1
|
Estermann MA, Grimm SA, Kitakule AS, Rodriguez KF, Brown PR, McClelland K, Amato CM, Yao HHC. NR2F2 regulation of interstitial cell fate in the embryonic mouse testis and its impact on differences of sex development. Nat Commun 2025; 16:3987. [PMID: 40295478 PMCID: PMC12038043 DOI: 10.1038/s41467-025-59183-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Testicular fetal Leydig cells produce androgens essential for male reproductive development. Impaired fetal Leydig cell differentiation leads to differences of sex development including hypospadias, cryptorchidism, and infertility. Despite fetal Leydig cells are thought to originate from proliferating progenitor cells in the testis interstitium, the precise mechanisms governing the interstitial cells to fetal Leydig cell transition remain elusive. Using mouse models and single-nucleus multiomics, we find that fetal Leydig cells arise from a Nr2f2-positive interstitial population. Embryonic deletion of Nr2f2 in mouse testes results in differences of sex development, including dysgenic testes, Leydig cell hypoplasia, cryptorchidism, and hypospadias. By combining single-nucleus multiomics and NR2F2 ChIP-seq we find that NR2F2 promotes the progenitor fate while suppresses Leydig cell differentiation by modulating key transcription factors and downstream genes. Our findings establish Nr2f2 as a crucial regulator of fetal Leydig cell differentiation and provide molecular insights into differences of sex development linked to Nr2f2 mutations.
Collapse
Affiliation(s)
- Martín Andrés Estermann
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Sara A Grimm
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Abigail S Kitakule
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Karina F Rodriguez
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Paula R Brown
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Kathryn McClelland
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Ciro M Amato
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
- Department of Surgery, Division of Urology, University of Missouri, Columbia, MO, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA.
| |
Collapse
|
2
|
Lei T, Yang Y, Yang WX. Luteinizing Hormone Regulates Testosterone Production, Leydig Cell Proliferation, Differentiation, and Circadian Rhythm During Spermatogenesis. Int J Mol Sci 2025; 26:3548. [PMID: 40332028 PMCID: PMC12027374 DOI: 10.3390/ijms26083548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Male reproductive health, particularly the regulation of spermatogenesis, is controlled by a complex combination of factors, including luteinizing hormone (LH) and its effects on Leydig cells (LCs). LH stimulates testosterone synthesis in LCs, which is critical for maintaining spermatogenesis and male fertility. This review examines the pathways through which LH regulates testosterone production, LC proliferation, differentiation, and circadian rhythm in human and non-human species. In particular, the signaling pathways of luteinizing hormone involved in testosterone production are discussed. Additionally, we explore LH's role in sperm maturation and quality, emphasizing its clinical implications in treating hypogonadotropic hypogonadism and diagnosing gonadal dysfunctions such as androgen insensitivity syndrome and precocious puberty. Furthermore, the potential of LH in assisted reproductive technologies for improving sperm quality is discussed. By highlighting key molecular mechanisms, this work provides insights into the therapeutic potential of LH in addressing male infertility and conditions of LC dysfunction.
Collapse
Affiliation(s)
| | | | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; (T.L.); (Y.Y.)
| |
Collapse
|
3
|
Wilhelm D, Perea-Gomez A, Newton A, Chaboissier MC. Gonadal sex determination in vertebrates: rethinking established mechanisms. Development 2025; 152:dev204592. [PMID: 40162719 DOI: 10.1242/dev.204592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Sex determination and differentiation are fundamental processes that are not only essential for fertility but also influence the development of many other organs, and hence, are important for species diversity and survival. In mammals, sex is determined by the inheritance of an X or a Y chromosome from the father. The Y chromosome harbours the testis-determining gene SRY, and it has long been thought that its absence is sufficient for ovarian development. Consequently, the ovarian pathway has been treated as a default pathway, in the sense that ovaries do not have or need a female-determining factor. Recently, a female-determining factor has been identified in mouse as the master regulator of ovarian development. Interestingly, this scenario was predicted as early as 1983. In this Review, we discuss the model predicted in 1983, how the mechanisms and genes currently known to be important for sex determination and differentiation in mammals have changed or supported this model, and finally, reflect on what these findings might mean for sex determination in other vertebrates.
Collapse
Affiliation(s)
- Dagmar Wilhelm
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Aitana Perea-Gomez
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Axel Newton
- TIGRR Lab, The School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | | |
Collapse
|
4
|
O’Neil EV, Dupont SM, Capel B. The basic helix-loop-helix transcription factor TCF4 recruits the Mediator Complex to activate gonadal genes and drive ovarian development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640455. [PMID: 40093061 PMCID: PMC11908221 DOI: 10.1101/2025.02.28.640455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The bipotential gonad is the precursor organ to both the ovary and testis and develops as part of the embryonic urogenital system. In mice, gonadogenesis initiates around embryonic day 9.5 (E9.5), when coelomic epithelial (CE) cells overlaying the mesonephric ducts proliferate and acquire the competence to differentiate into the two main cell types of the embryonic gonad, the pre-supporting cells and interstitial cell lineages. While some transcription factors that drive gonadal cell fate are known, HLH factors have not been investigated in this capacity. In the present study, we found that HLH binding sites are highly represented upstream of gonadal genes. We investigated the HLH factor Transcription Factor 4 (TCF4) which is expressed in the CE and GATA4+ somatic cells in both sexes prior to sex determination. TCF4 is maintained in ovarian pre-supporting cells and interstitial cells of both sexes but is silenced specifically in male pre-supporting cells. To characterize TCF4's role in gonad differentiation in vivo, we acquired a mutant mouse model that lacks the TCF4 DNA-binding domain and assessed morphology of the gonads at E15.5. While mutants develop gonads, we observed sex-specific effects on the gonads. Relative to wildtype littermates, SOX9 expression was higher in the Sertoli cells of XY Tcf4 STOP/STOP mutant testes, while FOXL2 and NR2F2 were reduced in the supporting and interstitial cell lineages of XX Tcf4 STOP/STOP mutant ovaries, respectively. Furthermore, the supporting: interstitial cell ratio was altered in XX Tcf4 STOP/STOP ovaries. These effects may occur downstream of changes to epigenetic programming or gene expression in somatic gonadal cells in mutant mice, as TCF4 binds the Mediator complex, RNA polymerase holoenzyme, and chromatin remodelers in early somatic cells. We hypothesize that TCF4 drives a gonadal program that advances female fate but is specifically silenced in male supporting cells as these pathways diverge.
Collapse
Affiliation(s)
- EV O’Neil
- Department of Cell Biology, Duke University School of Medicine, Durham NC 27710
| | - SM Dupont
- Department of Cell Biology, Duke University School of Medicine, Durham NC 27710
| | - B Capel
- Department of Cell Biology, Duke University School of Medicine, Durham NC 27710
| |
Collapse
|
5
|
Wang D, Xiang Y, Zhu Z, Liu J, Wang Y, Xu Z, Chen S, Dai C, Feng J, Chen J, Ma Q, Yang P. Cytological Effects of Cadmium Poisoning and the Protective Effect of Quercetin: A Mechanism Exploration based on the Testicular Lamina Propria. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2025; 31:ozae103. [PMID: 39405407 DOI: 10.1093/mam/ozae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/19/2024] [Accepted: 09/15/2024] [Indexed: 02/26/2025]
Abstract
This comprehensive study delved into the detrimental effects of cadmium (Cd), a toxic heavy metal, on the testicular lamina propria (LP), a key player in spermatogenesis, and the maintenance of testicular stem cell niches. Utilizing transmission electron microscopy, immunohistochemistry, and double-labeling immunofluorescence, the research characterized the structural and cellular components of mouse testicular LP under Cd exposure and investigated the protective effects of quercetin. The findings illustrated that Cd exposure results in significant morphological and cellular modifications within the LP, including the apoptosis of peritubular myoid cells, an upsurge in CD34+ stromal cells displaying anti-apoptotic behaviors, and an excessive production of collagen Type I fibers and extracellular matrix. Remarkably, quercetin effectively counteracted these adverse changes by reversing apoptosis, reducing the proliferation of CD34+ stromal cells, and addressing fibrosis markers, thereby mitigating the cellular damage induced by Cd. This study not only highlighted the critical impact of apoptosis and fibrosis in Cd-related testicular damage but also elucidated the protective mechanism of quercetin, laying the groundwork for future clinical applications in addressing testicular damage from heavy metal poisoning through cellular therapeutics and pharmacological interventions.
Collapse
Affiliation(s)
- Dian Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Yi Xiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Zhaoxuan Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Jiyue Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Yisheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Zeyu Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Si Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Chunyuan Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Jiasen Feng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| | - Jie Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 Puzhu South Road, Nanjing 211816, China
| | - Qianhui Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, No. 320 Yueyang Road, Shanghai 200031, China
| | - Ping Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang No. 1, Nanjing 210095, China
| |
Collapse
|
6
|
Estermann MA, Grimm S, Kitakule A, Rodriguez K, Brown P, McClelland K, Amato C, Yao HHC. NR2F2 regulation of interstitial to fetal Leydig cell differentiation in the testis: insights into differences of sex development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613312. [PMID: 39345510 PMCID: PMC11429913 DOI: 10.1101/2024.09.16.613312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Testicular fetal Leydig cells are a specialized cell type responsible for embryo masculinization. Fetal Leydig cells produce androgens, that induce the differentiation of male reproductive system and sexual characteristics. Deficiencies in Leydig cell differentiation leads to various disorders of sex development and male reproductive defects such as ambiguous genitalia, hypospadias, cryptorchidism, and infertility. Fetal Leydig cells are thought to originate from proliferating progenitor cells in the testis interstitium, marked by genes like Arx , Pdgfra , Tcf21 and Wnt5a . However, the precise mechanisms governing the transition from interstitial cells to fetal Leydig cells remain elusive. Through integrated approaches involving mouse models and single-nucleus multiomic analyses, we discovered that fetal Leydig cells originate from a Nr2f2 -positive non-steroidogenic interstitial cell population. Embryonic deletion of Nr2f2 in mouse testes resulted in disorders of sex development, including dysgenic testes, Leydig cell hypoplasia, cryptorchidism, and hypospadias. We found that NR2F2 promotes the progenitor cell fate while suppresses Leydig cell differentiation by directly and indirectly controlling a cohort of transcription factors and downstream genes. Bioinformatic analyses of single-nucleus ATAC-seq and NR2F2 ChIP-seq data revealed putative transcription factors co-regulating the process of interstitial to Leydig cell differentiation. Collectively, our findings not only highlight the critical role of Nr2f2 in orchestrating the transition from interstitial cells to fetal Leydig cells, but also provide molecular insight into the disorders of sex development as a result of Nr2f2 mutations.
