VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Krijgslaan 281, S9, 9000, Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Frans van Roy
- VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052, Ghent, Belgium.
- Molecular Cell Biology Unit, Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
| |
Collapse
|
19
|
Salz HK, Dawson EP, Heaney JD. Germ cell tumors: Insights from the Drosophila ovary and the mouse testis. Mol Reprod Dev 2017; 84:200-211. [PMID: 28079292 DOI: 10.1002/mrd.22779] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022]
Abstract
Ovarian and testicular germ cell tumors of young adults are thought to arise from defects in germ cell development, but the molecular mechanisms underlying malignant transformation are poorly understood. In this review, we focus on the biology of germ cell tumor formation in the Drosophila ovary and the mouse testis, for which evidence supports common underlying mechanisms, such as blocking initiation into the differentiation pathway, impaired lineage progression, and sexual identity instability. We then discuss how these concepts inform our understanding of the disease in humans. Mol. Reprod. Dev. 84: 200-211, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Helen K Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Emily P Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
20
|
Facchini G, Rossetti S, Cavaliere C, D'Aniello C, Di Franco R, Iovane G, Grimaldi G, Piscitelli R, Muto P, Botti G, Perdonà S, Veneziani BM, Berretta M, Montanari M. Exploring the molecular aspects associated with testicular germ cell tumors: a review. Oncotarget 2017; 9:1365-1379. [PMID: 29416701 PMCID: PMC5787445 DOI: 10.18632/oncotarget.22373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/05/2017] [Indexed: 11/25/2022] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most common solid tumors affecting young men. They constitute a distinct entity because of their embryonic origin and their unique biological behavior. Recent preclinical data regarding biological signaling machinery as well as genetic and epigenetic mechanisms associated with molecular patterns of tumors have contribute to explain the pathogenesis and the differentiation of TGCTs and to understand the mechanisms responsible for the development of resistance to treatment. In this review, we discuss the main genetic and epigenetic events associated with TGCTs development in order to better define their role in the pathogenesis of these tumors and in cisplatin-acquired resistance.
Collapse
Affiliation(s)
- Gaetano Facchini
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Carla Cavaliere
- Medical Oncology Unit, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Naples, Italy
| | - Rossella Di Franco
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gelsomina Iovane
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,S.S.D Oncologia Clinica Sperimentale Uro-Andrologica, Dipartimento Corp-S Assistenziale dei Percorsi Oncologici Uro-Genitale, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Giovanni Grimaldi
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale", IRCCS, Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy.,Scientific Management, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Sisto Perdonà
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, Aviano, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0, Linea Progettuale 14 per l'Implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy.,Department of Biology, College of Science and Technology, Temple University, Philadelphia, USA
| |
Collapse
|
21
|
Nettersheim D, Jostes S, Schneider S, Schorle H. Elucidating human male germ cell development by studying germ cell cancer. Reproduction 2017; 152:R101-13. [PMID: 27512122 DOI: 10.1530/rep-16-0114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/07/2016] [Indexed: 12/19/2022]
Abstract
Human germ cell development is regulated in a spatio-temporal manner by complex regulatory networks. Here, we summarize results obtained in germ cell tumors and respective cell lines and try to pinpoint similarities to normal germ cell development. This comparison allows speculating about the critical and error-prone mechanisms, which when disturbed, lead to the development of germ cell tumors. Short after specification, primordial germ cells express markers of pluripotency, which, in humans, persists up to the stage of fetal/infantile spermatogonia. Aside from the rare spermatocytic tumors, virtually all seminomas and embryonal carcinomas express markers of pluripotency and show signs of pluripotency or totipotency. Therefore, it appears that proper handling of the pluripotency program appears to be the most critical step in germ cell development in terms of tumor biology. Furthermore, data from mice reveal that germline cells display an epigenetic signature, which is highly similar to pluripotent cells. This signature (poised histone code, DNA hypomethylation) is required for the rapid induction of toti- and pluripotency upon fertilization. We propose that adult spermatogonial cells, when exposed to endocrine disruptors or epigenetic active substances, are prone to reinitiate the pluripotency program, giving rise to a germ cell tumor. The fact that pluripotent cells can be derived from adult murine and human testicular cells further corroborates this idea.
