1
|
Perrotta G, Condrea D, Ghyselinck NB. Meiosis and retinoic acid in the mouse fetal gonads: An unforeseen twist. Curr Top Dev Biol 2024; 161:59-88. [PMID: 39870439 DOI: 10.1016/bs.ctdb.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
In mammals, differentiation of germ cells is crucial for sexual reproduction, involving complex signaling pathways and environmental cues defined by the somatic cells of the gonads. This review examines the long-standing model positing that all-trans retinoic acid (ATRA) acts as a meiosis-inducing substance (MIS) in the fetal ovary by inducing expression of STRA8 in female germ cells, while CYP26B1 serves as a meiosis-preventing substance (MPS) in the fetal testis by degrading ATRA and preventing STRA8 expression in the male germ cells until postnatal development. Recent genetic studies in the mouse challenge this paradigm, revealing that meiosis initiation in female germ cells can occur independently of ATRA signaling, with key roles played by other intrinsic factors like DAZL and DMRT1, and extrinsic signals such as BMPs and vitamin C. Thus, ATRA can no longer be considered as 'the' long-searched MIS. Furthermore, evidence indicates that CYP26B1 does not prevent meiosis by degrading ATRA in the fetal testis, but acts by degrading an unidentified MIS or synthesizing an equally unknown MPS. By emphasizing the necessity of genetic loss-of-function approaches to accurately delineate the roles of signaling molecules such ATRA in vivo, this chapter calls for a reevaluation of the mechanisms instructing and preventing meiosis initiation in the fetal ovary and testis, respectively. It highlights the need for further research into the molecular identities of the signals involved in these processes.
Collapse
Affiliation(s)
- Giulia Perrotta
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Diana Condrea
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Norbert B Ghyselinck
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
| |
Collapse
|
2
|
Heidari B, Shirazi A, Akbari N, Barzegar-Amini M. Identification and Manipulation of Spermatogonial Stem Cells with the Aim of Inducing Spermatogenesis in Vitro. Reprod Sci 2024:10.1007/s43032-024-01709-2. [PMID: 39424678 DOI: 10.1007/s43032-024-01709-2] [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: 05/20/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Assisted reproduction techniques for infertile men with non-obstructive azoospermia require a sufficient number of functional germ cells produced in vitro. Understanding the mechanisms that allow the resumption of spermatogenesis outside the testicular environment is crucial for fertility preservation in these patients. A review of the literature was conducted using databases such as PubMed, Scopus and Web of Science, with keywords including "spermatogonial stem cell," "germ cells," "male factor infertility," and "enrichment and propagation of SSCs in vitro." Currently, two models-"in vivo" and "in vitro"-have been developed for producing haploid germ cells. The "in vivo" models include spermatogonial stem cell transplantation and testicular xenograft techniques. In contrast, the "in vitro" models consist of conventional culture systems, organ culture, and three-dimensional culture systems, all designed to induce spermatogenesis in vitro. These culture systems enable the simulation of the seminiferous epithelium in vitro, which facilitates better regulation of cell-signaling pathways that control the self-renewal and differentiation of SSCs. This review provides up-to-date information on the organization of SSCs, focusing on the identification, proliferation, and differentiation of spermatogonia in vitro.
Collapse
Affiliation(s)
- Banafsheh Heidari
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Nazanin Akbari
- Department of Biology, Shahid Beheshti University, Tehran, Iran
- Clinical Research Development Unit, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maral Barzegar-Amini
- Clinical Research Development Unit, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
El Omri-Charai R, Rwigemera A, Gilbert I, Langford A, Robert C, Sloboda DM, McGraw S, Delbes G. Erasure of DNA methylation in rat fetal germ cells is sex-specific and sensitive to maternal high-fat diet. J Dev Orig Health Dis 2024; 15:e19. [PMID: 39324180 DOI: 10.1017/s2040174424000230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
In mammals, DNA methylation (DNAme) erasure and reinstatement during embryo development and germline establishment are sensitive to the intrauterine environment. Maternal intake of a high-fat diet (HFD), associated with excessive gestational weight gain, has transgenerational effects on offspring health, which may be mediated by changes in DNAme in the germline. Here, we tested the impact of a maternal HFD on embryonic germline DNAme erasure using a rat strain that expresses green fluorescent protein specifically in germ cells. DNAme was analysed by methyl-seq capture in germ cells collected from male and female F1 gonads at gestational day 16. Our data show that although HFD induced global hypomethylation in both sexes, DNAme erasure in female germ cells was more advanced compared to male germ cells. The delay in DNAme erasure in males and the greater impact of HFD suggest that male germ cells are more vulnerable to alterations by exogenous factors.
Collapse
Affiliation(s)
- R El Omri-Charai
- Centre Armand Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - A Rwigemera
- Centre Armand Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| | - I Gilbert
- Department of Animal Sciences, Faculty of Agricultural and Food Sciences, Université Laval, Quebec, QC, Canada
| | - A Langford
- Department of Obstetrics and Gynecology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - C Robert
- Department of Animal Sciences, Faculty of Agricultural and Food Sciences, Université Laval, Quebec, QC, Canada
| | - D M Sloboda
- Departments of Biochemistry and Biomedical Sciences, Obstetrics and Gynecology and Pediatrics, McMaster University, Hamilton, ON, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - S McGraw
- Department of Obstetrics and Gynecology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche Azrieli du Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC, Canada
| | - G Delbes
- Centre Armand Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC, Canada
| |
Collapse
|
4
|
Yang M, Diaz F, Krause ART, Lei Y, Liu WS. Synergistic enhancement of the mouse Pramex1 and Pramel1 in repressing retinoic acid (RA) signaling during gametogenesis. Cell Biosci 2024; 14:28. [PMID: 38395975 PMCID: PMC10893636 DOI: 10.1186/s13578-024-01212-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND PRAME constitutes one of the largest multi-copy gene families in Eutherians, encoding cancer-testis antigens (CTAs) with leucine-rich repeats (LRR) domains, highly expressed in cancer cells and gametogenic germ cells. This study aims to elucidate genetic interactions between two members, Pramex1 and Pramel1, in the mouse Prame family during gametogenesis using a gene knockout approach. RESULT Single-gene knockout (sKO) of either Pramex1 or Pramel1 resulted in approximately 7% of abnormal seminiferous tubules, characterized by a Sertoli-cell only (SCO) phenotype, impacting sperm count and fecundity significantly. Remarkably, sKO female mice displayed normal reproductive functions. In contrast, Pramex1/Pramel1 double knockout (dKO) mice exhibited reduced fecundity in both sexes. In dKO females, ovarian primary follicle count decreased by 50% compared to sKO and WT mice, correlating with a 50% fecundity decrease. This suggested compensatory roles during oogenesis in Pramex1 or Pramel1 sKO females. Conversely, dKO males showed an 18% frequency of SCO tubules, increased apoptotic germ cells, and decreased undifferentiated spermatogonia compared to sKO and WT testes. Western blot analysis with PRAMEX1- or PRAMEL1-specific antibodies on sKO testes revealed compensatory upregulation of each protein (30-50%) in response to the other gene's deletion. Double KO males exhibited more severe defects in sperm count and litter size, surpassing Pramex1 and Pramel1 sKO accumulative effects, indicating a synergistic enhancement interaction during spermatogenesis. Additional experiments administering trans-retinoic acid (RA) and its inhibitor (WIN18,446) in sKO, dKO, and WT mice suggested that PRAMEX1 and PRAMEL1 synergistically repress the RA signaling pathway during spermatogenesis. CONCLUSION Data from sKO and dKO mice unveil a synergistic interaction via the RA signaling pathway between Pramex1 and Pramel1 genes during gametogenesis. This discovery sets the stage for investigating interactions among other members within the Prame family, advancing our understanding of multi-copy gene families involved in germ cell formation and function.
Collapse
Affiliation(s)
- Mingyao Yang
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Francisco Diaz
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Ana Rita T Krause
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, College of Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wan-Sheng Liu
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA.