Collapse
|
7
|
Baalbaki G, Lim V, Gillet AP, Verner MA, Vaillancourt C, Caron-Beaudoin E, Delbes G. Trace elements alone or in mixtures associated with unconventional natural gas exploitation affect rat fetal steroidogenesis and testicular development in vitro. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 357:124393. [PMID: 38901820 DOI: 10.1016/j.envpol.2024.124393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Biomonitoring studies have shown that pregnant women living in regions of unconventional natural gas (UNG) exploitation have higher levels of trace elements. Whether developmental endocrine disruption can be expected at these exposure levels during pregnancy is unclear. In this study, we aimed to test the impact of five trace elements alone or in mixtures using in vitro cell- and tissue-based assays relevant to endocrine disruption and development. Manganese, aluminum, strontium, barium, and cobalt were tested at concentrations including those representatives of human fetal exposure. Using transactivation assays, none of the tested elements nor their mixture altered the human estrogen receptor 1 or androgen receptor genomic signalling. In the rat fetal testis assay, an organ culture system, cobalt (5 μg/l), barium (500 μg/l) and strontium (500 μg/l) significantly increased testosterone secretion. Cobalt and strontium were associated with hyperplasia and/or hypertrophy of fetal Leydig cells. Mixing the five elements at concentrations where none had an effect individually stimulated testosterone secretion by the rat fetal testis paralleled by the significant increase of 3β-hydroxysteroid dehydrogenase protein level in comparison to the vehicle control. The mechanisms involved may be specific to the fetal testis as no effect was observed in the steroidogenic H295R cells. Our data suggest that some trace elements in mixture at concentrations representative of human fetal exposure can impact testis development and function. This study highlights the potential risk posed by UNG operations, especially for the most vulnerable populations, pregnant individuals, and their fetus.
Collapse
Affiliation(s)
- Ghida Baalbaki
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Victoria Lim
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Antoine P Gillet
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Marc-André Verner
- Université de Montréal, Department of Occupational and Environmental Health, Montreal, QC, Canada; Centre de Recherche en Santé Publique, Université de Montréal et CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montreal, QC, Canada
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada; Research Center, CIUSSS du Nord-de-l'Île-de-Montréal, Montreal, QC, Canada
| | - Elyse Caron-Beaudoin
- Department of Health and Society, University of Toronto Scarborough, Toronto, ON, Canada; Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Geraldine Delbes
- Institut National de la Recherche Scientifique (INRS), Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada.
| |
Collapse
|
8
|
Suen HC, Ou F, Miu KK, Wang Z, Chan WY, Liao J. The single-cell chromatin landscape in gonadal cell lineage specification. BMC Genomics 2024; 25:464. [PMID: 38741085 DOI: 10.1186/s12864-024-10376-1] [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] [Received: 10/31/2023] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.
Collapse
Affiliation(s)
- Hoi Ching Suen
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Fanghong Ou
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Kai-Kei Miu
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zhangting Wang
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai-Yee Chan
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jinyue Liao
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
9
|
Jiang K, Jorgensen JS. Fetal Leydig cells: What we know and what we don't. Mol Reprod Dev 2024; 91:e23739. [PMID: 38480999 PMCID: PMC11135463 DOI: 10.1002/mrd.23739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 05/24/2024]
Abstract
During male fetal development, testosterone plays an essential role in the differentiation and maturation of the male reproductive system. Deficient fetal testosterone production can result in variations of sex differentiation that may cause infertility and even increased tumor incidence later in life. Fetal Leydig cells in the fetal testis are the major androgen source in mammals. Although fetal and adult Leydig cells are similar in their functions, they are two distinct cell types, and therefore, the knowledge of adult Leydig cells cannot be directly applied to understanding fetal Leydig cells. This review summarizes our current knowledge of fetal Leydig cells regarding their cell biology, developmental biology, and androgen production regulation in rodents and human. Fetal Leydig cells are present in basement membrane-enclosed clusters in between testis cords. They originate from the mesonephros mesenchyme and the coelomic epithelium and start to differentiate upon receiving a Desert Hedgehog signal from Sertoli cells or being released from a NOTCH signal from endothelial cells. Mature fetal Leydig cells produce androgens. Human fetal Leydig cell steroidogenesis is LHCGR (Luteinizing Hormone Chronic Gonadotropin Receptor) dependent, while rodents are not, although other Gαs -protein coupled receptors might be involved in rodent steroidogenesis regulation. Fetal steroidogenesis ceases after sex differentiation is completed, and some fetal Leydig cells dedifferentiate to serve as stem cells for adult testicular cell types. Significant gaps are acknowledged: (1) Why are adult and fetal Leydig cells different? (2) What are bona fide progenitor and fetal Leydig cell markers? (3) Which signaling pathways and transcription factors regulate fetal Leydig cell steroidogenesis? It is critical to discover answers to these questions so that we can understand vulnerable targets in fetal Leydig cells and the mechanisms for androgen production that when disrupted, leads to variations in sex differentiation that range from subtle to complete sex reversal.
Collapse
Affiliation(s)
- Keer Jiang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
10
|
Taelman J, Czukiewska SM, Moustakas I, Chang YW, Hillenius S, van der Helm T, van der Meeren LE, Mei H, Fan X, Chuva de Sousa Lopes SM. Characterization of the human fetal gonad and reproductive tract by single-cell transcriptomics. Dev Cell 2024; 59:529-544.e5. [PMID: 38295793 PMCID: PMC10898717 DOI: 10.1016/j.devcel.2024.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024]
Abstract
During human fetal development, sex differentiation occurs not only in the gonads but also in the adjacent developing reproductive tract. However, while the cellular composition of male and female human fetal gonads is well described, that of the adjacent developing reproductive tract remains poorly characterized. Here, we performed single-cell transcriptomics on male and female human fetal gonads together with the adjacent developing reproductive tract from first and second trimesters, highlighting the morphological and molecular changes during sex differentiation. We validated different cell populations of the developing reproductive tract and gonads and compared the molecular signatures between the first and second trimesters, as well as between sexes, to identify conserved and sex-specific features. Together, our study provides insights into human fetal sex-specific gonadogenesis and development of the reproductive tract beyond the gonads.
Collapse
Affiliation(s)
- Jasin Taelman
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Sylwia M Czukiewska
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Ioannis Moustakas
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Yolanda W Chang
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Sanne Hillenius
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Talia van der Helm
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Lotte E van der Meeren
- Department of Pathology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands; Department of Pathology, Erasmus Medical Center, 3015 GD Rotterdam, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands
| | - Xueying Fan
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands.
| | - Susana M Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 ZC Leiden, the Netherlands; Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium.
| |
Collapse
|
11
|
Diawara M, Arsenault A, Charette SA, Martin LJ. The transcription factors Creb1 and Cebpb regulate Sox9 promoter activity in TM4 Sertoli cells. Gene 2023; 873:147477. [PMID: 37172798 DOI: 10.1016/j.gene.2023.147477] [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: 02/21/2023] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
In Sertoli cells, the Sox9 gene is essential for testicular development and normal spermatogenesis. SOX9 is critical for postnatal Sertoli cells differentiation and proliferation in the testis. However, the molecular mechanisms that specifically regulate its expression are not entirely understood. Sox9 expression is regulated by CREB1 and CEBPB in other biological contexts such as during chondrogenesis and in rat thyroid follicular cells. We hypothesized that Sox9 promoter activity is regulated by CREB1 and CEBPB in Sertoli cells. Our results show that Sox9 expression is dependent on the activation of these transcription factors by the cAMP/PKA signaling pathway in TM4 Sertoli cells. Chromatin immunoprecipitation and promoter/reporter luciferase assays with 5' promoter deletions and site-directed mutagenesis demonstrated that CREB1 is being recruited to a DNA regulatory element at -141 bp of the Sox9 promoter region. Such regulation is dependent on the cAMP/PKA signaling pathway, resulting in phosphorylation of CREB1. Activation of Sox9 expression by CEBPB may involve its recruitment to the proximal promoter region by protein-protein interaction with CREB1. Thus, we have shown that the Sox9 promoter is being regulated by the transcription factors CREB1 and CEBPB in TM4 Sertoli cells and involve their recruitment to the proximal promoter region.
Collapse
Affiliation(s)
- Mariama Diawara
- Biology Department, Université de Moncton, Moncton, New-Brunswick E1A 3E9, Canada
| | - Aurélie Arsenault
- Biology Department, Université de Moncton, Moncton, New-Brunswick E1A 3E9, Canada
| | - Sabrina Ayoub Charette
- Department of Nutritional Science, Temerty Faculty of Medicine, University of Toronto, M5S 1A8; Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, Ontario M5C 2T2, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, Moncton, New-Brunswick E1A 3E9, Canada.
| |
Collapse
|
12
|
Workman S, Wilson MJ. RNA sequencing and expression analysis reveal a role for Lhx9 in the haploinsufficient adult mouse ovary. Mol Reprod Dev 2023; 90:295-309. [PMID: 37084273 DOI: 10.1002/mrd.23686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/26/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
Understanding the molecular pathways that underpin ovarian development and function is vital for improving the research approaches to investigating fertility. Despite a significant improvement in our knowledge of molecular activity in the ovary, many questions remain unanswered in the quest to understand factors influencing fertility and ovarian pathologies such as cancer. Here, we present an investigation into the expression and function of the developmental transcription factor LIM Homeobox 9 (LHX9) in the adult mouse ovary. We have characterized Lhx9 expression in several cell types of the mature ovary across follicle stages. To evaluate possible LHX9 function in the adult ovary, we investigated ovarian anatomy and transcription in an Lhx9+/- knockout mouse model displaying subfertility. Despite a lack of gross anatomical differences between genotypes, RNA-sequencing found that 90 differentially expressed genes between Lhx9+/ - and Lhx9+/+ mice. Gene ontology analyses revealed a reduced expression of genes with major roles in ovarian steroidogenesis and an increased expression of genes associated with ovarian cancer. Analysis of the ovarian epithelium revealed Lhx9+/ - mice have a disorganized epithelial phenotype, corresponding to a significant increase in epithelial marker gene expression. These results provide an analysis of Lhx9 in the adult mouse ovary, suggesting a role in fertility and ovarian epithelial cancer.