Collapse
Affiliation(s)
- Daniel Nettersheim
- Department of Developmental PathologyInstitute of Pathology, University of Bonn Medical School, Bonn, Germany
| | - Sina Jostes
- Department of Developmental PathologyInstitute of Pathology, University of Bonn Medical School, Bonn, Germany
| | - Simon Schneider
- Department of Developmental PathologyInstitute of Pathology, University of Bonn Medical School, Bonn, Germany
| | - Hubert Schorle
- Department of Developmental PathologyInstitute of Pathology, University of Bonn Medical School, Bonn, Germany
| |
Collapse
|
22
|
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: 68] [Impact Index Per Article: 9.7] [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
|
23
|
Zhang T, Zarkower D. DMRT proteins and coordination of mammalian spermatogenesis. Stem Cell Res 2017; 24:195-202. [PMID: 28774758 DOI: 10.1016/j.scr.2017.07.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 10/19/2022] Open
Abstract
DMRT genes encode a deeply conserved family of transcription factors that share a unique DNA binding motif, the DM domain. DMRTs regulate development in a broad variety of metazoans and they appear to have controlled sexual differentiation for hundreds of millions of years. In mice, starting during embryonic development, three Dmrt genes act sequentially to help establish and maintain spermatogenesis. Dmrt1 has notably diverse functions that include repressing pluripotency genes and promoting mitotic arrest in embryonic germ cells, reactivating prospermatogonia perinatally, establishing and maintaining spermatogonial stem cells (SSCs), promoting spermatogonial differentiation, and controlling the mitosis/meiosis switch. Dmrt6 acts in differentiating spermatogonia to coordinate an orderly exit from the mitotic/spermatogonial program and allow proper timing of entry to the meiotic/spermatocyte program. Finally, Dmrt7 takes over during the first meiotic prophase to help choreograph a transition in histone modifications that maintains transcriptional silencing of the sex chromosomes. The combined action of these three Dmrt genes helps ensure robust and sustainable spermatogenesis.
Collapse
Affiliation(s)
- Teng Zhang
- Department of Genetics, Cell Biology, and Development, and Developmental Biology Center, University of Minnesota Medical School, 6-160 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA.
| | - David Zarkower
- Department of Genetics, Cell Biology, and Development, and Developmental Biology Center, University of Minnesota Medical School, 6-160 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA.
| |
Collapse
|
24
|
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: 3.7] [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
|
25
|
Reprogramming cell fate with a genome-scale library of artificial transcription factors. Proc Natl Acad Sci U S A 2016; 113:E8257-E8266. [PMID: 27930301 DOI: 10.1073/pnas.1611142114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Artificial transcription factors (ATFs) are precision-tailored molecules designed to bind DNA and regulate transcription in a preprogrammed manner. Libraries of ATFs enable the high-throughput screening of gene networks that trigger cell fate decisions or phenotypic changes. We developed a genome-scale library of ATFs that display an engineered interaction domain (ID) to enable cooperative assembly and synergistic gene expression at targeted sites. We used this ATF library to screen for key regulators of the pluripotency network and discovered three combinations of ATFs capable of inducing pluripotency without exogenous expression of Oct4 (POU domain, class 5, TF 1). Cognate site identification, global transcriptional profiling, and identification of ATF binding sites reveal that the ATFs do not directly target Oct4; instead, they target distinct nodes that converge to stimulate the endogenous pluripotency network. This forward genetic approach enables cell type conversions without a priori knowledge of potential key regulators and reveals unanticipated gene network dynamics that drive cell fate choices.
Collapse
|
26
|
Canovas S, Campos R, Aguilar E, Cibelli JB. Progress towards human primordial germ cell specification in vitro. Mol Hum Reprod 2016; 23:4-15. [PMID: 27798275 DOI: 10.1093/molehr/gaw069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/28/2016] [Indexed: 12/13/2022] Open
Abstract
Primordial germ cells (PGCs) have long been considered the link between one generation and the next. PGC specification begins in the early embryo as a result of a highly orchestrated combination of transcriptional and epigenetic mechanisms. Understanding the molecular events that lead to proper PGC development will facilitate the development of new treatments for human infertility as well as species conservation. This article describes the latest, most relevant findings about the mechanisms of PGC formation, emphasizing human PGC. It also discusses our own laboratory's progress in using transdifferentiation protocols to derive human PGCs (hPGCs). Our preliminary results arose from our pursuit of a sequential hPGC induction strategy that starts with the repression of lineage-specific factors in the somatic cell, followed by the reactivation of germ cell-related genes using specific master regulators, which can indeed reactivate germ cell-specific genes in somatic cells. While it is still premature to assume that fully functional human gametes can be obtained in a dish, our results, together with those recently published by others, provide strong evidence that generating their precursors, PGCs, is within reach.