| |
Collapse
|
5
|
Carver JJ, Zhu Y. Metzincin metalloproteases in PGC migration and gonadal sex conversion. Gen Comp Endocrinol 2023; 330:114137. [PMID: 36191636 DOI: 10.1016/j.ygcen.2022.114137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/13/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022]
Abstract
Development of a functional gonad includes migration of primordial germ cells (PGCs), differentiations of somatic and germ cells, formation of primary follicles or spermatogenic cysts with somatic gonadal cells, development and maturation of gametes, and subsequent releasing of mature germ cells. These processes require extensive cellular and tissue remodeling, as well as broad alterations of the surrounding extracellular matrix (ECM). Metalloproteases, including MMPs (matrix metalloproteases), ADAMs (a disintegrin and metalloproteinases), and ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs), are suggested to have critical roles in the remodeling of the ECM during gonad development. However, few research articles and reviews are available on the functions and mechanisms of metalloproteases in remodeling gonadal ECM, gonadal development, or gonadal differentiation. Moreover, most studies focused on the roles of transcription and growth factors in early gonad development and primary sex determination, leaving a significant knowledge gap on how differentially expressed metalloproteases exert effects on the ECM, cell migration, development, and survival of germ cells during the development and differentiation of ovaries or testes. We will review gonad development with focus on the evidence of metalloprotease involvements, and with an emphasis on zebrafish as a model for studying gonadal sex differentiation and metalloprotease functions.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Yong Zhu
- Department of Biology, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
6
|
Farini D, De Felici M. The Beginning of Meiosis in Mammalian Female Germ Cells: A Never-Ending Story of Intrinsic and Extrinsic Factors. Int J Mol Sci 2022; 23:ijms232012571. [PMID: 36293427 PMCID: PMC9604137 DOI: 10.3390/ijms232012571] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Meiosis is the unique division of germ cells resulting in the recombination of the maternal and paternal genomes and the production of haploid gametes. In mammals, it begins during the fetal life in females and during puberty in males. In both cases, entering meiosis requires a timely switch from the mitotic to the meiotic cell cycle and the transition from a potential pluripotent status to meiotic differentiation. Revealing the molecular mechanisms underlying these interrelated processes represents the essence in understanding the beginning of meiosis. Meiosis facilitates diversity across individuals and acts as a fundamental driver of evolution. Major differences between sexes and among species complicate the understanding of how meiosis begins. Basic meiotic research is further hindered by a current lack of meiotic cell lines. This has been recently partly overcome with the use of primordial-germ-cell-like cells (PGCLCs) generated from pluripotent stem cells. Much of what we know about this process depends on data from model organisms, namely, the mouse; in mice, the process, however, appears to differ in many aspects from that in humans. Identifying the mechanisms and molecules controlling germ cells to enter meiosis has represented and still represents a major challenge for reproductive medicine. In fact, the proper execution of meiosis is essential for fertility, for maintaining the integrity of the genome, and for ensuring the normal development of the offspring. The main clinical consequences of meiotic defects are infertility and, probably, increased susceptibility to some types of germ-cell tumors. In the present work, we report and discuss data mainly concerning the beginning of meiosis in mammalian female germ cells, referring to such process in males only when pertinent. After a brief account of this process in mice and humans and an historical chronicle of the major hypotheses and progress in this topic, the most recent results are reviewed and discussed.
Collapse
|
7
|
Alhasnani MA, Loeb S, Hall SJ, Caruolo Z, Simmonds F, Solano AE, Spade DJ. Interaction between mono-(2-ethylhexyl) phthalate and retinoic acid alters Sertoli cell development during fetal mouse testis cord morphogenesis. Curr Res Toxicol 2022; 3:100087. [PMID: 36189433 PMCID: PMC9520016 DOI: 10.1016/j.crtox.2022.100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/24/2022] Open
Abstract
Phthalic acid esters (phthalates) are a class of industrial chemicals that cause developmental and reproductive toxicity, but there are significant gaps in knowledge of phthalate toxicity mechanisms. There is evidence that phthalates disrupt retinoic acid signaling in the fetal testis, potentially disrupting control of spatial and temporal patterns of testis development. Our goal was to determine how a phthalate would interact with retinoic acid signaling during fetal mouse testis development. We hypothesized that mono-(2-ethylhexyl) phthalate (MEHP) would exacerbate the adverse effect of all-trans retinoic acid (ATRA) on seminiferous cord development in the mouse fetal testis. To test this hypothesis, gestational day (GD) 14 C57BL/6 mouse testes were isolated and cultured on media containing MEHP, ATRA, or a combination of both compounds. Cultured testes were collected for global transcriptome analysis after one day in culture and for histology and immunofluorescent analysis of Sertoli cell differentiation after three days in culture. ATRA disrupted seminiferous cord morphogenesis and induced aberrant FOXL2 expression. MEHP alone had no significant effect on cord development, but combined exposure to MEHP and ATRA increased the number of FOXL2-positive cells, reduced seminiferous cord number, and increased testosterone levels, beyond the effect of ATRA alone. In RNA-seq analysis, ATRA treatment and MEHP treatment resulted in differential expression of genes 510 and 134 genes, respectively, including 70 common differentially expressed genes (DEGs) between the two treatments, including genes with known roles in fetal testis development. MEHP DEGs included RAR target genes, genes involved in angiogenesis, and developmental patterning genes, including members of the homeobox superfamily. These results support the hypothesis that MEHP modulates retinoic acid signaling in the mouse fetal testis and provide insight into potential mechanisms by which phthalates disrupt seminiferous cord morphogenesis.
Collapse
Key Words
- ATRA, All-trans retinoic acid. CAS # 302-79-4
- DMSO, dimethyl sulfoxide
- Fetal testis development
- GD, gestational day
- GO, Gene Ontology
- IPA, Ingenuity Pathway Analysis
- ITCN, Image-based Tool for Counting Nuclei
- MEHP, mono-(2-ethylheyxl) phthalate. CAS # 4376-20-9
- MNGs, multinucleated germ cells
- PVC, polyvinyl chloride
- Phthalate toxicity
- Retinoic acid
- Sertoli cell
- TDS, testicular dysgenesis syndrome
Collapse
Affiliation(s)
- Maha A. Alhasnani
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Skylar Loeb
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Susan J. Hall
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Zachary Caruolo
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Faith Simmonds
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Amanda E. Solano
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Daniel J. Spade
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| |
Collapse
|
8
|
Loup B, Poumerol E, Jouneau L, Fowler PA, Cotinot C, Mandon-Pépin B. BPA disrupts meiosis I in oogonia by acting on pathways including cell cycle regulation, meiosis initiation and spindle assembly. Reprod Toxicol 2022; 111:166-177. [PMID: 35667523 DOI: 10.1016/j.reprotox.2022.06.001] [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: 01/02/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022]
Abstract
The negative in utero effects of bisphenol A (BPA) on female reproduction are of concern since the ovarian reserve of primordial follicles is constituted during the fetal period. This time-window is difficult to access, particularly in humans. Animal models and explant culture systems are, therefore, vital tools for investigating EDC impacts on primordial germ cells (PGCs). Here, we investigated the effects of BPA on prophase I meiosis in the fetal sheep ovary. We established an in vitro model of early gametogenesis through retinoic acid (RA)-induced differentiation of sheep PGCs that progressed through meiosis. Using this system, we demonstrated that BPA (3×10-7 M & 3×10-5M) exposure for 20 days disrupted meiotic initiation and completion in sheep oogonia and induced transcriptomic modifications of exposed explants. After exposure to the lowest concentrations of BPA (3×10-7M), only 2 probes were significantly up-regulated corresponding to NR2F1 and TMEM167A transcripts. In contrast, after exposure to 3×10-5M BPA, 446 probes were deregulated, 225 were down- and 221 were up-regulated following microarray analysis. Gene Ontology (GO) annotations of differentially expressed genes revealed that pathways mainly affected were involved in cell-cycle phase transition, meiosis and spindle assembly. Differences in key gene expression within each pathway were validated by qRT-PCR. This study provides a novel model for direct examination of the molecular pathways of environmental toxicants on early female gametogenesis and novel insights into the mechanisms by which BPA affects meiosis I. BPA exposure could thereby disrupt ovarian reserve formation by inhibiting meiotic progression of oocytes I and consequently by increasing atresia of primordial follicles containing defective oocytes.
Collapse
Affiliation(s)
- Benoit Loup
- Université Paris-Saclay, UVSQ, ENVA, INRAE, BREED, 78350, Jouy-en-Josas, France.
| | - Elodie Poumerol
- Université Paris-Saclay, UVSQ, ENVA, INRAE, BREED, 78350, Jouy-en-Josas, France.
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, ENVA, INRAE, BREED, 78350, Jouy-en-Josas, France.
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Corinne Cotinot
- Université Paris-Saclay, UVSQ, ENVA, INRAE, BREED, 78350, Jouy-en-Josas, France.
| | | |
Collapse
|
9
|
Tran KTD, Valli-Pulaski H, Colvin A, Orwig KE. Male fertility preservation and restoration strategies for patients undergoing gonadotoxic therapies†. Biol Reprod 2022; 107:382-405. [PMID: 35403667 PMCID: PMC9382377 DOI: 10.1093/biolre/ioac072] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
Medical treatments for cancers or other conditions can lead to permanent infertility. Infertility is an insidious disease that impacts not only the ability to have a biological child but also the emotional well-being of the infertile individuals, relationships, finances, and overall health. Therefore, all patients should be educated about the effects of their medical treatments on future fertility and about fertility preservation options. The standard fertility preservation option for adolescent and adult men is sperm cryopreservation. Sperms can be frozen and stored for a long period, thawed at a later date, and used to achieve pregnancy with existing assisted reproductive technologies. However, sperm cryopreservation is not applicable for prepubertal patients who do not yet produce sperm. The only fertility preservation option available to prepubertal boys is testicular tissue cryopreservation. Next-generation technologies are being developed to mature those testicular cells or tissues to produce fertilization-competent sperms. When sperm and testicular tissues are not available for fertility preservation, inducing pluripotent stem cells derived from somatic cells, such as blood or skin, may provide an alternative path to produce sperms through a process call in vitro gametogenesis. This review describes standard and experimental options to preserve male fertility as well as the experimental options to produce functional spermatids or sperms from immature cryopreserved testicular tissues or somatic cells.
Collapse
Affiliation(s)
- Kien T D Tran
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Hanna Valli-Pulaski
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Amanda Colvin
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Kyle E Orwig
- Correspondence: Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213, USA. Tel: 412-641-2460; E-mail:
| |
Collapse
|
10
|
Spiller C, Bowles J. Instructing Mouse Germ Cells to Adopt a Female Fate. Sex Dev 2022:1-13. [PMID: 35320803 DOI: 10.1159/000523763] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/20/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Germ cells are critical for the survival of our species. They are the only cells that undergo meiosis - the reductive form of cell division that is necessary for genetic reassortment of chromosomes and production of the haploid gametes, the sperm and eggs. Remarkably, the initial female/male fate decision in fetal germ cells does not depend on whether they are chromosomally XX or XY; rather, initial sexual fate is imposed by influences from the surrounding tissue. In mammals, the female germline is particularly precious: despite recent suggestions that germline stem cells exist in the ovary, it is still generally accepted that the ovarian reserve is finite, and its size is dependant on germ cells of the fetal ovary initiating meiosis in a timely manner. SUMMARY Prior to 2006, evidence suggested that gonadal germ cells initiate meiotic prophase I by default, but more recent data support a key role for the signalling molecule retinoic acid (RA) in instructing female germ cell fate. Newer findings also support a key meiosis-inducing role for another signalling molecule, bone morphogenic protein (BMP). Nonetheless, many questions remain. KEY MESSAGES Here, we review knowledge thus far regarding extrinsic and intrinsic determinants of a female germ cell fate, focusing on the mouse model.