Collapse
Affiliation(s)
- Stephanie Workman
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Megan J Wilson
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Neirijnck Y, Sararols P, Kühne F, Mayère C, Weerasinghe Arachchige LC, Regard V, Nef S, Schedl A. Single-cell transcriptomic profiling redefines the origin and specification of early adrenogonadal progenitors. Cell Rep 2023; 42:112191. [PMID: 36862551 DOI: 10.1016/j.celrep.2023.112191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/13/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023] Open
Abstract
Adrenal cortex and gonads represent the two major steroidogenic organs in mammals. Both tissues are considered to share a common developmental origin characterized by the expression of Nr5a1/Sf1. The precise origin of adrenogonadal progenitors and the processes driving differentiation toward the adrenal or gonadal fate remain, however, elusive. Here, we provide a comprehensive single-cell transcriptomic atlas of early mouse adrenogonadal development including 52 cell types belonging to twelve major cell lineages. Trajectory reconstruction reveals that adrenogonadal cells emerge from the lateral plate rather than the intermediate mesoderm. Surprisingly, we find that gonadal and adrenal fates have already diverged prior to Nr5a1 expression. Finally, lineage separation into gonadal and adrenal fates involves canonical versus non-canonical Wnt signaling and differential expression of Hox patterning genes. Thus, our study provides important insights into the molecular programs of adrenal and gonadal fate choice and will be a valuable resource for further research into adrenogonadal ontogenesis.
Collapse
Affiliation(s)
- Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; Université Côte d'Azur, CNRS, INSERM, IBV, 06108 Nice, France.
| | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | | | - Violaine Regard
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland.
| | - Andreas Schedl
- Université Côte d'Azur, CNRS, INSERM, IBV, 06108 Nice, France.
| |
Collapse
|
14
|
Di Persio S, Neuhaus N. Human spermatogonial stem cells and their niche in male (in)fertility: novel concepts from single-cell RNA-sequencing. Hum Reprod 2023; 38:1-13. [PMID: 36409992 PMCID: PMC9825264 DOI: 10.1093/humrep/deac245] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
The amount of single-cell RNA-sequencing (scRNA-seq) data produced in the field of human male reproduction has steadily increased. Transcriptional profiles of thousands of testicular cells have been generated covering the human neonatal, prepubertal, pubertal and adult period as well as different types of male infertility; the latter include non-obstructive azoospermia, cryptozoospermia, Klinefelter syndrome and azoospermia factor deletions. In this review, we provide an overview of transcriptional changes in different testicular subpopulations during postnatal development and in cases of male infertility. Moreover, we review novel concepts regarding the existence of spermatogonial and somatic cell subtypes as well as their crosstalk and provide corresponding marker genes to facilitate their identification. We discuss the potential clinical implications of scRNA-seq findings, the need for spatial information and the necessity to corroborate findings by exploring other levels of regulation, including at the epigenetic or protein level.
Collapse
Affiliation(s)
- Sara Di Persio
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Münster, Germany
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Münster, Germany
| |
Collapse
|
15
|
Bhattacharya I, Dey S. Emerging concepts on Leydig cell development in fetal and adult testis. Front Endocrinol (Lausanne) 2023; 13:1086276. [PMID: 36686449 PMCID: PMC9851038 DOI: 10.3389/fendo.2022.1086276] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Leydig cells (Lc) reside in the interstitial compartment of the testis and are the target of Luteinising hormone (LH) for Testosterone (T) production, thus critically regulates male fertility. Classical histological studies have identified two morphologically different populations of Lc during testicular development [fetal (FLc) and adult (ALc)]. Recent progress in ex vivo cell/organ culture, genome-wide analysis, genetically manipulated mouse models, lineage tracing, and single-cell RNA-seq experiments have revealed the diverse cellular origins with differential transcriptomic and distinct steroidogenic outputs of these populations. FLc originates from both coelomic epithelium and notch-active Nestin-positive perivascular cells located at the gonad-mesonephros borders, and get specified as Nr5a1 (previously known as Ad4BP/SF-1) expressing cells by embryonic age (E) 12.5 days in fetal mouse testes. These cells produce androstenedione (precursor of T, due to lack of HSD17β3 enzyme) and play critical a role in initial virilization and patterning of the male external genitalia. However, in neonatal testis, FLc undergoes massive regression/dedifferentiation and gradually gets replaced by T-producing ALc. Very recent studies suggest a small fraction (5-20%) of FLc still persists in adult testis. Both Nestin-positive perivascular cells and FLc are considered to be the progenitor populations for ALc. This minireview article summarizes the current understanding of Lc development in fetal and adult testes highlighting their common or diverse cellular (progenitor/stem) origins with respective functional significance in both rodents and primates. (227 words).
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Periye, Kerala, India
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
16
|
Himelreich Perić M, Takahashi M, Ježek D, Cunha GR. Early development of the human embryonic testis. Differentiation 2023; 129:4-16. [PMID: 35961887 DOI: 10.1016/j.diff.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/25/2023]
Abstract
Human gonadal development culminating in testicular differentiation is described through analysis of histologic sections derived from 33-day to 20-week human embryos/fetuses, focusing on early development (4-8 weeks of gestation). Our study updates the comprehensive studies of Felix (1912), van Wagenen and Simpson (1965), and Juric-Lekic et al. (2013), which were published in books and thus are unsearchable via PubMed. Human gonads develop from the germinal ridge, a thickening of coelomic epithelium on the medial side of the urogenital ridge. The bilateral urogenital ridges contain elements of the mesonephric kidney, namely the mesonephric duct, mesonephric tubules, and mesonephric glomeruli. The germinal ridge, into which primordial germ cells migrate, is initially recognized as a thickening of coelomic epithelium on the urogenital ridge late in the 4th week of gestation. Subsequently, in the 5th week of gestation, a dense mesenchyme develops sub-adjacent to the epithelium of the germinal ridge, and together these elements bulge into the coelomic cavity forming bilateral longitudinal ridges attached to the urogenital ridges. During development, primordial cells migrate into the germinal ridge and subsequently into testicular cords that form within the featureless dense mesenchyme of the germinal ridge at 6-8 weeks of gestation. The initial low density of testicular cords seen at 8 weeks remodels into a dense array of testicular cords surrounded by α-actin-positive myoid cells during the second trimester. Human testicular development shares many features with that of mice being derived from 4 elements: coelomic epithelium, sub-adjacent mesenchyme, primordial germ cells, and the mesonephros.
Collapse
Affiliation(s)
- Marta Himelreich Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia.
| | - Marta Takahashi
- Department of Communication Sciences, Catholic University of Croatia, 10000, Zagreb, Croatia
| | - Davor Ježek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia; Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
17
|
Role of mesonephric contribution to mouse testicular development revisited. Differentiation 2023; 129:109-119. [PMID: 35000816 DOI: 10.1016/j.diff.2021.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 01/25/2023]
Abstract
The role of the mesonephros in testicular development was re-evaluated by growing embryonic day 11.5 (E11.5) mouse testes devoid of mesonephros for 8-21 days in vivo under the renal capsule of castrated male athymic nude mice. This method provides improved growth conditions relative to previous studies based upon short-term (4-7 days) organ culture. Meticulous controls involved wholemount examination of dissected E11.5 mouse testes as well as serial sections of dissected E11.5 mouse testes which were indeed shown to be devoid of mesonephros. As expected, grafts of E11.5 mouse testes with mesonephros attached formed seminiferous tubules and also contained mesonephric derivatives. Grafts of E11.5 mouse testes without associated mesonephros also formed seminiferous tubules and never contained mesonephric derivatives. The consistent absence of mesonephric derivatives in grafts of E11.5 mouse testes grafted alone is further proof of the complete removal of the mesonephros from the E11.5 mouse testes. The testicular tissues that developed in grafts of E11.5 mouse testes alone contained canalized seminiferous tubules composed of Sox9-positive Sertoli cells as well as GENA-positive germ cells. The seminiferous tubules were surrounded by α-actin-positive myoid cells, and the interstitial space contained 3βHSD-1-positive Leydig cells. Grafts of E11.5 GFP mouse testes into wild-type hosts developed GFP-positive vasculature indicating that E11.5 mouse testes contain vascular precursors. These results indicate that the E11.5 mouse testis contains precursor cells for Sertoli cells, Leydig cells, myoid cells and vasculature whose development and differentiation are independent of cells migrating from the E11.5 mesonephros.