Collapse
Affiliation(s)
- S Canovas
- LARCEL, Centro Andaluz de Nanomedicina y Biotecnología (BIONAND), C/Severo Ochoa 35, Malaga 29590, Spain
| | - R Campos
- LARCEL, Centro Andaluz de Nanomedicina y Biotecnología (BIONAND), C/Severo Ochoa 35, Malaga 29590, Spain
| | - E Aguilar
- LARCEL, Centro Andaluz de Nanomedicina y Biotecnología (BIONAND), C/Severo Ochoa 35, Malaga 29590, Spain
| | - J B Cibelli
- LARCEL, Centro Andaluz de Nanomedicina y Biotecnología (BIONAND), C/Severo Ochoa 35, Malaga 29590, Spain .,Department of Physiology and Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
27
|
Zhang T, Oatley J, Bardwell VJ, Zarkower D. DMRT1 Is Required for Mouse Spermatogonial Stem Cell Maintenance and Replenishment. PLoS Genet 2016; 12:e1006293. [PMID: 27583450 PMCID: PMC5008761 DOI: 10.1371/journal.pgen.1006293] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/10/2016] [Indexed: 01/15/2023] Open
Abstract
Male mammals produce sperm for most of postnatal life and therefore require a robust germ line stem cell system, with precise balance between self-renewal and differentiation. Prior work established doublesex- and mab-3-related transcription factor 1 (Dmrt1) as a conserved transcriptional regulator of male sexual differentiation. Here we investigate the role of Dmrt1 in mouse spermatogonial stem cell (SSC) homeostasis. We find that Dmrt1 maintains SSCs during steady state spermatogenesis, where it regulates expression of Plzf, another transcription factor required for SSC maintenance. We also find that Dmrt1 is required for recovery of spermatogenesis after germ cell depletion. Committed progenitor cells expressing Ngn3 normally do not contribute to SSCs marked by the Id4-Gfp transgene, but do so when spermatogonia are chemically depleted using busulfan. Removal of Dmrt1 from Ngn3-positive germ cells blocks the replenishment of Id4-GFP-positive SSCs and recovery of spermatogenesis after busulfan treatment. Our data therefore reveal that Dmrt1 supports SSC maintenance in two ways: allowing SSCs to remain in the stem cell pool under normal conditions; and enabling progenitor cells to help restore the stem cell pool after germ cell depletion. The Dmrt1 gene is a deeply conserved gonadal regulator that is expressed in all mitotic germ cells of the mouse, including spermatogonial stem cells (SSCs). We previously showed that Dmrt1 controls the mitosis/meiosis switch in differentiating mouse spermatogonia. Here we have examined the role of Dmrt1 in undifferentiated spermatogonia and found that Dmrt1 plays two crucial roles in sustaining the population of SSCs. First, Dmrt1 is required to maintain the SSC pool during normal conditions: loss of Dmrt1 in SSCs causes loss of the SSC maintenance factor PLZF and differentiation of SSCs. This result suggests that Dmrt1 is necessary for SSC self-renewal. Second, Dmrt1 is required to replenish SSCs after germ line depletion. We found that Ngn3-positive transit amplifying cells normally do not contribute to Id4-positive SSCs, but can do so when germ cells are chemically depleted by busulfan treatment. However, when Dmrt1 is lost in committed progenitor cells the ability to replenish SSCs after cytotoxic stress is completely lost. Our results suggest that Dmrt1 is important for SSC homeostasis and may provide new avenues for SSC manipulation.
Collapse
Affiliation(s)
- Teng Zhang
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jon Oatley
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Vivian J. Bardwell
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota, United States of America
| | - David Zarkower
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
28
|
Sekita Y, Nakamura T, Kimura T. Reprogramming of germ cells into pluripotency. World J Stem Cells 2016; 8:251-259. [PMID: 27621759 PMCID: PMC4999652 DOI: 10.4252/wjsc.v8.i8.251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/08/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023] Open
Abstract
Primordial germ cells (PGCs) are precursors of all gametes, and represent the founder cells of the germline. Although developmental potency is restricted to germ-lineage cells, PGCs can be reprogrammed into a pluripotent state. Specifically, PGCs give rise to germ cell tumors, such as testicular teratomas, in vivo, and to pluripotent stem cells known as embryonic germ cells in vitro. In this review, we highlight the current knowledge on signaling pathways, transcriptional controls, and post-transcriptional controls that govern germ cell differentiation and de-differentiation. These regulatory processes are common in the reprogramming of germ cells and somatic cells, and play a role in the pathogenesis of human germ cell tumors.