Collapse
Affiliation(s)
- Cassy Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Zhao J, Lu P, Wan C, Huang Y, Cui M, Yang X, Hu Y, Zheng Y, Dong J, Wang M, Zhang S, Liu Z, Bian S, Wang X, Wang R, Ren S, Wang D, Yao Z, Chang G, Tang F, Zhao XY. Cell-fate transition and determination analysis of mouse male germ cells throughout development. Nat Commun 2021; 12:6839. [PMID: 34824237 PMCID: PMC8617176 DOI: 10.1038/s41467-021-27172-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022] Open
Abstract
Mammalian male germ cell development is a stepwise cell-fate transition process; however, the full-term developmental profile of male germ cells remains undefined. Here, by interrogating the high-precision transcriptome atlas of 11,598 cells covering 28 critical time-points, we demonstrate that cell-fate transition from mitotic to post-mitotic primordial germ cells is accompanied by transcriptome-scale reconfiguration and a transitional cell state. Notch signaling pathway is essential for initiating mitotic arrest and the maintenance of male germ cells' identities. Ablation of HELQ induces developmental arrest and abnormal transcriptome reprogramming of male germ cells, indicating the importance of cell cycle regulation for proper cell-fate transition. Finally, systematic human-mouse comparison reveals potential regulators whose deficiency contributed to human male infertility via mitotic arrest regulation. Collectively, our study provides an accurate and comprehensive transcriptome atlas of the male germline cycle and allows for an in-depth understanding of the cell-fate transition and determination underlying male germ cell development.
Collapse
Affiliation(s)
- Jiexiang Zhao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Ping Lu
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Biomedical Pioneering Innovation Center, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, 100871, Beijing, P. R. China
| | - Cong Wan
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Yaping Huang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Manman Cui
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Xinyan Yang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Yuqiong Hu
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Biomedical Pioneering Innovation Center, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, 100871, Beijing, P. R. China
| | - Yi Zheng
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Biomedical Pioneering Innovation Center, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, 100871, Beijing, P. R. China
| | - Mei Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Shu Zhang
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Biomedical Pioneering Innovation Center, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, 100871, Beijing, P. R. China
| | - Zhaoting Liu
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Shuhui Bian
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, P. R. China
| | - Xiaoman Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Rui Wang
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, P. R. China
| | - Shaofang Ren
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Dazhuang Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Zhaokai Yao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China
| | - Gang Chang
- Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, 518060, Shenzhen, Guangdong, P. R. China.
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics (ICG), School of Life Sciences, Peking University, 100871, Beijing, P. R. China.
- Biomedical Pioneering Innovation Center, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, 100871, Beijing, P. R. China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, P. R. China.
| | - Xiao-Yang Zhao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China.
- Guangdong Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, 510515, Guangzhou, Guangdong, P. R. China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), 510700, Guangzhou, Guangdong, P. R. China.
| |
Collapse
|
12
|
Feng CW, Burnet G, Spiller CM, Cheung FKM, Chawengsaksophak K, Koopman P, Bowles J. Identification of regulatory elements required for Stra8 expression in fetal ovarian germ cells of the mouse. Development 2021; 148:dev.194977. [PMID: 33574039 DOI: 10.1242/dev.194977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022]
Abstract
In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.
Collapse
Affiliation(s)
- Chun-Wei Feng
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Guillaume Burnet
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Cassy M Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Fiona Ka Man Cheung
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kallayanee Chawengsaksophak
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i. Vídenská 1083, 4 14220 Prague, Czech Republic
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia .,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
13
|
Vernet N, Condrea D, Mayere C, Féret B, Klopfenstein M, Magnant W, Alunni V, Teletin M, Souali-Crespo S, Nef S, Mark M, Ghyselinck NB. Meiosis occurs normally in the fetal ovary of mice lacking all retinoic acid receptors. SCIENCE ADVANCES 2020; 6:eaaz1139. [PMID: 32917583 PMCID: PMC7244263 DOI: 10.1126/sciadv.aaz1139] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/13/2020] [Indexed: 05/27/2023]
Abstract
Gametes are generated through a specialized cell differentiation process, meiosis, which, in ovaries of most mammals, is initiated during fetal life. All-trans retinoic acid (ATRA) is considered as the molecular signal triggering meiosis initiation. In the present study, we analyzed female fetuses ubiquitously lacking all ATRA nuclear receptors (RAR), obtained through a tamoxifen-inducible cre recombinase-mediated gene targeting approach. Unexpectedly, mutant oocytes robustly expressed meiotic genes, including the meiotic gatekeeper STRA8. In addition, ovaries from mutant fetuses grafted into adult recipient females yielded offspring bearing null alleles for all Rar genes. Thus, our results show that RAR are fully dispensable for meiotic initiation, as well as for the production of functional oocytes. Assuming that the effects of ATRA all rely on RAR, our study goes against the current model according to which meiosis is triggered by endogenous ATRA in the developing ovary. It therefore revives the search for the meiosis-inducing substance.
Collapse
Affiliation(s)
- Nadège Vernet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Diana Condrea
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Chloé Mayere
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Betty Féret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Muriel Klopfenstein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - William Magnant
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Violaine Alunni
- GenomEast platform, France Génomique consortium, IGBMC, 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Marius Teletin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| | - Sirine Souali-Crespo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Manuel Mark
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| | - Norbert B Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France.
| |
Collapse
|
14
|
Spade DJ, Hall SJ, Wortzel JD, Reyes G, Boekelheide K. All-trans Retinoic Acid Disrupts Development in Ex Vivo Cultured Fetal Rat Testes. II: Modulation of Mono-(2-ethylhexyl) Phthalate Toxicity. Toxicol Sci 2020; 168:149-159. [PMID: 30476341 DOI: 10.1093/toxsci/kfy283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Humans are universally exposed to low levels of phthalate esters (phthalates), which are used to plasticize polyvinyl chloride. Phthalates exert adverse effects on the development of seminiferous cords in the fetal testis through unknown toxicity pathways. To investigate the hypothesis that phthalates alter seminiferous cord development by disrupting retinoic acid (RA) signaling in the fetal testis, gestational day 15 fetal rat testes were exposed for 1-3 days to 10-6 M all-trans retinoic acid (ATRA) alone or in combination with 10-6-10-4 M mono-(2-ethylhexyl) phthalate (MEHP) in ex vivo culture. As previously reported, exogenous ATRA reduced seminiferous cord number. This effect was attenuated in a concentration-dependent fashion by MEHP co-exposure. ATRA and MEHP-exposed testes were depleted of DDX4-positive germ cells but not Sertoli cells. MEHP alone enhanced the expression of the RA receptor target Rbp1 and the ovary development-associated genes Wnt4 and Nr0b1, and suppressed expression of the Leydig cell marker, Star, and the germ cell markers, Ddx4 and Pou5f1. In co-exposures, MEHP predominantly enhanced the gene expression effects of ATRA, but the Wnt4 and Nr0b1 concentration-responses were nonlinear. Similarly, ATRA increased the number of cells expressing the granulosa cell marker FOXL2 in testis cultures, but this induction was attenuated by addition of MEHP. These results indicate that MEHP can both enhance and inhibit actions of ATRA during fetal testis development and provide evidence that RA signaling is a target for phthalate toxicity in the fetal testis.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Jeremy D Wortzel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Gerardo Reyes
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912.,Division of Natural Sciences, College of Mount Saint Vincent, Riverdale, New York 10471
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
15
|
Kim SM, Yokoyama T, Ng D, Ulu F, Yamazaki Y. Retinoic acid-stimulated ERK1/2 pathway regulates meiotic initiation in cultured fetal germ cells. PLoS One 2019; 14:e0224628. [PMID: 31682623 PMCID: PMC6827903 DOI: 10.1371/journal.pone.0224628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/17/2019] [Indexed: 11/18/2022] Open
Abstract
In murine fetal germ cells, retinoic acid (RA) is an extrinsic cue for meiotic initiation that stimulates transcriptional activation of the Stimulated by retinoic acid gene 8 (Stra8), which is required for entry of germ cells into meiotic prophase I. Canonically, the biological activities of RA are mediated by nuclear RA receptors. Recent studies in somatic cells found that RA noncanonically stimulates intracellular signal transduction pathways to regulate multiple cellular processes. In this study, using a germ cell culture system, we investigated (1) whether RA treatment activates any mitogen-activated protein kinase (MAPK) pathways in fetal germ cells at the time of sex differentiation, and (2) if this is the case, whether the corresponding RA-stimulated signaling pathway regulates Stra8 expression in fetal germ cells and their entry into meiosis. When XX germ cells at embryonic day (E) 12.5 were cultured with RA, the extracellular-signal-regulated kinase (ERK) 1/2 pathway was predominantly activated. MEK1/2 inhibitor (U0126) treatment suppressed the mRNA expressions of RA-induced Stra8 and meiotic marker genes (Rec8, Spo11, Dmc1, and Sycp3) in both XX and XY fetal germ cells. Furthermore, U0126 treatment dramatically reduced STRA8 protein levels and numbers of meiotic cells among cultured XX and XY fetal germ cells even in the presence of RA. Taken together, our results suggest the novel concept that the RA functions by stimulating the ERK1/2 pathway and that this activity is critical for Stra8 expression and meiotic progression in fetal germ cells.