Collapse
|
18
|
Cunha GR, Cao M, Aksel S, Derpinghaus A, Baskin LS. Mouse-human species differences in early testicular development and its implications. Differentiation 2023; 129:79-95. [PMID: 35667976 DOI: 10.1016/j.diff.2022.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/16/2022] [Accepted: 04/24/2022] [Indexed: 01/25/2023]
Abstract
The mouse has been used as a model of human organogenesis with the tacit assumption that morphogenetic and molecular mechanisms in mice are translatable to human organogenesis. While many morphogenetic and molecular mechanisms are shared in mice and humans, many anatomic, morphogenetic, and molecular differences have been noted. Two critical gaps in our knowledge prevent meaningful comparisons of mouse versus human testicular development: (a) human testicular development is profoundly under-represented in the literature, and (b) an absence of a detailed day-by-day ontogeny of mouse testicular development from E11.5 to E16.5 encompassing the ambisexual stage to seminiferous cord formation. To address these deficiencies, histologic and immunohistochemical studies were pursued in comparable stages of mouse and human testicular development with a particular emphasis on Leydig, Sertoli and myoid cells through review of the literature and new observations. For example, an androgen-receptor-positive testicular medulla is present in the developing human testis but not in the developing mouse testis. The human testicular medulla and associated mesonephros were historically described as the source of Sertoli cells in seminiferous cords. Consistent with this idea, the profoundly androgen receptor (AR)-positive human testicular medulla was shown to be a zone of mesenchymal to epithelial transition and a zone from which AR-positive cells appear to migrate into the human testicular cortex. While mouse Sertoli and Leydig cells have been proposed to arise from coelomic epithelium, Sertoli (SOX9) or Leydig (HSD3B1) cell markers are absent from the immediate coelomic zone of the developing human testis, perhaps because Leydig and Sertoli cell precursors are undifferentiated when they egress from the coelomic epithelium. The origin of mouse and human myoid cells remains unclear. This study provides a detailed comparison of the early stages of testicular development in human and mouse emphasizing differences in developmental processes.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
19
|
Li Y, Overland M, Derpinghaus A, Aksel S, Cao M, Ladwig N, Cunha GR, Baskin LS. Development of the human fetal testis: Morphology and expression of cellular differentiation markers. Differentiation 2023; 129:17-36. [PMID: 35490077 DOI: 10.1016/j.diff.2022.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 01/25/2023]
Abstract
A comprehensive immunohistochemical ontogeny of the developing human fetal testis has remained incomplete in the literature to date. We collected human fetal testes from 8 to 21 weeks of fetal age, as well as postnatal human testes at minipuberty, pre-pubertal, and pubertal stages. Immunohistochemistry was performed with a comprehensive panel of antigens targeting gonadocytes, Sertoli cells, fetal Leydig cells, peritubular myoid cells, and other hormonal and developmental targets. Testicular cords, precursor structures to seminiferous tubules, developed from 8 to 14 weeks of fetal age, separating the testis into the interstitial and intracordal compartments. Fetal gonadocytes were localized within the testicular cords and evaluated for Testis-Specific Protein Y, Octamer-binding transcription factor 4, Sal-like protein 4, and placental alkaline phosphatase expression. Fetal Sertoli cells were also localized in the testicular cords and evaluated for SRY-box Transcription Factor 9, inhibin, and anti-Mullerian hormone expression. Fetal Leydig cells were present in the interstitium and stained for cytochrome p450c17 and calretinin, while interstitial peritubular myoid cells were examined using smooth muscle α-actin staining. Androgen receptor expression was localized close to the testicular medulla at 8 weeks and then around the testicular cords in the interstitium as they matured in structure. Postnatal staining showed that Testis-Specific Protein Y remained positive of male gonadocytes throughout adulthood. Anti-Mullerian hormone, SRY-box Transcription Factor 9, and Steroidogenic factor 1 are expressed by the postnatal Sertoli cells at all ages examined. Leydig cell markers cytochrome p450c17 and calretinin are expressed during mini-puberty and puberty, but not expressed during the pre-pubertal period. Smooth muscle α-actin and androgen receptor were not expressed during mini-puberty or pre-puberty, but again expressed during the pubertal period. The ontogenic map of the human fetal and postnatal testicular structure and expression patterns described here will serve as a reference for future investigations into normal and abnormal testicular development.
Collapse
Affiliation(s)
- Yi Li
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Maya Overland
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Nicholas Ladwig
- Department of Pathology, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| |
Collapse
|
20
|
Aksel S, Cao M, Derpinghaus A, Baskin LS, Cunha GR. Ontogeny of mouse Sertoli, Leydig and peritubular myoid cells from embryonic day 10 to adulthood. Differentiation 2023; 129:96-108. [PMID: 35317954 DOI: 10.1016/j.diff.2022.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023]
Abstract
We present a comprehensive description of the differentiating somatic cell types (Sertoli, Leydig, and peritubular myoid cells) of the mouse testis from embryonic day 10.5 (E10.5) to adulthood, postnatal day 60 (P60). Immunohistochemistry was used to analyze expression of: Sox9 (a Sertoli cell marker), 3βHSD-1 (a fetal Leydig cell marker), 3βHSD-6 (an adult Leydig cell marker), α-actin (a peritubular myoid cell marker), and androgen receptor (a marker of all three somatic cell types). The temporal-spatial expression of these markers was used to interrogate findings of earlier experimental studies on the origin of Sertoli, Leydig and peritubular myoid cells, as well as extend previous descriptive studies across a broader developmental period (E10.5-P60). Such comparisons demonstrate inconsistencies that require further examination and raise questions regarding conservation of developmental mechanisms across higher vertebrate species.
Collapse
Affiliation(s)
- Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| |
Collapse
|
21
|
Human and mouse gonadal development. Differentiation 2023; 129:1-3. [PMID: 36272880 DOI: 10.1016/j.diff.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/28/2022] [Indexed: 01/25/2023]
|
22
|
Dissecting the fate of Foxl2-expressing cells in fetal ovary using lineage tracing and single-cell transcriptomics. Cell Discov 2022; 8:139. [PMID: 36575161 PMCID: PMC9794781 DOI: 10.1038/s41421-022-00492-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/25/2022] [Indexed: 12/28/2022] Open
Abstract
Gonad somatic cells acquire sex-specific fates during sex determination. In XX gonad, a subset of somatic cells expresses Foxl2 after sex determination which is considered the progenitor of granulosa cells. However, whether these cells also contribute to other cell types at later developmental stages is unknown. In the present study, the cell fate of Foxl2-expressing cells in fetal ovaries was analyzed by lineage tracing and single-cell transcriptomics. We found that Foxl2-expressing cells gave rise to three cell types at later developmental stages, including granulosa cells, theca-interstitial cells, and stromal cells. Series single-cell RNA sequencing revealed FOXL2-positive cells were divided into two clusters at P0. One group further differentiated into granulosa cells and Theca-G (Theca-interstitial cells derived from granulosa) at P14. Another group was classified as stromal cell lineage, then a small portion of them further differentiated into 3β-HSD-positive Theca-S (Theca-interstitial cells derived from stroma). Cyp17a1 was expressed in Theca-S, but not in Theca-G. This study demonstrated that Folx2-expressing cells in XX gonad after sex determination are multipotent and theca-interstitial cells are derived from different progenitors. Our data provided an important resource, at single-cell resolution, for a better understanding of somatic cell differentiation in ovary development.
Collapse
|
23
|
Zamfirescu AM, Yatsenko AS, Shcherbata HR. Notch signaling sculpts the stem cell niche. Front Cell Dev Biol 2022; 10:1027222. [PMID: 36605720 PMCID: PMC9810114 DOI: 10.3389/fcell.2022.1027222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Adult stem cells depend on their niches for regulatory signaling that controls their maintenance, division, and their progeny differentiation. While communication between various types of stem cells and their niches is becoming clearer, the process of stem cell niche establishment is still not very well understood. Model genetic organisms provide simplified systems to address various complex questions, for example, how is a stem cell niche formed? What signaling cascades induce the stem cell niche formation? Are the mechanisms of stem cell niche formation conserved? Notch signaling is an evolutionarily conserved pathway first identified in fruit flies, crucial in fate acquisition and spatiotemporal patterning. While the core logic behind its activity is fairly simple and requires direct cell-cell interaction, it reaches an astonishing complexity and versatility by combining its different modes of action. Subtleties such as equivalency between communicating cells, their physical distance, receptor and ligand processing, and endocytosis can have an effect on the way the events unfold, and this review explores some important general mechanisms of action, later on focusing on its involvement in stem cell niche formation. First, looking at invertebrates, we will examine how Notch signaling induces the formation of germline stem cell niche in male and female Drosophila. In the developing testis, a group of somatic gonadal precursor cells receive Delta signals from the gut, activating Notch signaling and sealing their fate as niche cells even before larval hatching. Meanwhile, the ovarian germline stem cell niche is built later during late larval stages and requires a two-step process that involves terminal filament formation and cap cell specification. Intriguingly, double security mechanisms of Notch signaling activation coordinated by the soma or the germline control both steps to ensure the robustness of niche assembly. Second, in the vast domains of mammalian cellular signaling, there is an emerging picture of Notch being an active player in a variety of tissues in health and disease. Notch involvement has been shown in stem cell niche establishment in multiple organs, including the brain, muscle, and intestine, where the stem cell niches are essential for the maintenance of adult stem cells. But adult stem cells are not the only cells looking for a home. Cancer stem cells use Notch signaling at specific stages to gain an advantage over endogenous tissue and overpower it, at the same time acquiring migratory and invasive abilities to claim new tissues (e.g., bone) as their territory. Moreover, in vitro models such as organoids reveal similar Notch employment when it comes to the developing stem cell niches. Therefore, a better understanding of the processes regulating stem cell niche assembly is key for the fields of stem cell biology and regenerative medicines.
Collapse
Affiliation(s)
| | | | - Halyna R. Shcherbata
- Mount Desert Island Biological Laboratory, Bar Harbor, ME, United States,*Correspondence: Halyna R. Shcherbata,
| |
Collapse
|
24
|
Fan M, Yang W, Zhang W, Zhang L. The ontogenic gonadal transcriptomes provide insights into sex change in the ricefield eel Monopterus albus. BMC ZOOL 2022; 7:56. [PMID: 37170354 PMCID: PMC10127409 DOI: 10.1186/s40850-022-00155-4] [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: 05/28/2022] [Accepted: 10/20/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The ricefield eel is a freshwater protogynous hermaphrodite fish and has become an important aquaculture species in China. The sex change of ricefield eel is impeding its aquaculture practice, particularly the large-scale artificial breeding. Many studies including transcriptomes of mixed gonadal samples from different individuals have been aimed to elucidate mechanisms underlying the sex change. However, the key physiological factors involved in the initiation of sex change remain to be identified. RESULTS: The present study performed transcriptomic analysis on gonadal samples of different sexual stages obtained through biopsy from the same fish undergoing sex change. A total of 539,764,816 high-quality reads were generated from twelve cDNA libraries of gonadal tissues at female (F), early intersexual (EI), mid-intersexual (MI), and late intersexual (LI) stages of three individual sex-changing fish. Pairwise comparisons between EI and F, MI and EI, and LI and MI identified 886, 319, and 10,767 differentially expressed genes (DEGs), respectively. Realtime quantitative PCR analysis of 12 representative DEGs showed similar expression profiles to those inferred from transcriptome data, suggesting the reliability of RNA-seq data for gene expression analysis. The expression of apoeb, csl2, and enpp2 was dramatically increased and peaked at EI while that of cyp19a1a, wnt4a, fgf16, and foxl2a significantly downregulated from F to EI and remained at very low levels during subsequent development until LI, which suggests that apoeb, csl2, enpp2, cyp19a1a, wnt4a, fgf16, and foxl2a may be closely associated with the initiation of sex change of ricefield eels. CONCLUSIONS Collectively, results of the present study confirmed that the down-regulation of female-related genes, such as cyp19a1a, wnt4a, fgf16, and foxl2a, is important for the sex change of ricefield eels. More importantly, some novel genes, including apoeb, csl2, and enpp2, were shown to be expressed with peak values at EI, which are potentially involved in the initiation of sex change. The present transcriptomic data may provide an important research resource for further unraveling the mechanisms underlying the sex change and testicular development in ricefield eels as well as other teleosts.