Collapse
|
29
|
Lanza DG, Dawson EP, Rao P, Heaney JD. Misexpression of cyclin D1 in embryonic germ cells promotes testicular teratoma initiation. Cell Cycle 2016; 15:919-30. [PMID: 26901436 DOI: 10.1080/15384101.2016.1149272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Testicular teratomas result from anomalies in embryonic germ cell development. In the 129 family of inbred mouse strains, teratomas arise during the same developmental period that male germ cells normally enter G1/G0 mitotic arrest and female germ cells initiate meiosis (the mitotic:meiotic switch). Dysregulation of this switch associates with teratoma susceptibility and involves three germ cell developmental abnormalities seemingly critical for tumor initiation: delayed G1/G0 mitotic arrest, retention of pluripotency, and misexpression of genes normally restricted to embryonic female and adult male germ cells. One misexpressed gene, cyclin D1 (Ccnd1), is a known regulator of cell cycle progression and an oncogene in many tissues. Here, we investigated whether Ccnd1 misexpression in embryonic germ cells is a determinant of teratoma susceptibility in mice. We found that CCND1 localizes to teratoma-susceptible germ cells that fail to enter G1/G0 arrest during the mitotic:meiotic switch and is the only D-type cyclin misexpressed during this critical developmental time frame. We discovered that Ccnd1 deficiency in teratoma-susceptible mice significantly reduced teratoma incidence and suppressed the germ cell proliferation and pluripotency abnormalities associated with tumor initiation. Importantly, Ccnd1 expression was dispensable for somatic cell development and male germ cell specification and maturation in tumor-susceptible mice, implying that the mechanisms by which Ccnd1 deficiency reduced teratoma incidence were germ cell autonomous and specific to tumorigenesis. We conclude that misexpression of Ccnd1 in male germ cells is a key component of a larger pro-proliferative program that disrupts the mitotic:meiotic switch and predisposes 129 inbred mice to testicular teratocarcinogenesis.
Collapse
Affiliation(s)
- Denise G Lanza
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA
| | - Emily P Dawson
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA
| | - Priya Rao
- b Department of Pathology , MD Anderson Cancer Center, The University of Texas , Houston , TX , USA
| | - Jason D Heaney
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,c Dan L Duncan Cancer Center, Baylor College of Medicine , Houston , TX , USA.,d Center For Reproductive Medicine, Baylor College of Medicine , Houston , TX , USA
| |
Collapse
|
30
|
Sanchez A, Amatruda JF. Zebrafish Germ Cell Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:479-94. [PMID: 27165367 DOI: 10.1007/978-3-319-30654-4_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Germ cell tumors (GCTs) are malignant cancers that arise from embryonic precursors known as Primordial Germ Cells. GCTs occur in neonates, children, adolescents and young adults and can occur in the testis, the ovary or extragonadal sites. Because GCTs arise from pluripotent cells, the tumors can exhibit a wide range of different histologies. Current cisplatin-based combination therapies cures most patients, however at the cost of significant toxicity to normal tissues. While GWAS studies and genomic analysis of human GCTs have uncovered somatic mutations and loci that might confer tumor susceptibility, little is still known about the exact mechanisms that drive tumor development, and animal models that faithfully recapitulate all the different GCT subtypes are lacking. Here, we summarize current understanding of germline development in humans and zebrafish, describe the biology of human germ cell tumors, and discuss progress and prospects for zebrafish GCT models that may contribute to better understanding of human GCTs.
Collapse
Affiliation(s)
- Angelica Sanchez
- Departments of Pediatrics and Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - James F Amatruda
- Departments of Pediatrics, Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
31
|
Kjartansdóttir KR, Reda A, Panula S, Day K, Hultenby K, Söder O, Hovatta O, Stukenborg JB. A Combination of Culture Conditions and Gene Expression Analysis Can Be Used to Investigate and Predict hES Cell Differentiation Potential towards Male Gonadal Cells. PLoS One 2015; 10:e0144029. [PMID: 26630562 PMCID: PMC4667967 DOI: 10.1371/journal.pone.0144029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/12/2015] [Indexed: 12/12/2022] Open
Abstract
Human embryonic stem cell differentiation towards various cell types belonging to ecto-, endo- and mesodermal cell lineages has been demonstrated, with high efficiency rates using standardized differentiation protocols. However, germ cell differentiation from human embryonic stem cells has been very inefficient so far. Even though the influence of various growth factors has been evaluated, the gene expression of different cell lines in relation to their differentiation potential has not yet been extensively examined. In this study, the potential of three male human embryonic stem cell lines to differentiate towards male gonadal cells was explored by analysing their gene expression profiles. The human embryonic stem cell lines were cultured for 14 days as monolayers on supporting human foreskin fibroblasts or as spheres in suspension, and were differentiated using BMP7, or spontaneous differentiation by omitting exogenous FGF2. TLDA analysis revealed that in the undifferentiated state, these cell lines have diverse mRNA profiles and exhibit significantly different potentials for differentiation towards the cell types present in the male gonads. This potential was associated with important factors directing the fate of the male primordial germ cells in vivo to form gonocytes, such as SOX17 or genes involved in the NODAL/ACTIVIN pathway, for example. Stimulation with BMP7 in suspension culture resulted in up-regulation of cytoplasmic SOX9 protein expression in all three lines. The observation that human embryonic stem cells differentiate towards germ and somatic cells after spontaneous and BMP7-induced stimulation in suspension emphasizes the important role of somatic cells in germ cell differentiation in vitro.