Collapse
Affiliation(s)
- Sung-Min Kim
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States of America
| | - Toshifumi Yokoyama
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States of America
- Department of Animal Science, Kobe University, Kobe, Hyogo, Japan
| | - Dylan Ng
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States of America
| | - Ferhat Ulu
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States of America
| | - Yukiko Yamazaki
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States of America
- * E-mail:
| |
Collapse
|
16
|
Divergent Roles of CYP26B1 and Endogenous Retinoic Acid in Mouse Fetal Gonads. Biomolecules 2019; 9:biom9100536. [PMID: 31561560 PMCID: PMC6843241 DOI: 10.3390/biom9100536] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 11/19/2022] Open
Abstract
In female mammals, germ cells enter meiosis in the fetal ovaries, while in males, meiosis is prevented until postnatal development. Retinoic acid (RA) is considered the main inducer of meiotic entry, as it stimulates Stra8 which is required for the mitotic/meiotic switch. In fetal testes, the RA-degrading enzyme CYP26B1 prevents meiosis initiation. However, the role of endogenous RA in female meiosis entry has never been demonstrated in vivo. In this study, we demonstrate that some effects of RA in mouse fetal gonads are not recapitulated by the invalidation or up-regulation of CYP26B1. In organ culture of fetal testes, RA stimulates testosterone production and inhibits Sertoli cell proliferation. In the ovaries, short-term inhibition of RA-signaling does not decrease Stra8 expression. We develop a gain-of-function model to express CYP26A1 or CYP26B1. Only CYP26B1 fully prevents STRA8 induction in female germ cells, confirming its role as part of the meiotic prevention machinery. CYP26A1, a very potent RA degrading enzyme, does not impair the formation of STRA8-positive cells, but decreases Stra8 transcription. Collectively, our data reveal that CYP26B1 has other activities apart from metabolizing RA in fetal gonads and suggest a role of endogenous RA in amplifying Stra8, rather than being the initial inducer of Stra8. These findings should reactivate the quest to identify meiotic preventing or inducing substances.
Collapse
|
17
|
Mäkelä JA, Koskenniemi JJ, Virtanen HE, Toppari J. Testis Development. Endocr Rev 2019; 40:857-905. [PMID: 30590466 DOI: 10.1210/er.2018-00140] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022]
Abstract
Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades, and these are described along with the description of molecular regulation of testis development.
Collapse
Affiliation(s)
- Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jaakko J Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Helena E Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
18
|
Abstract
Germ cells are the stem cells of the species. Thus, it is critical that we have a good understanding of how they are specified, how the somatic cells instruct and support them, how they commit to one or other sex, and how they ultimately develop into functional gametes. Here, we focus on specifics of how sexual fate is determined during fetal life. Because the majority of relevant experimental work has been done using the mouse model, we focus on that species. We review evidence regarding the identity of instructive signals from the somatic cells, and the molecular responses that occur in germ cells in response to those extrinsic signals. In this way we aim to clarify progress to date regarding the mechanisms underlying the mitotic to meiosis switch in germ cells of the fetal ovary, and those involved in adopting and securing male fate in germ cells of the fetal testis.
Collapse
Affiliation(s)
- Cassy Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
19
|
Spade DJ, Dere E, Hall SJ, Schorl C, Freiman RN, Boekelheide K. All-Trans Retinoic Acid Disrupts Development in Ex Vivo Cultured Fetal Rat Testes. I: Altered Seminiferous Cord Maturation and Testicular Cell Fate. Toxicol Sci 2019; 167:546-558. [PMID: 30329139 PMCID: PMC6358251 DOI: 10.1093/toxsci/kfy260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Exposure to excess retinoic acid (RA) disrupts the development of the mammalian testicular seminiferous cord. However, the molecular events surrounding RA-driven loss of cord structure have not previously been examined. To investigate the mechanisms associated with this adverse developmental effect, fetal rat testes were isolated on gestational day 15, after testis determination and the initiation of cord development, and cultured in media containing all-trans RA (ATRA; 10-8 to 10-6 M) or vehicle for 3 days. ATRA exposure resulted in a concentration-dependent decrease in the number of seminiferous cords per testis section and number of germ cells, assessed by histopathology and immunohistochemistry. Following 1 day of culture, genome-wide expression profiling by microarray demonstrated that ATRA exposure altered biological processes related to retinoid metabolism and gonadal sex determination. Real-time RT-PCR analysis confirmed that ATRA enhanced the expression of the key ovarian development gene Wnt4 and the antitestis gene Nr0b1 in a concentration-dependent manner. After 3 days of culture, ATRA-treated testes contained both immunohistochemically DMRT1-positive and FOXL2-positive somatic cells, providing evidence of disrupted testicular cell fate maintenance following ATRA exposure. We conclude that exogenous RA disrupts seminiferous cord development in ex vivo cultured fetal rat testes, resulting in a reduction in seminiferous cord number, and interferes with maintenance of somatic cell fate by enhancing expression of factors that promote ovarian development.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Edward Dere
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
- Division of Urology, Rhode Island Hospital, Providence, Rhode Island 02903
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Christoph Schorl
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912
| | - Richard N Freiman
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
20
|
Bejarano I, Rodríguez AB, Pariente JA. Apoptosis Is a Demanding Selective Tool During the Development of Fetal Male Germ Cells. Front Cell Dev Biol 2018; 6:65. [PMID: 30003081 PMCID: PMC6031705 DOI: 10.3389/fcell.2018.00065] [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: 04/03/2018] [Accepted: 06/08/2018] [Indexed: 02/06/2023] Open
Abstract
Apoptosis is widely known to play a major role on diseases related to male infertility. Diseases of the male genital tract as defective spermatogenesis, decreased sperm motility, sperm DNA fragmentation, testicular torsion, varicocele and immunological infertility are strongly related to apoptotic cell death. Apoptosis must not be considered only as a fail on germ cell physiology or a secondary effect of certain pathologies and exogenous hazardous agents. Apoptosis orchestrates correct function and development of the male germ cell from the early embryonic stages of gonadal differentiation to the fertilization. In this review we have tried to address a reading frame of the main knowledge about apoptosis in male germ cell development. Focussing on mechanisms concerning cellular apoptosis, which are independent of exogenous stimuli, we aimed to highlight that apoptosis is a selective instrument that guarantees the delivery of genetic message to offspring.
Collapse
Affiliation(s)
| | | | - José A. Pariente
- Neuroimmunophysiology and Chrononutrition Research Group, Department of Physiology, Faculty of Science, University of Extremadura, Badajoz, Spain
| |
Collapse
|
21
|
Western PS. Epigenomic drugs and the germline: Collateral damage in the home of heritability? Mol Cell Endocrinol 2018; 468:121-133. [PMID: 29471014 DOI: 10.1016/j.mce.2018.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 02/07/2023]
Abstract
The testis and ovary provide specialised environments that nurture germ cells and facilitate their maturation, culminating in the production of mature gametes that can found the following generation. The sperm and egg not only transmit genetic information, but also epigenetic modifications that affect the development and physiology of offspring. Importantly, the epigenetic information contained in mature sperm and oocytes can be influenced by a range of environmental factors, such as diet, chemicals and drugs. An increasing range of studies are revealing how gene-environment interactions are mediated through the germline. Outside the germline, altered epigenetic state is common in a range of diseases, including many cancers. As epigenetic modifications are reversible, pharmaceuticals that directly target epigenetic modifying proteins have been developed and are delivering substantial benefits to patients, particularly in oncology. While providing the most effective patient treatment is clearly the primary concern, some patients will want to conceive children after treatment. However, the impacts of epigenomic drugs on the male and female gametes are poorly understood and whether these drugs will have lasting effects on patients' germline epigenome and subsequent offspring remains largely undetermined. Currently, evidence based clinical guidelines for use of epigenomic drugs in patients of reproductive age are limited in this context. Developing a deeper understanding of the epigenetic mechanisms regulating the germline epigenome and its impact on inherited traits and disease susceptibility is required to determine how specific epigenomic drugs might affect the germline and inheritance. Understanding these potential effects will facilitate the development of informed clinical guidelines appropriate for the use of epigenomic drugs in patients of reproductive age, ultimately improving the safety of these therapies in the clinic.
Collapse
Affiliation(s)
- Patrick S Western
- Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
22
|
Pui HP, Saga Y. NANOS2 acts as an intrinsic regulator of gonocytes-to-spermatogonia transition in the murine testes. Mech Dev 2018; 149:27-40. [DOI: 10.1016/j.mod.2018.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 02/07/2023]
|
23
|
Abstract
Sexual reproduction crucially depends on the production of sperm in males and oocytes in females. Both types of gamete arise from the same precursor, the germ cells. We review the events that characterize the development of germ cells during fetal life as they commit to, and prepare for, oogenesis or spermatogenesis. In females, fetal germ cells enter meiosis, whereas in males they delay meiosis and instead lose pluripotency, activate an irreversible program of prospermatogonial differentiation, and temporarily cease dividing. Both pathways involve sex-specific molecular signals from the somatic cells of the developing gonads and a suite of intrinsic receptors, signal transducers, transcription factors, RNA stability factors, and epigenetic modulators that act in complex, interconnected positive and negative regulatory networks. Understanding these networks is important in the contexts of the etiology, diagnosis, and treatment of infertility and gonadal cancers, and in efforts to augment human and animal fertility using stem cell approaches.