Collapse
Affiliation(s)
- Miao Fan
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wei Yang
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Present address: Institute of Biomedical Engineering, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China
| | - Weimin Zhang
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Biology Department, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Lihong Zhang
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
- Biology Department, School of Life Sciences, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
25
|
Inoue M, Baba T, Takahashi F, Terao M, Yanai S, Shima Y, Saito D, Sugihara K, Miura T, Takada S, Suyama M, Ohkawa Y, Morohashi KI. Tmsb10 triggers fetal Leydig differentiation by suppressing the RAS/ERK pathway. Commun Biol 2022; 5:974. [PMID: 36109592 PMCID: PMC9478096 DOI: 10.1038/s42003-022-03941-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
Leydig cells in fetal testes play crucial roles in masculinizing fetuses through androgen production. Gene knockout studies have revealed that growth factors are implicated in fetal Leydig cell (FLC) differentiation, but little is known about the mechanisms regulating this process. We investigate this issue by characterizing FLC progenitor cells using single-cell RNA sequencing. The sequence datasets suggest that thymosin β10 (Tmsb10) is transiently upregulated in the progenitors. While studying the function of Tmsb10, we reveal that platelet-derived growth factor (PDGF) regulates ciliogenesis through the RAS/ERK and PI3K/AKT pathways, and thereby promotes desert hedgehog (DHH)-dependent FLC differentiation. Tmsb10 expressed in the progenitor cells induces their differentiation into FLCs by suppressing the RAS/ERK pathway. Through characterizing the transiently expressed Tmsb10 in the FLC progenitors, this study unveils the molecular process of FLC differentiation and shows that it is cooperatively induced by DHH and PDGF. Investigation of fetal Leydig progenitors shows that thymosin β10 (Tmsb10) suppresses the RAS/ERK pathway, inducing progenitor differentiation into fetal Leydig cells.
Collapse
|
26
|
Abe SI. Behavior and Functional Roles of CD34 + Mesenchymal Cells in Mammalian Testes. Int J Mol Sci 2022; 23:9585. [PMID: 36076981 PMCID: PMC9455925 DOI: 10.3390/ijms23179585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/19/2022] Open
Abstract
Mammalian testes consist of seminiferous tubules within which Sertoli cells line up at the periphery and nurse germ cells, and of interstitia that harbor various cells such as peritubular myoid cells (PMCs), Leydig cells (LCs), vascular endothelial cells, immune cells such as macrophages, and mesenchymal (stromal) cells. Morphological studies have recently reported the presence of telocytes with telopodes in the interstitium of adult mouse, rat, and human testes. CD34+PDGFRα+ telocytes with long and moniliform telopodes form reticular networks with various cell types such as LCs, PMCs, and vessels, indicating their potential functions in cell-cell communications and tissue homeostasis. Functional studies have recently been performed on testicular interstitial cells and CD34+ cells, using 3D re-aggregate cultures of dissociated testicular cells, and cell cultures. Direct observation of CD34+ cells and adult LCs (ALCs) revealed that CD34+ cells extend thin cytoplasmic processes (telopodes), move toward the LC-CD34+ cell-re-aggregates, and finally enter into the re-aggregates, indicating the chemotactic behavior of CD34+ telocytes toward ALCs. In mammalian testes, important roles of mesenchymal interstitial cells as stem/progenitors in the differentiation and regeneration of LCs have been reported. Here, reports on testicular telocytes so far obtained are reviewed, and future perspectives on the studies of testicular telocytes are noted.
Collapse
Affiliation(s)
- Shin-Ichi Abe
- Faculty of Health Science, Kumamoto Health Science University, Kumamoto 861-5598, Japan
| |
Collapse
|
27
|
Ademi H, Djari C, Mayère C, Neirijnck Y, Sararols P, Rands CM, Stévant I, Conne B, Nef S. Deciphering the origins and fates of steroidogenic lineages in the mouse testis. Cell Rep 2022; 39:110935. [PMID: 35705036 DOI: 10.1016/j.celrep.2022.110935] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 03/22/2022] [Accepted: 05/19/2022] [Indexed: 11/03/2022] Open
Abstract
Leydig cells (LCs) are the major androgen-producing cells in the testis. They arise from steroidogenic progenitors (SPs), whose origins, maintenance, and differentiation dynamics remain largely unknown. Single-cell transcriptomics reveal that the mouse steroidogenic lineage is specified as early as embryonic day 12.5 (E12.5) and has a dual mesonephric and coelomic origin. SPs specifically express the Wnt5a gene and evolve rapidly. At E12.5 and E13.5, they give rise first to an intermediate population of pre-LCs, and finally to fetal LCs. At E16.5, SPs possess the characteristics of the dormant progenitors at the origin of adult LCs and are also transcriptionally closely related to peritubular myoid cells (PMCs). In agreement with our in silico analysis, in vivo lineage tracing indicates that Wnt5a-expressing cells are bona fide progenitors of PMCs as well as fetal and adult LCs, contributing to most of the LCs present in the fetal and adult testis.
Collapse
Affiliation(s)
- Herta Ademi
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Cyril Djari
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Chris M Rands
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland; Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | - Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Béatrice Conne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
28
|
Chen M, Cen C, Wang N, Shen Z, Wang M, Liu B, Li J, Cui X, Wang Y, Gao F. The functions of Wt1 in mouse gonad development and somatic cells differentiation. Biol Reprod 2022; 107:269-274. [PMID: 35244683 DOI: 10.1093/biolre/ioac050] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/27/2022] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Wilms' tumour 1 (Wt1) encodes a zinc finger nuclear transcription factor which is mutated in 15-20% of Wilms' tumor, a pediatric kidney tumor. Wt1 has been found to be involved in the development of many organs. In gonads, Wt1 is expressed in genital ridge somatic cells before sex determination, and its expression is maintained in Sertoli cells and granulosa cells after sex determination. It has been demonstrated that Wt1 is required for the survival of the genital ridge cells. Homozygous mutation of Wt1 causes gonad agenesis. Recent studies find that Wt1 plays important roles in lineage specification and maintenance of gonad somatic cells. In this review, we will summarize the recent research works about Wt1 in gonadal somatic cell differentiation.
Collapse
Affiliation(s)
- Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Changhuo Cen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhiming Shen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Mengyue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bowen Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jiayi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiuhong Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yanbo Wang
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, Inner Mongolia, 028000, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, P. R. China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
29
|
Estermann MA, Major AT, Smith CA. Genetic Regulation of Avian Testis Development. Genes (Basel) 2021; 12:1459. [PMID: 34573441 PMCID: PMC8470383 DOI: 10.3390/genes12091459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 11/30/2022] Open
Abstract
As in other vertebrates, avian testes are the site of spermatogenesis and androgen production. The paired testes of birds differentiate during embryogenesis, first marked by the development of pre-Sertoli cells in the gonadal primordium and their condensation into seminiferous cords. Germ cells become enclosed in these cords and enter mitotic arrest, while steroidogenic Leydig cells subsequently differentiate around the cords. This review describes our current understanding of avian testis development at the cell biology and genetic levels. Most of this knowledge has come from studies on the chicken embryo, though other species are increasingly being examined. In chicken, testis development is governed by the Z-chromosome-linked DMRT1 gene, which directly or indirectly activates the male factors, HEMGN, SOX9 and AMH. Recent single cell RNA-seq has defined cell lineage specification during chicken testis development, while comparative studies point to deep conservation of avian testis formation. Lastly, we identify areas of future research on the genetics of avian testis development.
Collapse
Affiliation(s)
| | | | - Craig Allen Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (M.A.E.); (A.T.M.)
| |
Collapse
|
30
|
Li H, Chang HM, Lin YM, Shi Z, Leung PCK. TGF-β1 inhibits microvascular-like formation by decreasing VCAM1 and ICAM1 via the upregulation of SNAIL in human granulosa cells. Mol Cell Endocrinol 2021; 535:111395. [PMID: 34265344 DOI: 10.1016/j.mce.2021.111395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 06/27/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
Three major endothelial cell junctional adhesion molecules (VCAM1, ICAM1 and E-SELECTIN) play important roles in the process of angiogenesis, a progression of extensive physiological vascularization that occurs during the formation of the corpus luteum. Our previous studies demonstrated that TGF-β1 is a negative regulator of luteinization and progesterone production in luteinized human granulosa (hGL) cells. Whether TGF-β1 can regulate the expression of these endothelial cell adhesion molecules and subsequent angiogenesis in hGL cells remains to be elucidated. Using dual inhibition approaches (small molecular inhibitors and siRNA-based knockdown), we provided the first data showing that TGF-β1 significantly upregulates the expression of the SNAIL transcription factor, which in turn suppresses the expression of VCAM1 and ICAM1 in hGL cells. Additionally, we demonstrate that the suppressive effects on the expression of VCAM1 and ICAM1 induced by TGF-β1 treatment were most likely via an ALK5-mediated SMAD-dependent signaling pathway. Furthermore, functional studies showed that hGL cells cultured on Matrigel exhibited two typical endothelial cell phenotypes, microvascular-like formation and a sprouting microvascular pattern. Notably, these phenotypes were significantly suppressed by either TGF-β1 treatment or knockdown of VCAM1 and ICAM1. Our findings suggest that TGF-β1 plays a potential role in the inhibition of granulosa cell angiogenesis by downregulating the expression of VCAM1 and ICAM1 during follicular development and corpus luteum formation.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Yung-Ming Lin
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Zhendan Shi
- Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada.