Collapse
Affiliation(s)
- Kristín Rós Kjartansdóttir
- Department of Women’s and Children’s Health, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-171 76, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, SE-141 86, Huddinge, Sweden
- Laboratory of Reproductive Biology, Scientific Unit, Horsens Hospital, DK-8700, Horsens, Denmark
| | - Ahmed Reda
- Department of Women’s and Children’s Health, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-171 76, Stockholm, Sweden
| | - Sarita Panula
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, SE-141 86, Huddinge, Sweden
| | - Kelly Day
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, SE-141 86, Huddinge, Sweden
| | - Kjell Hultenby
- Division of Clinical Research Centre, Department of Laboratory Medicine, Karolinska Institutet, SE-141 86, Huddinge, Sweden
| | - Olle Söder
- Department of Women’s and Children’s Health, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-171 76, Stockholm, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, SE-141 86, Huddinge, Sweden
| | - Jan-Bernd Stukenborg
- Department of Women’s and Children’s Health, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-171 76, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
32
|
Rossitto M, Philibert P, Poulat F, Boizet-Bonhoure B. Molecular events and signalling pathways of male germ cell differentiation in mouse. Semin Cell Dev Biol 2015; 45:84-93. [PMID: 26454096 DOI: 10.1016/j.semcdb.2015.09.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022]
Abstract
Germ cells, the precursors of gametes, represent a unique cell lineage that is able to differentiate into spermatozoa or oocytes depending on the chromosomal sex of the organism. In the mammalian embryonic gonad, commitment to oogenesis involves pre-meiotic DNA replication and entry into the first meiotic division; whereas, commitment to spermatogenesis involves inhibition of meiotic initiation, suppression of pluripotency, mitotic arrest and expression of specific markers that will control the development of the male germ cells. The crucial decision made by the germ line to commit to either a male or a female fate has been partially explained by genetic and ex vivo studies in mice which have implicated a complex network of regulatory genes, numerous factors and pathways. Besides the reproductive failure that may follow a deregulation of this complex network, the germ cells may, in view of their proliferative and pluripotent nature, act as precursors of potential malignant transformation and as putative targets for exogenous environmental compounds. Our review summarizes and discusses recent developments that have improved our understanding on how germ cell precursors are committed to a male or a female cell fate in the mouse gonad.
Collapse
Affiliation(s)
- Moïra Rossitto
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Pascal Philibert
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Francis Poulat
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| | - Brigitte Boizet-Bonhoure
- Genetic and Development Department, Institute of Human Genetics, CNRS UPR1142, Montpellier, France.
| |
Collapse
|
33
|
Economou C, Tsakiridis A, Wymeersch FJ, Gordon-Keylock S, Dewhurst RE, Fisher D, Medvinsky A, Smith AJH, Wilson V. Intrinsic factors and the embryonic environment influence the formation of extragonadal teratomas during gestation. BMC DEVELOPMENTAL BIOLOGY 2015; 15:35. [PMID: 26453549 PMCID: PMC4599726 DOI: 10.1186/s12861-015-0084-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 09/18/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pluripotent cells are present in early embryos until the levels of the pluripotency regulator Oct4 drop at the beginning of somitogenesis. Elevating Oct4 levels in explanted post-pluripotent cells in vitro restores their pluripotency. Cultured pluripotent cells can participate in normal development when introduced into host embryos up to the end of gastrulation. In contrast, pluripotent cells efficiently seed malignant teratocarcinomas in adult animals. In humans, extragonadal teratomas and teratocarcinomas are most frequently found in the sacrococcygeal region of neonates, suggesting that these tumours originate from cells in the posterior of the embryo that either reactivate or fail to switch off their pluripotent status. However, experimental models for the persistence or reactivation of pluripotency during embryonic development are lacking. METHODS We manually injected embryonic stem cells into conceptuses at E9.5 to test whether the presence of pluripotent cells at this stage correlates with teratocarcinoma formation. We then examined the effects of reactivating embryonic Oct4 expression ubiquitously or in combination with Nanog within the primitive streak (PS)/tail bud (TB) using a transgenic mouse line and embryo chimeras carrying a PS/TB-specific heterologous gene expression cassette respectively. RESULTS Here, we show that pluripotent cells seed teratomas in post-gastrulation embryos. However, at these stages, induced ubiquitous expression of Oct4 does not lead to restoration of pluripotency (indicated by Nanog expression) and tumour formation in utero, but instead causes a severe phenotype in the extending anteroposterior axis. Use of a more restricted T(Bra) promoter transgenic system enabling inducible ectopic expression of Oct4 and Nanog specifically in the posteriorly-located primitive streak (PS) and tail bud (TB) led to similar axial malformations to those induced by Oct4 alone. These cells underwent induction of pluripotency marker expression in Epiblast Stem Cell (EpiSC) explants derived from somitogenesis-stage embryos, but no teratocarcinoma formation was observed in vivo. CONCLUSIONS Our findings show that although pluripotent cells with teratocarcinogenic potential can be produced in vitro by the overexpression of pluripotency regulators in explanted somitogenesis-stage somatic cells, the in vivo induction of these genes does not yield tumours. This suggests a restrictive regulatory role of the embryonic microenvironment in the induction of pluripotency.