Collapse
Affiliation(s)
- Cassy Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia;
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
24
|
Induction of male germ cell-like lineage from chicken fetal bone marrow stem cells with chicken testis extract. BIOTECHNOL BIOPROC E 2017. [DOI: 10.1007/s12257-016-0629-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
25
|
Wen Y, He W, Jiang M, Zeng M, Cai L. Deriving cells expressing markers of female germ cells from premature ovarian failure patient-specific induced pluripotent stem cells. Regen Med 2017; 12:143-152. [PMID: 28244827 DOI: 10.2217/rme-2016-0074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: We proposed a two-step protocol for deriving cells expressing markers of female germ cells (FGCs) from premature ovarian failure patient-specific induced pluripotent stem cells (POF-iPSCs). Material & methods: We cultured POF-iPSCs in suspension and pretreated them with TGFβ-1 (1 ng/ml) for 2 days and continued with both TGFβ-1 and BMP4 (50 ng/ml) for 5 more days. Then changed to media containing retinoic acid (1 μM) and 5% follicular fluid for another 7 days. Expression of markers of different stages of FGCs were detected. Results: c-KIT, STELLA/DPPA3, VASA/DDX4, SCP3, GDF9 and ZP3 were positively detected and statistically significant different when compared with control groups. Conclusion: Our in vitro system was beneficial for POF-iPSCs differentiated cells to express STELLA, VASA and SCP3, which were the markers of meiosis initiation of FGCs.
Collapse
Affiliation(s)
- Yanfei Wen
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 6 East Longkou Road, Guangzhou, China
- Center for Reproductive Medicine, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China
| | - Wen He
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 6 East Longkou Road, Guangzhou, China
| | - Manbo Jiang
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 6 East Longkou Road, Guangzhou, China
- Department of Reproductive Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Minhui Zeng
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 6 East Longkou Road, Guangzhou, China
- Memorial hospital of Sun Yat-sen University, Guangzhou, China
| | - Liuhong Cai
- Center for Reproductive Medicine, The Third Affiliated Hospital, Sun Yat-sen University, 6 East Longkou Road, Guangzhou, China
| |
Collapse
|
26
|
Teletin M, Vernet N, Ghyselinck NB, Mark M. Roles of Retinoic Acid in Germ Cell Differentiation. Curr Top Dev Biol 2017; 125:191-225. [PMID: 28527572 DOI: 10.1016/bs.ctdb.2016.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The modalities of gametogenesis differ markedly between sexes. Female are born with a definitive reserve of oocytes whose size is crucial to ensure fertility. Male fertility, in contrast, relies on a tightly regulated balance between germ cell self-renewal and differentiation, which operates throughout life, according to recurring spatial and temporal patterns. Genetic and pharmacological studies conducted in the mouse and discussed in this review have revealed that all-trans retinoic acid and its nuclear receptors are major players of gametogenesis and are instrumental to fertility in both sexes.
Collapse
Affiliation(s)
- Marius Teletin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Centre National de la Recherche Scientifique (CNRS), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; Université de Strasbourg (UNISTRA), Strasbourg, France; Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France
| | - Nadège Vernet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Centre National de la Recherche Scientifique (CNRS), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; Université de Strasbourg (UNISTRA), Strasbourg, France
| | - Norbert B Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Centre National de la Recherche Scientifique (CNRS), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; Université de Strasbourg (UNISTRA), Strasbourg, France
| | - Manuel Mark
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Centre National de la Recherche Scientifique (CNRS), Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France; Université de Strasbourg (UNISTRA), Strasbourg, France; Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France.
| |
Collapse
|
27
|
Kasimanickam VR. Expression of retinoic acid-metabolizing enzymes, ALDH1A1, ALDH1A2, ALDH1A3, CYP26A1, CYP26B1 and CYP26C1 in canine testis during post-natal development. Reprod Domest Anim 2016; 51:901-909. [PMID: 27569851 DOI: 10.1111/rda.12756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022]
Abstract
Mammalian spermatogenesis involves highly regulated temporal and spatial dynamics, carefully controlled by several signalling processes. Retinoic acid (RA) signalling could have a critical role in spermatogenesis by promoting spermatogonia differentiation, adhesion of germ cells to Sertoli cells, and release of mature spermatids. An optimal testicular RA concentration is maintained by retinaldehyde dehydrogenases (ALDHs), which oxidize RA precursors to produce RA, whereas the CYP26 class of enzymes catabolizes (oxidize) RA into inactive metabolites. The objective was to elucidate gene expression of these RA-metabolizing enzymes (ALDH1A1, ALDH1A2, ALDH1A3, CYP26A1, CYP26B1 and CYP26C1) and their protein presence in testes of young, peripubertal and adult dogs. Genes encoding RA-synthesizing isozymes ALDH1A1, ALDH1A2 and ALDH1A3 and RA-catabolizing isomers CYP26A1, CYP26B1 and CYP26C1 were expressed in testis at varying levels during testicular development from birth to adulthood in dogs. Based on detailed analyses of mRNA expression patterns, ALDH1A2 was regarded as a primary RA-synthesizing enzyme and CYP26B1 as a critical RA-hydrolysing enzyme; presumably, these genes have vital roles in maintaining RA homeostasis, which is imperative to spermatogenesis and other testicular functions in post-natal canine testis.
Collapse
Affiliation(s)
- V R Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.,Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
28
|
Brill JA, Yildirim S, Fabian L. Phosphoinositide signaling in sperm development. Semin Cell Dev Biol 2016; 59:2-9. [PMID: 27321976 DOI: 10.1016/j.semcdb.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/15/2016] [Indexed: 01/15/2023]
Abstract
Phosphatidylinositol phosphates (PIPs)1 are membrane lipids with crucial roles during cell morphogenesis, including the establishment of cytoskeletal organization, membrane trafficking, cell polarity, cell-cycle control and signaling. Recent studies in mice (Mus musculus), fruit flies (Drosophila melanogaster) and other organisms have defined germ cell intrinsic requirements for these lipids and their regulatory enzymes in multiple aspects of sperm development. In particular, PIP levels are crucial in germline stem cell maintenance, spermatogonial proliferation and survival, spermatocyte cytokinesis, spermatid polarization, sperm tail formation, nuclear shaping, and production of mature, motile sperm. Here, we briefly review the stages of spermatogenesis and discuss the roles of PIPs and their regulatory enzymes in male germ cell development.
Collapse
Affiliation(s)
- Julie A Brill
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sukriye Yildirim
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| | - Lacramioara Fabian
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| |
Collapse
|
29
|
Regulators in the apoptotic pathway during spermatogenesis: Killers or guards? Gene 2016; 582:97-111. [DOI: 10.1016/j.gene.2016.02.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/19/2016] [Accepted: 02/03/2016] [Indexed: 01/24/2023]
|
30
|
Kato Y, Katsuki T, Kokubo H, Masuda A, Saga Y. Dazl is a target RNA suppressed by mammalian NANOS2 in sexually differentiating male germ cells. Nat Commun 2016; 7:11272. [PMID: 27072294 PMCID: PMC4833867 DOI: 10.1038/ncomms11272] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 03/08/2016] [Indexed: 12/26/2022] Open
Abstract
Evolutionally conserved Nanos RNA-binding proteins play crucial roles in germ cell development. While a mammalian Nanos family protein, NANOS2, is required for sexual differentiation of male (XY) germ cells in mice, the underlying mechanisms and the identities of its target RNAs in vivo remain elusive. Using comprehensive microarray analysis and a bacterial artificial chromosome transgenic system, here we identify Dazl, a germ cell-specific gene encoding an RNA-binding protein implicated in translation, as a crucial target of NANOS2. Importantly, removal of the Dazl 3'-untranslated region in XY germ cells stabilizes the Dazl mRNA, resulting in elevated meiotic gene expression, abnormal resumption of the cell cycle and impaired processing-body formation, reminiscent of Nanos2-knockout phenotypes. Furthermore, our data suggest that NANOS2 acts as an antagonist of the DAZL protein. We propose a dual system of NANOS2-mediated suppression of Dazl expression as a pivotal molecular mechanism promoting sexual differentiation of XY germ cells.
Collapse
Affiliation(s)
- Yuzuru Kato
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, SOKENDAI, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Takeo Katsuki
- Kavli Institute for Brain and Mind, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Hiroki Kokubo
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Aki Masuda
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Yumiko Saga
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, SOKENDAI, Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
31
|
Xu L, Chang G, Ma T, Wang H, Chen J, Li Z, Guo X, Wan F, Ren L, Lu W, Chen G. Piwil1 mediates meiosis during spermatogenesis in chicken. Anim Reprod Sci 2016; 166:99-108. [DOI: 10.1016/j.anireprosci.2016.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 10/22/2022]
|
32
|
Abstract
Germ cells are the precursors of the sperm and oocytes and hence are critical for survival of the species. In mammals, they are specified during fetal life, migrate to the developing gonads and then undergo a critical period during which they are instructed, by the soma, to adopt the appropriate sexual fate. In a fetal ovary, germ cells enter meiosis and commit to oogenesis, whereas in a fetal testis, they avoid entry into meiosis and instead undergo mitotic arrest and mature toward spermatogenesis. Here, we discuss what we know so far about the regulation of sex-specific differentiation of germ cells, considering extrinsic molecular cues produced by somatic cells, as well as critical intrinsic changes within the germ cells. This review focuses almost exclusively on our understanding of these events in the mouse model.