| |
Collapse
|
31
|
Scarlet D, Handschuh S, Reichart U, Podico G, Ellerbrock RE, Demyda-Peyrás S, Canisso IF, Walter I, Aurich C. Sexual Differentiation and Primordial Germ Cell Distribution in the Early Horse Fetus. Animals (Basel) 2021; 11:2422. [PMID: 34438878 PMCID: PMC8388682 DOI: 10.3390/ani11082422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
It was the aim of this study to characterize the development of the gonads and genital ducts in the equine fetus around the time of sexual differentiation. This included the identification and localization of the primordial germ cell population. Equine fetuses between 45 and 60 days of gestation were evaluated using a combination of micro-computed tomography scanning, immunohistochemistry, and multiplex immunofluorescence. Fetal gonads increased in size 23-fold from 45 to 60 days of gestation, and an even greater increase was observed in the metanephros volume. Signs of mesonephros atrophy were detected during this time. Tubular structures of the fetal testes were present from day 50 onwards, whereas cell clusters dominated in the fetal ovary. The genital ducts were well-differentiated and presented a lumen in all samples. No sign of mesonephric or paramesonephric duct degeneration was detected. Expression of AMH was strong in the fetal testes but absent in ovaries. Irrespective of sex, primordial germ cells selectively expressed LIN28. Migration of primordial germ cells from the mesonephros to the gonad was detected at 45 days, but not at 60 days of development. Their number and distribution within the gonad were influenced (p < 0.05) by fetal sex. Most primordial germ cells (86.8 ± 3.2% in females and 84.6 ± 4.7% in males) were characterized as pluripotent according to co-localization with CD117. However, only a very small percentage of primordial germ cells were proliferating (7.5 ± 1.7% in females and 3.2 ± 1.2% in males) based on co-localization with Ki67. It can be concluded that gonadal sexual differentiation in the horse occurs asynchronously with regard to sex but already before 45 days of gestation.
Collapse
Affiliation(s)
- Dragos Scarlet
- Obstetrics, Gynecology and Andrology, Department for Small Animals and Horses, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
- Institute of Veterinary Anatomy and Clinic of Reproductive Medicine, Vetsuisse Faculty Zürich, Winterthurerstrasse 260, 8057 Zürich, Switzerland
| | - Stephan Handschuh
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
| | - Ursula Reichart
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
| | - Giorgia Podico
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Robyn E. Ellerbrock
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Sebastián Demyda-Peyrás
- Department of Animal Production, School of Veterinary Sciences, National University of La Plata and CONICET CCT-La Plata, Calle 60 and 118 S/N, 1900 La Plata, Argentina;
| | - Igor F. Canisso
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Ingrid Walter
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Christine Aurich
- Center for Artificial Insemination and Embryo Transfer, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| |
Collapse
|
32
|
Estermann MA, Hirst CE, Major AT, Smith CA. The homeobox gene TGIF1 is required for chicken ovarian cortical development and generation of the juxtacortical medulla. Development 2021; 148:dev199646. [PMID: 34387307 PMCID: PMC8406534 DOI: 10.1242/dev.199646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
During early embryogenesis in amniotic vertebrates, the gonads differentiate into either ovaries or testes. The first cell lineage to differentiate gives rise to the supporting cells: Sertoli cells in males and pre-granulosa cells in females. These key cell types direct the differentiation of the other cell types in the gonad, including steroidogenic cells. The gonadal surface epithelium and the interstitial cell populations are less well studied, and little is known about their sexual differentiation programs. Here, we show the requirement of the homeobox transcription factor gene TGIF1 for ovarian development in the chicken embryo. TGIF1 is expressed in the two principal ovarian somatic cell populations: the cortex and the pre-granulosa cells of the medulla. TGIF1 expression is associated with an ovarian phenotype in estrogen-mediated sex reversal experiments. Targeted misexpression and gene knockdown indicate that TGIF1 is required, but not sufficient, for proper ovarian cortex formation. In addition, TGIF1 is identified as the first known regulator of juxtacortical medulla development. These findings provide new insights into chicken ovarian differentiation and development, specifically cortical and juxtacortical medulla formation.
Collapse
Affiliation(s)
| | | | | | - Craig Allen Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton VIC 3800, Australia
| |
Collapse
|
33
|
Kothandapani A, Larsen MC, Lee J, Jorgensen JS, Jefcoate CR. Distinctive functioning of STARD1 in the fetal Leydig cells compared to adult Leydig and adrenal cells. Impact of Hedgehog signaling via the primary cilium. Mol Cell Endocrinol 2021; 531:111265. [PMID: 33864885 DOI: 10.1016/j.mce.2021.111265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
STARD1 stimulates cholesterol transfer to mitochondrial CYP11A1 for conversion to pregnenolone. A cholesterol-binding START domain is guided by an N-terminal domain in a cell selective manner. Fetal and adult Leydig cells (FLC, ALC) show distinct Stard1 regulation. sm- FISH microscopy, which resolves individual molecules of Stard1 mRNA, shows uniformly high basal expression in each FLC. In ALC, in vivo, and cultured MA-10 cells, basal Stard1 expression is minimal. PKA activates loci asynchronously, with delayed splicing/export of 3.5 kb mRNA to mitochondria. After 60 min, ALC transition to an integrated mRNA delivery to mitochondria that is seen in FLC. Sertoli cells cooperate in Stard1 stimulation in FLC by delivering DHH to the primary cilium. There PTCH, SMO and cholesterol cooperate to release GLI3 to activate the Stard1 locus, probably by directing histone changes. ALC lack cilia. PKA then primes locus activation. FLC and ALC share similar SIK/CRTC/CREB regulation characterized for adrenal cells.
Collapse
Affiliation(s)
- Anbarasi Kothandapani
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
34
|
Shen YC, Shami AN, Moritz L, Larose H, Manske GL, Ma Q, Zheng X, Sukhwani M, Czerwinski M, Sultan C, Chen H, Gurczynski SJ, Spence JR, Orwig KE, Tallquist M, Li JZ, Hammoud SS. TCF21 + mesenchymal cells contribute to testis somatic cell development, homeostasis, and regeneration in mice. Nat Commun 2021; 12:3876. [PMID: 34162856 PMCID: PMC8222243 DOI: 10.1038/s41467-021-24130-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Testicular development and function rely on interactions between somatic cells and the germline, but similar to other organs, regenerative capacity declines in aging and disease. Whether the adult testis maintains a reserve progenitor population remains uncertain. Here, we characterize a recently identified mouse testis interstitial population expressing the transcription factor Tcf21. We found that TCF21lin cells are bipotential somatic progenitors present in fetal testis and ovary, maintain adult testis homeostasis during aging, and act as potential reserve somatic progenitors following injury. In vitro, TCF21lin cells are multipotent mesenchymal progenitors which form multiple somatic lineages including Leydig and myoid cells. Additionally, TCF21+ cells resemble resident fibroblast populations reported in other organs having roles in tissue homeostasis, fibrosis, and regeneration. Our findings reveal that the testis, like other organs, maintains multipotent mesenchymal progenitors that can be potentially leveraged in development of future therapies for hypoandrogenism and/or infertility.
Collapse
Affiliation(s)
- Yu-Chi Shen
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Lindsay Moritz
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Hailey Larose
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Gabriel L Manske
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Qianyi Ma
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Xianing Zheng
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Czerwinski
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Caleb Sultan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Haolin Chen
- Biochemistry and Molecular Biology, Bloomberg School of Public Health, John Hopkins, USA
| | | | - Jason R Spence
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michelle Tallquist
- University of Hawaii, Center for Cardiovascular Research, Honolulu, HI, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
35
|
Li SY, Gu X, Heinrich A, Hurley EG, Capel B, DeFalco T. Loss of Mafb and Maf distorts myeloid cell ratios and disrupts fetal mouse testis vascularization and organogenesis†. Biol Reprod 2021; 105:958-975. [PMID: 34007995 DOI: 10.1093/biolre/ioab098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
Testis differentiation is initiated when Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. Sertoli cells are essential for testis development, but cell types within the interstitial compartment, such as immune and endothelial cells, are also critical for organ formation. Our previous work implicated macrophages in fetal testis morphogenesis, but little is known about genes underlying immune cell development during organogenesis. Here we examine the role of the immune-associated genes Mafb and Maf in mouse fetal gonad development, and we demonstrate that deletion of these genes leads to aberrant hematopoiesis manifested by supernumerary gonadal monocytes. Mafb; Maf double knockout embryos underwent initial gonadal sex determination normally, but exhibited testicular hypervascularization, testis cord formation defects, Leydig cell deficit, and a reduced number of germ cells. In general, Mafb and Maf alone were dispensable for gonad development; however, when both genes were deleted, we observed significant defects in testicular morphogenesis, indicating that Mafb and Maf work redundantly during testis differentiation. These results demonstrate previously unappreciated roles for Mafb and Maf in immune and vascular development and highlight the importance of interstitial cells in gonadal differentiation.
Collapse
Affiliation(s)
- Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anna Heinrich
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Emily G Hurley
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA.,Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| |
Collapse
|
36
|
Gu X, Li SY, DeFalco T. Immune and vascular contributions to organogenesis of the testis and ovary. FEBS J 2021; 289:2386-2408. [PMID: 33774913 PMCID: PMC8476657 DOI: 10.1111/febs.15848] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/07/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Gonad development is a highly regulated process that coordinates cell specification and morphogenesis to produce sex-specific organ structures that are required for fertility, such as testicular seminiferous tubules and ovarian follicles. While sex determination occurs within specialized gonadal supporting cells, sexual differentiation is evident throughout the entire organ, including within the interstitial compartment, which contains immune cells and vasculature. While immune and vascular cells have been traditionally appreciated for their supporting roles during tissue growth and homeostasis, an increasing body of evidence supports the idea that these cell types are critical drivers of sexually dimorphic morphogenesis of the gonad. Myeloid immune cells, such as macrophages, are essential for multiple aspects of gonadogenesis and fertility, including for forming and maintaining gonadal vasculature in both sexes at varying stages of life. While vasculature is long known for supporting organ growth and serving as an export mechanism for gonadal sex steroids in utero, it is also an important component of fetal testicular morphogenesis and differentiation; additionally, it is vital for ovarian corpus luteal function and maintenance of pregnancy. These findings point toward a new paradigm in which immune cells and blood vessels are integral components of sexual differentiation and organogenesis. In this review, we discuss the state of the field regarding the diverse roles of immune and vascular cells during organogenesis of the testis and ovary and highlight outstanding questions in the field that could stimulate new research into these previously underappreciated constituents of the gonad.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
37
|
Oikonomakos I, Weerasinghe Arachchige LC, Schedl A. Developmental mechanisms of adrenal cortex formation and their links with adult progenitor populations. Mol Cell Endocrinol 2021; 524:111172. [PMID: 33484742 DOI: 10.1016/j.mce.2021.111172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
The adrenal cortex is the main steroid producing organ of the human body. Studies on adrenal tissue renewal have been neglected for many years, but recent intensified research has seen tremendous progress in our understanding of the formation and homeostasis of this organ. However, cell turnover of the adrenal cortex appears to be complex and several cell populations have been identified that can differentiate into steroidogenic cells and contribute to adrenal cortex renewal. The purpose of this review is to provide an overview of how the adrenal cortex develops and how stem cell populations relate to its developmental progenitors. Finally, we will summarize present and future approaches to harvest the potential of progenitor/stem cells for future cell replacement therapies.