Collapse
Affiliation(s)
- Constantinos Economou
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Anestis Tsakiridis
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Filip J Wymeersch
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sabrina Gordon-Keylock
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Robert E Dewhurst
- Drug Discovery Unit, Telethon Kids Institute, PO Box 855, West Perth, WA, 6872, Australia
| | - Dawn Fisher
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Alexander Medvinsky
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Andrew J H Smith
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Valerie Wilson
- MRC Centre for Regenerative Medicine, School of Biological Sciences, SCRM Building, The University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
34
|
Zhang T, Murphy MW, Gearhart MD, Bardwell VJ, Zarkower D. The mammalian Doublesex homolog DMRT6 coordinates the transition between mitotic and meiotic developmental programs during spermatogenesis. Development 2014; 141:3662-71. [PMID: 25249458 DOI: 10.1242/dev.113936] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In mammals, a key transition in spermatogenesis is the exit from spermatogonial differentiation and mitotic proliferation and the entry into spermatocyte differentiation and meiosis. Although several genes that regulate this transition have been identified, how it is controlled and coordinated remains poorly understood. Here, we examine the role in male gametogenesis of the Doublesex-related gene Dmrt6 (Dmrtb1) in mice and find that Dmrt6 plays a crucial role in directing germ cells through the mitotic-to-meiotic germ cell transition. DMRT6 protein is expressed in late mitotic spermatogonia. In mice of the C57BL/6J strain, a null mutation in Dmrt6 disrupts spermatogonial differentiation, causing inappropriate expression of spermatogonial differentiation factors, including SOHLH1, SOHLH2 and DMRT1 as well as the meiotic initiation factor STRA8, and causing most late spermatogonia to undergo apoptosis. In mice of the 129Sv background, most Dmrt6 mutant germ cells can complete spermatogonial differentiation and enter meiosis, but they show defects in meiotic chromosome pairing, establishment of the XY body and processing of recombination foci, and they mainly arrest in mid-pachynema. mRNA profiling of Dmrt6 mutant testes together with DMRT6 chromatin immunoprecipitation sequencing suggest that DMRT6 represses genes involved in spermatogonial differentiation and activates genes required for meiotic prophase. Our results indicate that Dmrt6 plays a key role in coordinating the transition in gametogenic programs from spermatogonial differentiation and mitosis to spermatocyte development and meiosis.
Collapse
Affiliation(s)
- Teng Zhang
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark W Murphy
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micah D Gearhart
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vivian J Bardwell
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - David Zarkower
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| |
Collapse
|
35
|
Moniot B, Ujjan S, Champagne J, Hirai H, Aritake K, Nagata K, Dubois E, Nidelet S, Nakamura M, Urade Y, Poulat F, Boizet-Bonhoure B. Prostaglandin D2 acts through the Dp2 receptor to influence male germ cell differentiation in the foetal mouse testis. Development 2014; 141:3561-71. [DOI: 10.1242/dev.103408] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Through intercellular signalling, the somatic compartment of the foetal testis is able to program primordial germ cells to undergo spermatogenesis. Fibroblast growth factor 9 and several members of the transforming growth factor β superfamily are involved in this process in the foetal testis, counteracting the induction of meiosis by retinoic acid and activating germinal mitotic arrest. Here, using in vitro and in vivo approaches, we show that prostaglandin D2 (PGD2), which is produced through both L-Pgds and H-Pgds enzymatic activities in the somatic and germ cell compartments of the foetal testis, plays a role in mitotic arrest in male germ cells by activating the expression and nuclear localization of the CDK inhibitor p21Cip1 and by repressing pluripotency markers. We show that PGD2 acts through its Dp2 receptor, at least in part through direct effects in germ cells, and contributes to the proper differentiation of male germ cells through the upregulation of the master gene Nanos2. Our data identify PGD2 signalling as an early pathway that acts in both paracrine and autocrine manners, and contributes to the differentiation of germ cells in the foetal testis.