Collapse
Affiliation(s)
| | - Josephine Bowles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
33
|
Abby E, Tourpin S, Ribeiro J, Daniel K, Messiaen S, Moison D, Guerquin J, Gaillard JC, Armengaud J, Langa F, Toth A, Martini E, Livera G. Implementation of meiosis prophase I programme requires a conserved retinoid-independent stabilizer of meiotic transcripts. Nat Commun 2016; 7:10324. [PMID: 26742488 PMCID: PMC4729902 DOI: 10.1038/ncomms10324] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/27/2015] [Indexed: 12/28/2022] Open
Abstract
Sexual reproduction is crucially dependent on meiosis, a conserved, specialized cell division programme that is essential for the production of haploid gametes. Here we demonstrate that fertility and the implementation of the meiotic programme require a previously uncharacterized meiosis-specific protein, MEIOC. Meioc invalidation in mice induces early and pleiotropic meiotic defects in males and females. MEIOC prevents meiotic transcript degradation and interacts with an RNA helicase that binds numerous meiotic mRNAs. Our results indicate that proper engagement into meiosis necessitates the specific stabilization of meiotic transcripts, a previously little-appreciated feature in mammals. Remarkably, the upregulation of MEIOC at the onset of meiosis does not require retinoic acid and STRA8 signalling. Thus, we propose that the complete induction of the meiotic programme requires both retinoic acid-dependent and -independent mechanisms. The latter process involving post-transcriptional regulation likely represents an ancestral mechanism, given that MEIOC homologues are conserved throughout multicellular animals. Meiosis is a cell division program that produces haploid gametes and is initiated by a retinoic acid-dependent process. Here the authors report that a meiosis-specific protein, MEIOC, is upregulated in a retinoic acid-independent manner and is required to stabilise meiosis-specific transcripts.
Collapse
Affiliation(s)
- Emilie Abby
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Sophie Tourpin
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Jonathan Ribeiro
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Katrin Daniel
- Molecular Cell Biology Group/Experimental Center, Institute of Physiological Chemistry, Medical School, MTZ, Dresden University of Technology, Fiedlerstrasse 42, Dresden 01307, Germany
| | - Sébastien Messiaen
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Delphine Moison
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Justine Guerquin
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Jean-Charles Gaillard
- CEA, DSV/IBITEC-S/SPI/Li2D, Laboratory 'Innovative Technologies for Detection and Diagnostic', CEA-Marcoule, BP 17171, Bagnols-sur-Cèze F-30200, France
| | - Jean Armengaud
- CEA, DSV/IBITEC-S/SPI/Li2D, Laboratory 'Innovative Technologies for Detection and Diagnostic', CEA-Marcoule, BP 17171, Bagnols-sur-Cèze F-30200, France
| | - Francina Langa
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, Paris 75015, France
| | - Attila Toth
- Molecular Cell Biology Group/Experimental Center, Institute of Physiological Chemistry, Medical School, MTZ, Dresden University of Technology, Fiedlerstrasse 42, Dresden 01307, Germany
| | - Emmanuelle Martini
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| | - Gabriel Livera
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, UMR-967, BP 6, Fontenay-aux-Roses 92265, France.,CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses 92265, France.,INSERM, Unité 967, Fontenay-aux-Roses F-92265, France.,Université Paris-Sud, UMR-967, Fontenay-aux-Roses F-92265, France
| |
Collapse
|
34
|
Çinar L, Kartal D, Ergin C, Aksoy H, Karadag MA, Aydin T, Cinar E, Borlu M. The effect of systemic isotretinoin on male fertility. Cutan Ocul Toxicol 2015; 35:296-9. [PMID: 26653640 DOI: 10.3109/15569527.2015.1119839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND/OBJECTIVE Acne vulgaris is one of the most common diseases of the youth. Systemic isotretinoin is the only drug which acts on all of the etiopathogenic mechanisms of acne. Isotretinoin has some well-known side effects. Besides these, there is a suspicion whether it causes infertility or not. In this study, we aimed to evaluate the effects of systemic isotretinoin on male fertility. METHODS Eighty one male patients, who were older than 18 years of age, and had severe or refractory acne vulgaris were included in the study. They were given a total dose of 120 mg/kg of systemic isotretinoin over a period of six months. Before and after the study, the spermiogram parameters of the patients were evaluated to show any possible effect on male fertility. The patients' total testosterone, follicle stimulating hormone and luteinizing hormone levels were also evaluated. RESULTS All of the spermiogram parameters changed positively (p < 0.05). There was no significant change in the hormone levels. CONCLUSION Systemic isotretinoin has a positive effect on male fertility. Since the hormone levels did not change significantly, this positive effect of isotretinoin is not via the hypothalamic-pituitary-gonadal axis but can be due to its regenerative and proliferative effects on the testes.
Collapse
Affiliation(s)
- Levent Çinar
- a Faculty of Medicine , Dermatology and Venereology Department, Erciyes University , Kayseri , Turkey
| | - Demet Kartal
- a Faculty of Medicine , Dermatology and Venereology Department, Erciyes University , Kayseri , Turkey
| | - Can Ergin
- b Dermatology and Venereology Department, Diskapi Yildirim Beyazit Education and Research Hospital , Ankara , Turkey
| | - Huseyin Aksoy
- c Obstetrics and Gynecology Department, Kayseri Military Hospital , Kayseri , Turkey
| | - Mert Ali Karadag
- d Urology Department, Faculty of Medicine, Kafkas University , Kayseri , Turkey , and
| | - Turgut Aydin
- e Obstetrics and Gynecology Department, Kayseri Acibadem Hospital , Kayseri , Turkey
| | - Elif Cinar
- e Obstetrics and Gynecology Department, Kayseri Acibadem Hospital , Kayseri , Turkey
| | - Murat Borlu
- a Faculty of Medicine , Dermatology and Venereology Department, Erciyes University , Kayseri , Turkey
| |
Collapse
|
35
|
Niedenberger BA, Busada JT, Geyer CB. Marker expression reveals heterogeneity of spermatogonia in the neonatal mouse testis. Reproduction 2015; 149:329-38. [PMID: 25737569 DOI: 10.1530/rep-14-0653] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Prospermatogonia transition to type A spermatogonia, which provide the source for the spermatogonial stem cell (SSC) pool. A percentage of these type A spermatogonia then differentiate to enter meiosis as spermatocytes by ∼P10. It is currently unclear as to when these distinct populations are initially formed in the neonatal testis, and when the expression of markers both characteristic of and required for the adult undifferentiated and differentiating states is established. In this study, we compared expression of known spermatogonial cell fate markers during normal development and in response to the differentiation signal provided by retinoic acid (RA). We found that some markers for the undifferentiated state (ZBTB16/PLZF and CDH1) were expressed in nearly all spermatogonia from P1 through P7. In contrast, differentiation markers (STRA8 and KIT) appeared in a subset of spermatogonia at P4, coincident with the onset of RA signaling. GFRA1, which was present in nearly all prospermatogonia at P1, was only retained in STRA8/KIT- spermatogonia. From P4 through P10, there was a great deal of heterogeneity in the male germ cell population in terms of expression of markers, as markers characteristic of the undifferentiated (except GFRA1) and differentiating states were co-expressed through this interval. After P10, these fate markers diverged to mark distinct populations of undifferentiated and differentiating spermatogonia, and this pattern was maintained in juvenile (P18) and adult (P>60) testes. Taken together, these results reveal that the spermatogonia population is heterogeneous during the first wave of spermatogenesis, and indicate that neonatal spermatogonia may not serve as an ideal substitute for studying the function of adult spermatogonia.
Collapse
Affiliation(s)
- Bryan A Niedenberger
- Department of Anatomy and Cell Biology Brody School of Medicine and East Carolina Diabetes and Obesity Institute East Carolina University, Greenville, North Carolina 27834, USA
| | - Jonathan T Busada
- Department of Anatomy and Cell Biology Brody School of Medicine and East Carolina Diabetes and Obesity Institute East Carolina University, Greenville, North Carolina 27834, USA
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology Brody School of Medicine and East Carolina Diabetes and Obesity Institute East Carolina University, Greenville, North Carolina 27834, USA Department of Anatomy and Cell Biology Brody School of Medicine and East Carolina Diabetes and Obesity Institute East Carolina University, Greenville, North Carolina 27834, USA
| |
Collapse
|
36
|
Busada JT, Geyer CB. The Role of Retinoic Acid (RA) in Spermatogonial Differentiation. Biol Reprod 2015; 94:10. [PMID: 26559678 PMCID: PMC4809555 DOI: 10.1095/biolreprod.115.135145] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/06/2015] [Indexed: 12/22/2022] Open
Abstract
Retinoic acid (RA) directs the sequential, but distinct, programs of spermatogonial differentiation and meiotic differentiation that are both essential for the generation of functional spermatozoa. These processes are functionally and temporally decoupled, as they occur in distinct cell types that arise over a week apart, both in the neonatal and adult testis. However, our understanding is limited in terms of what cellular and molecular changes occur downstream of RA exposure that prepare differentiating spermatogonia for meiotic initiation. In this review, we describe the process of spermatogonial differentiation and summarize the current state of knowledge regarding RA signaling in spermatogonia.