Collapse
Affiliation(s)
- Ioannis Oikonomakos
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108, Nice, France.
| | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, 06108, Nice, France.
| |
Collapse
|
38
|
Estermann MA, Major AT, Smith CA. Gonadal Sex Differentiation: Supporting Versus Steroidogenic Cell Lineage Specification in Mammals and Birds. Front Cell Dev Biol 2020; 8:616387. [PMID: 33392204 PMCID: PMC7775416 DOI: 10.3389/fcell.2020.616387] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/07/2020] [Indexed: 01/16/2023] Open
Abstract
The gonads of vertebrate embryos are unique among organs because they have a developmental choice; ovary or testis formation. Given the importance of proper gonad formation for sexual development and reproduction, considerable research has been conducted over the years to elucidate the genetic and cellular mechanisms of gonad formation and sexual differentiation. While the molecular trigger for gonadal sex differentiation into ovary of testis can vary among vertebrates, from egg temperature to sex-chromosome linked master genes, the downstream molecular pathways are largely conserved. The cell biology of gonadal formation and differentiation has long thought to also be conserved. However, recent discoveries point to divergent mechanisms of gonad formation, at least among birds and mammals. In this mini-review, we provide an overview of cell lineage allocation during gonadal sex differentiation in the mouse model, focusing on the key supporting and steroidogenic cells and drawing on recent insights provided by single cell RNA-sequencing. We compare this data with emerging information in the chicken model. We highlight surprising differences in cell lineage specification between species and identify gaps in our current understanding of the cell biology underlying gonadogenesis.
Collapse
|
39
|
Nagahama Y, Chakraborty T, Paul-Prasanth B, Ohta K, Nakamura M. Sex determination, gonadal sex differentiation, and plasticity in vertebrate species. Physiol Rev 2020; 101:1237-1308. [PMID: 33180655 DOI: 10.1152/physrev.00044.2019] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A diverse array of sex determination (SD) mechanisms, encompassing environmental to genetic, have been found to exist among vertebrates, covering a spectrum from fixed SD mechanisms (mammals) to functional sex change in fishes (sequential hermaphroditic fishes). A major landmark in vertebrate SD was the discovery of the SRY gene in 1990. Since that time, many attempts to clone an SRY ortholog from nonmammalian vertebrates remained unsuccessful, until 2002, when DMY/dmrt1by was discovered as the SD gene of a small fish, medaka. Surprisingly, however, DMY/dmrt1by was found in only 2 species among more than 20 species of medaka, suggesting a large diversity of SD genes among vertebrates. Considerable progress has been made over the last 3 decades, such that it is now possible to formulate reasonable paradigms of how SD and gonadal sex differentiation may work in some model vertebrate species. This review outlines our current understanding of vertebrate SD and gonadal sex differentiation, with a focus on the molecular and cellular mechanisms involved. An impressive number of genes and factors have been discovered that play important roles in testicular and ovarian differentiation. An antagonism between the male and female pathway genes exists in gonads during both sex differentiation and, surprisingly, even as adults, suggesting that, in addition to sex-changing fishes, gonochoristic vertebrates including mice maintain some degree of gonadal sexual plasticity into adulthood. Importantly, a review of various SD mechanisms among vertebrates suggests that this is the ideal biological event that can make us understand the evolutionary conundrums underlying speciation and species diversity.
Collapse
Affiliation(s)
- Yoshitaka Nagahama
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Faculty of Biological Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Tapas Chakraborty
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan.,Karatsu Satellite of Aqua-Bioresource Innovation Center, Kyushu University, Karatsu, Japan
| | - Bindhu Paul-Prasanth
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidapeetham, Kochi, Kerala, India
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan
| | - Masaru Nakamura
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Research Center, Okinawa Churashima Foundation, Okinawa, Japan
| |
Collapse
|
40
|
Stévant I, Kühne F, Greenfield A, Chaboissier MC, Dermitzakis ET, Nef S. Dissecting Cell Lineage Specification and Sex Fate Determination in Gonadal Somatic Cells Using Single-Cell Transcriptomics. Cell Rep 2020; 26:3272-3283.e3. [PMID: 30893600 DOI: 10.1016/j.celrep.2019.02.069] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/07/2019] [Accepted: 02/19/2019] [Indexed: 01/02/2023] Open
Abstract
Sex determination is a unique process that allows the study of multipotent progenitors and their acquisition of sex-specific fates during differentiation of the gonad into a testis or an ovary. Using time series single-cell RNA sequencing (scRNA-seq) on ovarian Nr5a1-GFP+ somatic cells during sex determination, we identified a single population of early progenitors giving rise to both pre-granulosa cells and potential steroidogenic precursor cells. By comparing time series single-cell RNA sequencing of XX and XY somatic cells, we provide evidence that gonadal supporting cells are specified from these early progenitors by a non-sex-specific transcriptomic program before pre-granulosa and Sertoli cells acquire their sex-specific identity. In XX and XY steroidogenic precursors, similar transcriptomic profiles underlie the acquisition of cell fate but with XX cells exhibiting a relative delay. Our data provide an important resource, at single-cell resolution, for further interrogation of the molecular and cellular basis of mammalian sex determination.
Collapse
Affiliation(s)
- Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland; SIB, Swiss Institute of Bioinformatics, University of Geneva, 1211 Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | | | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland; SIB, Swiss Institute of Bioinformatics, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
41
|
Adaptation of Human Testicular Niche Cells for Pluripotent Stem Cell and Testis Development Research. Tissue Eng Regen Med 2020; 17:223-235. [PMID: 32114677 DOI: 10.1007/s13770-020-00240-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/02/2020] [Accepted: 01/16/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Human testicular cells are greatly valuable to the research community as tools for studying testicular physiology and the effects of environmental pollutants. Because adult testicular cells have a limited self-organization capacity and life span, we investigated whether human pluripotent stem cells (hPSCs) can be used together with testicular cells to move a step closer toward making an optimal model of the human testis. METHODS We used in vitro culture of donor testicular cells under serum-containing and chemically defined conditions. CRISPR-Cas9 technology was applied to introduce fluorescent transgenes (mCherry2 and EGFP) into hPSCs and testicular cells. hPSC-derived spheroids were co-cultured with human testicular cells in mini-spin bioreactors. RESULTS Traditional cell culture conditions used for maintenance of testicular somatic cells generally contain serum and pose limitations on evaluating the role of active molecules on cell functions. We established that chemically defined culture conditions can be used to maintain testicular cells without the loss of proliferative activity. These cultures demonstrate marker expression which is characteristic of common testicular cell types: Sertoli, Leydig, endothelial, myoid cells, and macrophages. In order to model testicular physiology, it is important to be able to perform live cell microscopy. Thus, we generated fluorescent protein-expressing human testicular cells and hPSCs and demonstrated that these cell types can be successfully co-cultured for prolonged periods of time in a three-dimensional microenvironment. CONCLUSION Our research extends the possible applications of human testis-derived somatic cells and shows that they can be used together with hPSCs for further studies of human male reproductive biology.
Collapse
|
42
|
Guo J, Nie X, Giebler M, Mlcochova H, Wang Y, Grow EJ, Kim R, Tharmalingam M, Matilionyte G, Lindskog C, Carrell DT, Mitchell RT, Goriely A, Hotaling JM, Cairns BR. The Dynamic Transcriptional Cell Atlas of Testis Development during Human Puberty. Cell Stem Cell 2020; 26:262-276.e4. [PMID: 31928944 PMCID: PMC7298616 DOI: 10.1016/j.stem.2019.12.005] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/03/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022]
Abstract
The human testis undergoes dramatic developmental and structural changes during puberty, including proliferation and maturation of somatic niche cells, and the onset of spermatogenesis. To characterize this understudied process, we profiled and analyzed single-cell transcriptomes of ∼10,000 testicular cells from four boys spanning puberty and compared them to those of infants and adults. During puberty, undifferentiated spermatogonia sequentially expand and differentiate prior to the initiation of gametogenesis. Notably, we identify a common pre-pubertal progenitor for Leydig and myoid cells and delineate candidate factors controlling pubertal differentiation. Furthermore, pre-pubertal Sertoli cells exhibit two distinct transcriptional states differing in metabolic profiles before converging to an alternative single mature population during puberty. Roles for testosterone in Sertoli cell maturation, antimicrobial peptide secretion, and spermatogonial differentiation are further highlighted through single-cell analysis of testosterone-suppressed transfemale testes. Taken together, our transcriptional atlas of the developing human testis provides multiple insights into developmental changes and key factors accompanying male puberty.