Collapse
Affiliation(s)
- Brigitte Moniot
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Safdar Ujjan
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Julien Champagne
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Hiroyuki Hirai
- Department of Advanced Technology and Development, BML, Matoba, Kawagoe, Saitama 350-1101, Japan
| | - Kosuke Aritake
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Osaka 565-0874, Japan
| | - Kinya Nagata
- Department of Advanced Technology and Development, BML, Matoba, Kawagoe, Saitama 350-1101, Japan
| | - Emeric Dubois
- Plateforme MGX, Functional Genomic Institute, CNRS UMR 5203 – INSERM U 661, Montpellier 34094, Cedex 05, France
| | - Sabine Nidelet
- Plateforme MGX, Functional Genomic Institute, CNRS UMR 5203 – INSERM U 661, Montpellier 34094, Cedex 05, France
| | - Masataka Nakamura
- Human Gene Sciences Center, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yoshihiro Urade
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Osaka 565-0874, Japan
| | - Francis Poulat
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Brigitte Boizet-Bonhoure
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| |
Collapse
|
36
|
Poulain M, Frydman N, Tourpin S, Muczynski V, Mucsynski V, Souquet B, Benachi A, Habert R, Rouiller-Fabre V, Livera G. Involvement of doublesex and mab-3-related transcription factors in human female germ cell development demonstrated by xenograft and interference RNA strategies. Mol Hum Reprod 2014; 20:960-71. [PMID: 25082981 DOI: 10.1093/molehr/gau058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We identified three doublesex and mab-3-related transcription factors (DMRT) that were sexually differentially expressed in human fetal gonads and present in the ovaries at the time of meiotic initiation. These were also identified in murine embryonic female germ cells. Among these, we focused on DMRTA2 (DMRT5), whose function is unknown in the developing gonads, and clarified its role in human female fetal germ cells, using an original xenograft model. Early human fetal ovaries (8-11 weeks post-fertilization) were grafted into nude mice. Grafted ovaries developed normally, with no apparent overt changes, when compared with ungrafted ovaries at equivalent developmental stages. Appropriate germ cell density, mitotic/meiotic transition, markers of meiotic progression and follicle formation were evident. Four weeks after grafting, mice were treated with siRNA, specifically targeting human DMRTA2 mRNA. DMRTA2 inhibition triggered an increase in undifferentiated FUT4-positive germ cells and a decrease in the percentage of meiotic γH2AX-positive germ cells, when compared with mice that were injected with control siRNA. Interestingly, the expression of markers associated with pre-meiotic germ cell differentiation was also impaired, as was the expression of DMRTB1 (DMRT6) and DMRTC2 (DMRT7). This study reveals, for the first time, the requirement of DMRTA2 for normal human female embryonic germ cell development. DMRTA2 appears to be necessary for proper differentiation of oogonia, prior to entry into meiosis, in the human species. Additionally, we developed a new model of organ xenografting, coupled with RNA interference, which provides a useful tool for genetic investigations of human germline development.