Collapse
Affiliation(s)
- Jonathan T Busada
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
37
|
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: 1.0] [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
|
38
|
Hogg K, Western PS. Differentiation of Fetal Male Germline and Gonadal Progenitor Cells Is Disrupted in Organ Cultures Containing Knockout Serum Replacement. Stem Cells Dev 2015; 24:2899-911. [PMID: 26393524 DOI: 10.1089/scd.2015.0196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Germ cells occupy a unique place in development through their requirement to maintain underlying totipotency while producing highly differentiated gametes. This is reflected in the expression of key regulators of pluripotency in the fetal germline and the ability of these cells to form pluripotent stem cells and germ cell tumors. Culture of whole fetal testes, including germ cells, provides a key model for studying gonad and germline development, but it is critical that such models mimic physiological development as closely as possible. We aimed to determine the effects of differing culture conditions, including serum-free and serum-containing conditions, on fetal germ cell and testis development. We tested a commonly used model that employs knockout serum replacement (KSR) to provide more defined culture conditions than media containing fetal bovine serum (FBS). In FBS conditions, cell cycle parameters in germ and Sertoli cells closely resembled normal development. In contrast, KSR significantly inhibited male germ cell entry into mitotic arrest, a key milestone in male germline development. Moreover, KSR disrupted molecular control of cell cycle and inhibited the transcription of a range of male germ cell differentiation markers. In the somatic compartment, KSR stimulated proliferation and inhibited differentiation in Sertoli cells. These data demonstrate that KSR substantially alters germ and somatic cell differentiation in fetal testis culture and should not be used to replicate normal gonadal development. In contrast, basal media with or without serum support germ and supporting cell differentiation and cell cycle dynamics that are in line with in vivo characteristics.
Collapse
Affiliation(s)
- Kirsten Hogg
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Victoria, Australia .,2 Department of Molecular and Translational Science, Monash University , Melbourne, Victoria, Australia
| | - Patrick S Western
- 1 Centre for Genetic Diseases, Hudson Institute of Medical Research , Clayton, Victoria, Australia .,2 Department of Molecular and Translational Science, Monash University , Melbourne, Victoria, Australia
| |
Collapse
|
39
|
RA induces differentiation of multipotent P19 cells towards male germ cell. In Vitro Cell Dev Biol Anim 2014; 51:85-91. [PMID: 25537091 DOI: 10.1007/s11626-014-9746-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 03/03/2014] [Indexed: 01/04/2023]
Abstract
Generating male germ cells in vitro from multipotent stem cells is still a challenge for stem cell biologists. The difficulty is caused by the lack of knowledge about spermatogenesis molecular-controlling mechanisms. In vivo, PGCs differentiate into male germ cells in a very complicated environment through many middle steps. In this study, we use the pluripotent p19 cells to test their responses to different retinoic acid (RA) concentrations by evaluating markers for stem cells (bmp4, egr3), primordial germ cells (ddx4), spermatogonia (c-kit), premeiotic cells (stra8), and male germ cells (dazl and plzf). We have found that cyp26b1, which will catalyze RA, increases dramatically in p19 cells 1 d after RA treatment. Bmp3, egr3, and stra8 are stimulated after 1 d of RA treatment and then recover to normal after 3 d of RA treatment. C-kit keeps being expressed when treated with 10 nM-4 μM RA. Dazl and plzf are gained after 3 d of stimulation. The morphology of RA (100 nM-4 μM)-treated cells changes distinctively, and cell colonies are formed. Typical neural cell-like and germ cell-like morphologies appear in the 100 nM and 4 μM RA groups, respectively. We conclude that 100-500 nM RA can cause responses in p19 cells, but a high concentration of RA (1-4 μM) can drive these pluripotent cells' differentiation towards male germ cells. However, high concentrations of RA are also toxic. Some colonies that survived from 4 μM RA begin to express ddx4 and c-kit. Selection of the c-kit(+), dazl(+), and ddx4(+) cells after RA stimulation and creating a special culture medium for their propagation might benefit successful spermatogenesis induction in vitro.
Collapse
|
40
|
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: 2.8] [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
|
41
|
Busada JT, Kaye EP, Renegar RH, Geyer CB. Retinoic acid induces multiple hallmarks of the prospermatogonia-to-spermatogonia transition in the neonatal mouse. Biol Reprod 2014; 90:64. [PMID: 24478393 DOI: 10.1095/biolreprod.113.114645] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In mammals, most neonatal male germ cells (prospermatogonia) are quiescent and located in the center of the testis cords. In response to an unknown signal, prospermatogonia transition into spermatogonia, reenter the cell cycle, divide, and move to the periphery of the testis cords. In mice, these events occur by 3-4 days postpartum (dpp), which temporally coincides with the onset of retinoic acid (RA) signaling in the neonatal testis. RA has a pivotal role in initiating germ cell entry into meiosis in both sexes, yet little is known about the mechanisms and about cellular changes downstream of RA signaling. We examined the role of RA in mediating the prospermatogonia-to-spermatogonia transition in vivo and found 24 h of precocious RA exposure-induced germ cell changes mimicking those that occur during the endogenous transition at 3-4 dpp. These changes included: 1) spermatogonia proliferation; 2) maturation of cellular organelles; and 3), expression of markers characteristic of differentiating spermatogonia. We found that germ cell exposure to RA did not lead to cellular loss from apoptosis but rather resulted in a delay of ∼2 days in their entry into meiosis. Taken together, our results indicate that exogenous RA induces multiple hallmarks of the transition of prospermatogonia to spermatogonia prior to their entry into meiosis.
Collapse
Affiliation(s)
- Jonathan T Busada
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | | | | | | |
Collapse
|
42
|
Habert R, Muczynski V, Grisin T, Moison D, Messiaen S, Frydman R, Benachi A, Delbes G, Lambrot R, Lehraiki A, N'tumba-Byn T, Guerquin MJ, Levacher C, Rouiller-Fabre V, Livera G. Concerns about the widespread use of rodent models for human risk assessments of endocrine disruptors. Reproduction 2014; 147:R119-29. [PMID: 24497529 PMCID: PMC3959776 DOI: 10.1530/rep-13-0497] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal testis is a major target of endocrine disruptors (EDs). During the last 20 years, we have developed an organotypic culture system that maintains the function of the different fetal testis cell types and have used this approach as a toxicological test to evaluate the effects of various compounds on gametogenesis and steroidogenesis in rat, mouse and human testes. We named this test rat, mouse and human fetal testis assay. With this approach, we compared the effects of six potential EDs ((mono-(2-ethylhexyl) phthalate (MEHP), cadmium, depleted uranium, diethylstilboestrol (DES), bisphenol A (BPA) and metformin) and one signalling molecule (retinoic acid (RA)) on the function of rat, mouse and human fetal testis at a comparable developmental stage. We found that the response is similar in humans and rodents for only one third of our analyses. For instance, RA and MEHP have similar negative effects on gametogenesis in the three species. For another third of our analyses, the threshold efficient concentrations that disturb gametogenesis and/or steroidogenesis differ as a function of the species. For instance, BPA and metformin have similar negative effects on steroidogenesis in human and rodents, but at different threshold doses. For the last third of our analyses, the qualitative response is species specific. For instance, MEHP and DES affect steroidogenesis in rodents, but not in human fetal testis. These species differences raise concerns about the extrapolation of data obtained in rodents to human health risk assessment and highlight the need of rigorous comparisons of the effects in human and rodent models, when assessing ED risk.
Collapse
Affiliation(s)
- René Habert
- Unit of Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, BP 6, 92265 Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Saba R, Wu Q, Saga Y. CYP26B1 promotes male germ cell differentiation by suppressing STRA8-dependent meiotic and STRA8-independent mitotic pathways. Dev Biol 2014; 389:173-81. [PMID: 24576537 DOI: 10.1016/j.ydbio.2014.02.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 01/31/2014] [Accepted: 02/17/2014] [Indexed: 11/18/2022]
Abstract
Germ cell sex is defined by factors derived from somatic cells. CYP26B1 is known to be a male sex-promoting factor that inactivates retinoic acid (RA) in somatic cells. In CYP26B1-null XY gonads, germ cells are exposed to a higher level of RA than in normal XY gonads and this activates Stra8 to induce meiosis while male-specific gene expression is suppressed. However, it is unknown whether meiotic entry by an elevated level of RA is responsible for the suppression of male-type gene expression. To address this question, we have generated Cyp26b1/Stra8 double knockout (dKO) embryos. We successfully suppressed the induction of meiosis in CYP26B1-null XY germ cells by removing the Stra8 gene. Concomitantly, we found that the male genetic program represented by the expression of NANOS2 and DNMT3L was totally rescued in about half of dKO germ cells, indicating that meiotic entry causes the suppression of male differentiation. However, half of the germ cells still failed to enter the appropriate male pathway in the dKO condition. Using microarray analyses together with immunohistochemistry, we found that KIT expression was accompanied by mitotic activation, but was canceled by inhibition of the RA signaling pathway. Taken together, we conclude that inhibition of RA is one of the essential factors to promote male germ cell differentiation, and that CYP26B1 suppresses two distinct genetic programs induced by RA: a Stra8-dependent meiotic pathway, and a Stra8-independent mitotic pathway.