Collapse
Affiliation(s)
- Jingtao Guo
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; The Andrology Laboratory, Department of Surgery (Andrology/Urology), Center for Reconstructive Urology and Men's Health, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Xichen Nie
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Maria Giebler
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX39DS, UK
| | - Hana Mlcochova
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX39DS, UK
| | - Yueqi Wang
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Edward J Grow
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Robin Kim
- Section of Transplantation, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Melissa Tharmalingam
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; Royal Hospital for Children and Young People, Edinburgh EH91LF, UK
| | - Gabriele Matilionyte
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; Royal Hospital for Children and Young People, Edinburgh EH91LF, UK
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala 751 85, Sweden
| | - Douglas T Carrell
- The Andrology Laboratory, Department of Surgery (Andrology/Urology), Center for Reconstructive Urology and Men's Health, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; Royal Hospital for Children and Young People, Edinburgh EH91LF, UK
| | - Anne Goriely
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX39DS, UK
| | - James M Hotaling
- The Andrology Laboratory, Department of Surgery (Andrology/Urology), Center for Reconstructive Urology and Men's Health, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
| | - Bradley R Cairns
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
43
|
Chen P, Zirkin BR, Chen H. Stem Leydig Cells in the Adult Testis: Characterization, Regulation and Potential Applications. Endocr Rev 2020; 41:5610863. [PMID: 31673697 PMCID: PMC7753054 DOI: 10.1210/endrev/bnz013] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/25/2019] [Indexed: 01/20/2023]
Abstract
Androgen deficiency (hypogonadism) affects males of all ages. Testosterone replacement therapy (TRT) is effective in restoring serum testosterone and relieving symptoms. TRT, however, is reported to have possible adverse effects in part because administered testosterone is not produced in response to the hypothalamic-pituitary-gonadal (HPG) axis. Progress in stem cell biology offers potential alternatives for treating hypogonadism. Adult Leydig cells (ALCs) are generated by stem Leydig cells (SLCs) during puberty. SLCs persist in the adult testis. Considerable progress has been made in the identification, isolation, expansion and differentiation of SLCs in vitro. In addition to forming ALCs, SLCs are multipotent, with the ability to give rise to all 3 major cell lineages of typical mesenchymal stem cells, including osteoblasts, adipocytes, and chondrocytes. Several regulatory factors, including Desert hedgehog and platelet-derived growth factor, have been reported to play key roles in the proliferation and differentiation of SLCs into the Leydig lineage. In addition, stem cells from several nonsteroidogenic sources, including embryonic stem cells, induced pluripotent stem cells, mature fibroblasts, and mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord have been transdifferentiated into Leydig-like cells under a variety of induction protocols. ALCs generated from SLCs in vitro, as well as Leydig-like cells, have been successfully transplanted into ALC-depleted animals, restoring serum testosterone levels under HPG control. However, important questions remain, including: How long will the transplanted cells continue to function? Which induction protocol is safest and most effective? For translational purposes, more work is needed with primate cells, especially human.
Collapse
Affiliation(s)
- Panpan Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Barry R Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Haolin Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Anesthesiology, Perioperative Medicine, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
44
|
N-Cadherin Is Critical for the Survival of Germ Cells, the Formation of Steroidogenic Cells, and the Architecture of Developing Mouse Gonads. Cells 2019; 8:cells8121610. [PMID: 31835801 PMCID: PMC6952792 DOI: 10.3390/cells8121610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023] Open
Abstract
Normal gonad development assures the fertility of the individual. The properly functioning gonads must contain a sufficient number of the viable germ cells, possess a correct architecture and tissue structure, and assure the proper hormonal regulation. This is achieved by the interplay between the germ cells and different types of somatic cells. N-cadherin coded by the Cdh2 gene plays a critical role in this interplay. To gain an insight into the role of N-cadherin in the development of mouse gonads, we used the Cre-loxP system to knock out N-cadherin separately in two cell lines: the SF1+ somatic cells and the OCT4+ germ cells. We observed that N-cadherin plays a key role in the survival of both female and male germ cells. However, the N-cadherin is not necessary for the differentiation of the Sertoli cells or the initiation of the formation of testis cords or ovigerous cords. In the later stages of gonad development, N-cadherin is important for the maintenance of testis cord structure and is required for the formation of steroidogenic cells. In the ovaries, N-cadherin is necessary for the formation of the ovarian follicles. These results indicate that N-cadherin plays a major role in gonad differentiation, structuralization, and function.
Collapse
|
45
|
Grinspon RP, Rey RA. Molecular Characterization of XX Maleness. Int J Mol Sci 2019; 20:ijms20236089. [PMID: 31816857 PMCID: PMC6928850 DOI: 10.3390/ijms20236089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
Androgens and anti-Müllerian hormone (AMH), secreted by the foetal testis, are responsible for the development of male reproductive organs and the regression of female anlagen. Virilization of the reproductive tract in association with the absence of Müllerian derivatives in the XX foetus implies the existence of testicular tissue, which can occur in the presence or absence of SRY. Recent advancement in the knowledge of the opposing gene cascades driving to the differentiation of the gonadal ridge into testes or ovaries during early foetal development has provided insight into the molecular explanation of XX maleness.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| |
Collapse
|
46
|
Capel B. WOMEN IN REPRODUCTIVE SCIENCE: To be or not to be a testis. Reproduction 2019; 158:F101-F111. [PMID: 31265995 PMCID: PMC9945370 DOI: 10.1530/rep-19-0151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/25/2019] [Indexed: 11/08/2022]
Abstract
Work that established the testis as the driver of male development, and the Y chromosome as the bearer of the male-determining gene, established a working model, and set the stage for the molecular age of mammalian sex determination. The discovery and characterization of Sry/SRY at the top of the hierarchy in mammals launched the field in two major directions. The first was to identify the downstream transcription factors and other molecular players that drive the bifurcation of Sertoli and granulosa cell differentiation. The second major direction was to understand organogenesis of the early bipotential gonad, and how divergence of its two distinct morphogenetic pathways (testis and ovary) is regulated at the cellular level. This review will summarize the early discoveries soon after Sry was identified and focus on my study of the gonad as a model of organogenesis.
Collapse
Affiliation(s)
- Blanche Capel
- 1Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
47
|
Bhattacharya I, Sen Sharma S, Majumdar SS. Pubertal orchestration of hormones and testis in primates. Mol Reprod Dev 2019; 86:1505-1530. [DOI: 10.1002/mrd.23246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology & BiotechnologyHNB Garhwal University, Srinagar CampusSrinagar India
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Souvik Sen Sharma
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Subeer S. Majumdar
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
- Gene and Protein Engineering LabNational Institute of Animal BiotechnologyHyderabad India
| |
Collapse
|
48
|
Deciphering Cell Lineage Specification during Male Sex Determination with Single-Cell RNA Sequencing. Cell Rep 2019; 22:1589-1599. [PMID: 29425512 DOI: 10.1016/j.celrep.2018.01.043] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/21/2017] [Accepted: 01/12/2018] [Indexed: 11/20/2022] Open
Abstract
The gonad is a unique biological system for studying cell-fate decisions. However, major questions remain regarding the identity of somatic progenitor cells and the transcriptional events driving cell differentiation. Using time-series single-cell RNA sequencing on XY mouse gonads during sex determination, we identified a single population of somatic progenitor cells prior to sex determination. A subset of these progenitors differentiates into Sertoli cells, a process characterized by a highly dynamic genetic program consisting of sequential waves of gene expression. Another subset of multipotent cells maintains their progenitor state but undergoes significant transcriptional changes restricting their competence toward a steroidogenic fate required for the differentiation of fetal Leydig cells. Our findings confirm the presence of a unique multipotent progenitor population in the gonadal primordium that gives rise to both supporting and interstitial lineages. These also provide the most granular analysis of the transcriptional events occurring during testicular cell-fate commitment.
Collapse
|
49
|
Stévant I, Nef S. Genetic Control of Gonadal Sex Determination and Development. Trends Genet 2019; 35:346-358. [PMID: 30902461 DOI: 10.1016/j.tig.2019.02.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/15/2019] [Accepted: 02/23/2019] [Indexed: 10/27/2022]
Abstract
Sex determination is the process by which the bipotential gonads develop as either testes or ovaries. With two distinct potential outcomes, the gonadal primordium offers a unique model for the study of cell fate specification and how distinct cell populations diverge from multipotent progenitors. This review focuses on recent advances in our understanding of the genetic programs and epigenetic mechanisms that regulate gonadal sex determination and the regulation of cell fate commitment in the bipotential gonads. We rely primarily on mouse data to illuminate the complex and dynamic genetic programs controlling cell fate decision and sex-specific cell differentiation during gonadal formation and gonadal sex determination.
Collapse
Affiliation(s)
- Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland; SIB, Swiss Institute of Bioinformatics, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
50
|
Piprek RP, Kloc M, Kubiak JZ. Matrix metalloproteinase-dependent regulation of extracellular matrix shapes the structure of sexually differentiating mouse gonads. Differentiation 2019; 106:23-34. [PMID: 30852470 DOI: 10.1016/j.diff.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 11/19/2022]
Abstract
The extracellular matrix (ECM) proteins play an important role in the establishment of the sex-dependent structure of developing gonads. The matrix metalloproteinases (MMPs) are the major players in the regulation of ECM. Our hypothesis was that the MMPs-dependent regulation of EMC is crucial for the establishment of the correct, either testis or ovary, structure of developing gonad. We cultured developing mouse gonads in vitro in the presence of the MMPs inhibitors (α-2-macroglobulin, leupeptin, phosphoramidon) or the MMPs activator, APMA (4-aminophenylmercuric acetate). These inhibitors and activator inhibit/activate, to a different degree, matrix metalloproteinases, but the exact mechanism of inhibition/activation remains unknown. We found that the MMP inhibitors increased accumulation of ECM in the developing gonads. The α-2-macroglobulin had the weakest, and the phosphoramidon the strongest effect on the ECM and the structure of the gonads. The α-2-macroglobulin caused a slight increase of ECM and did not disrupt the gonad structure. Leupeptin led to the strong accumulation of ECM, resulted in the formation of the structures resembling testis cords in both testes and ovaries, and caused increase of apoptosis and complete loss of germ cells. Phosphoramidon caused the strongest accumulation of ECM, which separated individual cells and completely prevented intercellular adhesion both in the testes and in the ovaries. As a result of aberrant morphology, the sex of the phosphoramidon-treated gonads was morphologically unrecognizable. The APMA - the activator of MMP caused ECM loss, which led to the loss of cell adhesion, cell dispersion and an aberrant morphology of the gonads. These results indicate that the ECM accumulation is MMPs-dependent and that the correct amount and distribution of ECM during gonad development plays a key role in the formation of the gonad structure.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jacek Z Kubiak
- Univ Rennes, CNRS, Institute of Genetics and Development of Rennes, UMR 6290, Cell Cycle Group, Faculty of Medicine, F-35000, Rennes, France; Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| |
Collapse
|