Collapse
Affiliation(s)
- Marine Poulain
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France AP-HP, University Paris-Sud, Reproductive Biology Unit, Clamart F-92140, France
| | - Nelly Frydman
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France AP-HP, University Paris-Sud, Reproductive Biology Unit, Clamart F-92140, France
| | - Sophie Tourpin
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Vincent Muczynski
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Vincent Mucsynski
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Benoit Souquet
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Alexandra Benachi
- AP-HP, University Paris-Sud, Department of Obstetrics and Gynecology, Clamart F-92140, France
| | - René Habert
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Virginie Rouiller-Fabre
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| | - Gabriel Livera
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR 967, Fontenay aux Roses F-92265, France CEA, DSV, iRCM, SCSR, LDG, Fontenay aux Roses F-92265, France INSERM, Unité 967, Fontenay aux Roses F-92265, France University Paris-Sud, UMR 967, Fontenay aux Roses F-92265, France
| |
Collapse
|
37
|
Schemmer J, Araúzo-Bravo MJ, Haas N, Schäfer S, Weber SN, Becker A, Eckert D, Zimmer A, Nettersheim D, Schorle H. Transcription factor TFAP2C regulates major programs required for murine fetal germ cell maintenance and haploinsufficiency predisposes to teratomas in male mice. PLoS One 2013; 8:e71113. [PMID: 23967156 PMCID: PMC3742748 DOI: 10.1371/journal.pone.0071113] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/26/2013] [Indexed: 12/27/2022] Open
Abstract
Maintenance and maturation of primordial germ cells is controlled by complex genetic and epigenetic cascades, and disturbances in this network lead to either infertility or malignant aberration. Transcription factor TFAP2C has been described to be essential for primordial germ cell maintenance and to be upregulated in several human germ cell cancers. Using global gene expression profiling, we identified genes deregulated upon loss of Tfap2c in embryonic stem cells and primordial germ cell-like cells. We show that loss of Tfap2c affects many aspects of the genetic network regulating germ cell biology, such as downregulation of maturation markers and induction of markers indicative for somatic differentiation, cell cycle, epigenetic remodeling and pluripotency. Chromatin-immunoprecipitation analyses demonstrated binding of TFAP2C to regulatory regions of deregulated genes (Sfrp1, Dmrt1, Nanos3, c-Kit, Cdk6, Cdkn1a, Fgf4, Klf4, Dnmt3b and Dnmt3l) suggesting that these genes are direct transcriptional targets of TFAP2C in primordial germ cells. Since Tfap2c deficient primordial germ cell-like cells display cancer related deregulations in epigenetic remodeling, cell cycle and pluripotency control, the Tfap2c-knockout allele was bred onto 129S2/Sv genetic background. There, mice heterozygous for Tfap2c develop with high incidence germ cell cancer resembling human pediatric germ cell tumors. Precursor lesions can be observed as early as E16.5 in developing testes displaying persisting expression of pluripotency markers. We further demonstrate that mice with a heterozygous deletion of the TFAP2C target gene Nanos3 are also prone to develop teratomas. These data highlight TFAP2C as a critical and dose-sensitive regulator of germ cell fate.
Collapse
Affiliation(s)
- Jana Schemmer
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Marcos J. Araúzo-Bravo
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental Biology, Münster, Germany
| | - Natalie Haas
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Sabine Schäfer
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Susanne N. Weber
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Astrid Becker
- University of Bonn Medical School, Institute for Reconstructive Neurobiology, Life&Brain Center, Bonn, Germany
| | - Dawid Eckert
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Andreas Zimmer
- University of Bonn Medical School, Institute for Reconstructive Neurobiology, Life&Brain Center, Bonn, Germany
| | - Daniel Nettersheim
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| | - Hubert Schorle
- University of Bonn Medical School, Institute of Pathology, Department of Developmental Pathology, Bonn, Germany
| |
Collapse
|
38
|
Zechel JL, Doerner SK, Lager A, Tesar PJ, Heaney JD, Nadeau JH. Contrasting effects of Deadend1 (Dnd1) gain and loss of function mutations on allelic inheritance, testicular cancer, and intestinal polyposis. BMC Genet 2013; 14:54. [PMID: 23773267 PMCID: PMC3693958 DOI: 10.1186/1471-2156-14-54] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 06/07/2013] [Indexed: 11/10/2022] Open
Abstract
Background Certain mutations in the Deadend1 (Dnd1) gene are the most potent modifiers of testicular germ cell tumor (TGCT) susceptibility in mice and rats. In the 129 family of mice, the Dnd1Ter mutation significantly increases occurrence of TGCT-affected males. To test the hypothesis that he Dnd1Ter allele is a loss-of-function mutation; we characterized the consequences of a genetically-engineered loss-of-function mutation in mice, and compared these results with those for Dnd1Ter. Results We found that intercrossing Dnd1+/KO heterozygotes to generate a complete loss-of-function led to absence of Dnd1KO/KO homozygotes and significantly reduced numbers of Dnd1+/KO heterozygotes. Further crosses showed that Dnd1Ter partially rescues loss of Dnd1KO mice. We also found that loss of a single copy of Dnd1 in Dnd1KO/+ heterozygotes did not affect baseline occurrence of TGCT-affected males and that Dnd1Ter increased TGCT risk regardless whether the alternative allele was loss-of-function (Dnd1KO) or wild-type (Dnd1+). Finally, we found that the action of Dnd1Ter was not limited to testicular cancer, but also significantly increased polyp number and burden in the Apc+/Min model of intestinal polyposis. Conclusion These results show that Dnd1 is essential for normal allelic inheritance and that Dnd1Ter has a novel combination of functions that significantly increase risk for both testicular and intestinal cancer.
Collapse
Affiliation(s)
- Jennifer L Zechel
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland OH 44106, USA
| | | | | | | | | | | |
Collapse
|
39
|
Haugen TB. Genfeil kan gi testikkelkreft. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2013. [DOI: 10.4045/tidsskr.13.0957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|