Collapse
Affiliation(s)
- Rie Saba
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan.
| | - Quan Wu
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI, 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Yumiko Saga
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
44
|
Feng CW, Bowles J, Koopman P. Control of mammalian germ cell entry into meiosis. Mol Cell Endocrinol 2014; 382:488-497. [PMID: 24076097 DOI: 10.1016/j.mce.2013.09.026] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 08/24/2013] [Accepted: 09/20/2013] [Indexed: 11/21/2022]
Abstract
Germ cells are unique in undergoing meiosis to generate oocytes and sperm. In mammals, meiosis onset is before birth in females, or at puberty in males, and recent studies have uncovered several regulatory steps involved in initiating meiosis in each sex. Evidence suggests that retinoic acid (RA) induces expression of the critical pre-meiosis gene Stra8 in germ cells of the fetal ovary, pubertal testis and adult testis. In the fetal testis, CYP26B1 degrades RA, while FGF9 further antagonises RA signalling to suppress meiosis. Failsafe mechanisms involving Nanos2 may further suppress meiosis in the fetal testis. Here, we draw together the growing knowledge relating to these meiotic control mechanisms, and present evidence that they are co-ordinately regulated and that additional factors remain to be identified. Understanding this regulatory network will illuminate not only how the foundations of mammalian reproduction are laid, but also how mis-regulation of these steps can result in infertility or germline tumours.
Collapse
Affiliation(s)
- Chun-Wei Feng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Josephine Bowles
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
45
|
Effects of vitamin A on in vitro maturation of pre-pubertal mouse spermatogonial stem cells. PLoS One 2013; 8:e82819. [PMID: 24349372 PMCID: PMC3857286 DOI: 10.1371/journal.pone.0082819] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 11/06/2013] [Indexed: 11/19/2022] Open
Abstract
Testicular tissue cryopreservation is the only potential option for fertility preservation in pre-pubertal boys exposed to gonadotoxic treatment. Completion of spermatogenesis after in vitro maturation is one of the future uses of harvested testicular tissue. The purpose of the current study was to evaluate the effects of vitamin A on in vitro maturation of fresh and frozen-thawed mouse pre-pubertal spermatogonial stem cells in an organ culture system. Pre-pubertal CD1 mouse fresh testes were cultured for 7 (D7), 9 (D9) and 11 (D11) days using an organ culture system. Basal medium was supplemented with different concentrations of retinol (Re) or retinoic acid (RA) alone or in combination. Seminiferous tubule morphology (tubule diameter, intra-tubular cell type), intra-tubular cell death and proliferation (PCNA antibody) and testosterone level were assessed at D7, D9 and D11. Pre-pubertal mouse testicular tissue were frozen after a soaking temperature performed at -7°C, -8°C or -9°C and after thawing, were cultured for 9 days, using the culture medium preserving the best fresh tissue functionality. Retinoic acid at 10-6M and retinol at 3.3.10-7M, as well as retinol 10-6M are favourable for seminiferous tubule growth, maintenance of intra-tubular cell proliferation and germ cell differentiation of fresh pre-pubertal mouse spermatogonia. Structural and functional integrity of frozen-thawed testicular tissue appeared to be well-preserved after soaking temperature at -8°C, after 9 days of organotypic culture using 10-6M retinol. RA and Re can control in vitro germ cell proliferation and differentiation. Re at a concentration of 10-6M maintains intra-tubular cell proliferation and the ability of spermatogonia to initiate spermatogenesis in fresh and frozen pre-pubertal mouse testicular tissue using a soaking temperature at -8°C. Our data suggested a possible human application for in vitro maturation of cryopreserved pre-pubertal testicular tissue.
Collapse
|
46
|
Rodríguez-Marí A, Cañestro C, BreMiller RA, Catchen JM, Yan YL, Postlethwait JH. Retinoic acid metabolic genes, meiosis, and gonadal sex differentiation in zebrafish. PLoS One 2013; 8:e73951. [PMID: 24040125 PMCID: PMC3769385 DOI: 10.1371/journal.pone.0073951] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/24/2013] [Indexed: 11/18/2022] Open
Abstract
To help understand the elusive mechanisms of zebrafish sex determination, we studied the genetic machinery regulating production and breakdown of retinoic acid (RA) during the onset of meiosis in gonadogenesis. Results uncovered unexpected mechanistic differences between zebrafish and mammals. Conserved synteny and expression analyses revealed that cyp26a1 in zebrafish and its paralog Cyp26b1 in tetrapods independently became the primary genes encoding enzymes available for gonadal RA-degradation, showing lineage-specific subfunctionalization of vertebrate genome duplication (VGD) paralogs. Experiments showed that zebrafish express aldh1a2, which encodes an RA-synthesizing enzyme, in the gonad rather than in the mesonephros as in mouse. Germ cells in bipotential gonads of all zebrafish analyzed were labeled by the early meiotic marker sycp3, suggesting that in zebrafish, the onset of meiosis is not sexually dimorphic as it is in mouse and is independent of Stra8, which is required in mouse but was lost in teleosts. Analysis of dead-end knockdown zebrafish depleted of germ cells revealed the germ cell-independent onset and maintenance of gonadal aldh1a2 and cyp26a1 expression. After meiosis initiated, somatic cell expression of cyp26a1 became sexually dimorphic: up-regulated in testes but not ovaries. Meiotic germ cells expressing the synaptonemal complex gene sycp3 occupied islands of somatic cells that lacked cyp26a1 expression, as predicted by the hypothesis that Cyp26a1 acts as a meiosis-inhibiting factor. Consistent with this hypothesis, females up-regulated cyp26a1 in oocytes that entered prophase-I meiotic arrest, and down-regulated cyp26a1 in oocytes resuming meiosis. Co-expression of cyp26a1 and the pluripotent germ cell stem cell marker pou5f1(oct4) in meiotically arrested oocytes was consistent with roles in mouse to promote germ cell survival and to prevent apoptosis, mechanisms that are central for tipping the sexual fate of gonads towards the female pathway in zebrafish.
Collapse
Affiliation(s)
- Adriana Rodríguez-Marí
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- Departament de Genètica, Universitat de Barcelona, Barcelona, Spain
| | - Cristian Cañestro
- Departament de Genètica, Universitat de Barcelona, Barcelona, Spain
- * E-mail: (JHP); (CC)
| | - Ruth A. BreMiller
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Julian M. Catchen
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Yi-Lin Yan
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - John H. Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- * E-mail: (JHP); (CC)
| |
Collapse
|
47
|
Culty M. Gonocytes, from the Fifties to the Present: Is There a Reason to Change the Name?1. Biol Reprod 2013; 89:46. [DOI: 10.1095/biolreprod.113.110544] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
48
|
Dong R, Yang S, Jiao J, Wang T, Shi H, Zhou L, Zhang Y, Wang D. Characterization of Stra8 in Southern catfish (Silurus meridionalis): evidence for its role in meiotic initiation. BMC Mol Biol 2013; 14:11. [PMID: 23697400 PMCID: PMC3668142 DOI: 10.1186/1471-2199-14-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/15/2013] [Indexed: 01/04/2023] Open
Abstract
Background RA (retinoic acid) signal pathway has been proved to be required for germ cell meiotic initiation in mammals, aves and amphibians. Stra8 (Stimulated by retinoic acid gene 8) is an important factor in RA signal pathway. However, the role of RA and Stra8 in germ cell meiotic initiation in teleosts is poorly characterized. Results In this study, the full length cDNA of Stra8 was cloned from Southern catfish (Silurus meridionalis), and its spatio-temporal expression profiles were analyzed. The Stra8 cDNA (1606 bp) includes 163 bp 5’-UTR (untranslated region), 456 bp 3’-UTR, and an ORF (open reading frame) of 987 bp, encoding a polypeptide of 328 aa. Phylogenetic analysis revealed its existence in some primitive teleosts, such as Siluriformes and Salmoniformes. Tissue distribution analysis by RT-PCR showed that Stra8 is specifically expressed in gonads. By real-time PCR, in situ hybridization and immunohistochemistry, the highest expression level of Stra8/Stra8 was detected in 50 and 130 dah (day after hatching), the premeiotic stage of germ cells in XX and XY gonads, respectively. Conclusions Our results suggest that Stra8 might be involved in germ cell meiotic initiation in S. meridionalis as it did in tetrapods.
Collapse
Affiliation(s)
- Ranran Dong
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Li M, Yu M, Zhu H, Song W, Hua J. The effects of Nanos2 on Boule and Stra8 in male germline stem cells (mGSCs). Mol Biol Rep 2013; 40:4383-9. [PMID: 23644984 DOI: 10.1007/s11033-013-2527-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/29/2013] [Indexed: 11/29/2022]
Abstract
The mitosis-meiosis switch is a key event in the differentiation of germ cells. Meiosis is important in development biology, however, it has not been clear what is the regulation mechanism in mammals. Our previous study showed that Boule could activate Stra8 directly and result in the meiosis initiation of dairy goat male germline stem cells (mGSCs). Nanos2, a RNA-binding protein, plays critical roles in the suppression of meiosis by preventing Stra8 expression and maintain the male germ cell development. The main purpose of this study was to explore whether Nanos2 represses Stra8 transcription through Boule or not. We found ectopic over-expression of Nanos2 in GC-1 and mGSCs down-regulated Stra8 transcription and translation, and Boule expression was not affected. It was in consistent with our expectation that RA could up-regulate Boule and Stra8 expression, but down-regulate Nanos2 expression in mGSCs. In dairy goat, the expression levels of Boule and Stra8 would rise with the increase of age, but the expression level of Nanos2 in 90 dpp and adult testis had not shown a clear change. In conclusion, Nanos2 represses Stra8 expression but not through Boule in dairy goat mGSCs.
Collapse
Affiliation(s)
- Mingzhao Li
- Key Lab for Animal Biotechnology of Agriculture Ministry, Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | | | | | | | | |
Collapse
|
50
|
In vitro and in vivo differentiation of induced pluripotent stem cells into male germ cells. Biochem Biophys Res Commun 2013; 433:286-91. [DOI: 10.1016/j.bbrc.2013.02.107] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 02/18/2013] [Indexed: 11/21/2022]
|