1
|
Wijayarathna R, de Geus ED, Genovese R, Gearing LJ, Wray-McCann G, Sreenivasan R, Hasan H, Fijak M, Stanton P, Fietz D, Pilatz A, Schuppe HC, Tate MD, Hertzog PJ, Hedger MP. Interferon epsilon is produced in the testis and protects the male reproductive tract against virus infection, inflammation and damage. PLoS Pathog 2024; 20:e1012702. [PMID: 39621805 PMCID: PMC11637430 DOI: 10.1371/journal.ppat.1012702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/12/2024] [Accepted: 10/29/2024] [Indexed: 12/14/2024] Open
Abstract
The testis is a reservoir for viruses that can cause persistent infection and adversely affect male reproductive health, an observation commonly attributed to deficiencies in inducible antiviral defence mechanisms. In this study, we demonstrate that interferon-epsilon (IFNε), a type I interferon initially discovered in female reproductive epithelia, is constitutively expressed by meiotic and post-meiotic spermatogenic cells, Leydig cells and macrophages in mouse testes. A similar distribution pattern was observed in human testes. Mice lacking IFNɛ were more susceptible to Zika virus-induced inflammation and damage of the testis and epididymis compared to wild-type mice. Exogenous IFNε treatment reduced the viral infection burden in cultured human testicular cells by inducing interferon-stimulated gene expression, and reducing inflammatory gene expression and cell damage. Treatment was more effective when administered prior to infection. These data indicate a critical role for constitutively-expressed IFNɛ in limiting viral infection and inflammatory damage in the male reproductive tract.
Collapse
Affiliation(s)
- Rukmali Wijayarathna
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Eveline D. de Geus
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Australia
| | - Rosemary Genovese
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, Australia
| | - Linden J. Gearing
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Australia
| | - Georgie Wray-McCann
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Australia
| | - Rajini Sreenivasan
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, Australia
| | - Hiba Hasan
- Institute of Anatomy and Cell Biology, Justus-Liebig University, Giessen, Germany
| | - Monika Fijak
- Institute of Anatomy and Cell Biology, Justus-Liebig University, Giessen, Germany
| | - Peter Stanton
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, Australia
| | - Daniela Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Adrian Pilatz
- Department of Urology, Paediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Paediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Michelle D. Tate
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Australia
| | - Paul J. Hertzog
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Australia
| | - Mark P. Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
2
|
Yoshimura S, Omotehara T, Nakata H, Birch LA, Prins GS, Ichimura K, Itoh M. Mesonephric tubules expressing estrogen and androgen receptors remain in the rete ovarii of adult mice. Cell Tissue Res 2024:10.1007/s00441-024-03931-5. [PMID: 39579219 DOI: 10.1007/s00441-024-03931-5] [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: 01/17/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024]
Abstract
The rete ovarii and epoophoron in females are homologous structures of the rete testis and efferent/epididymal duct in males and are derived from the developing rete cells and mesonephric tubules, respectively. Sex steroid hormones play a critical role in reproductive function for both sexes, and we recently reported expression patterns of sex steroid receptors in developing male reproductive tracts. However, their expression patterns in females remain unclear. We, therefore, investigated the three-dimensional structure and expression patterns of sex steroid receptors in the rete ovarii and epoophoron of fetal and adult female mice. In adult females, the epoophoron was not adherent to the rete ovarii. The rete ovarii had a bursa-like structure, with its extra-ovarian region protruding toward the epoophoron. A marker for mesonephric tubules, PAX2 (Paired box 2), was detected in the epoophoron and a small population of epithelial cells in the extra-ovarian rete ovarii. These epithelial cells expressed estrogen receptor and androgen receptor. During development, mesonephric tubules were adherent to the rete ovarii at first, but as the development proceeded, the continuity was lost due to the interruption of the tubule rather than separation between the tip of the tubule and rete ovarii. These findings suggest that epithelial cells, originating from the mesonephric tubules, persist even in the adult rete ovarii with maintained expressions of receptors for estrogen and androgen.
Collapse
Affiliation(s)
- Shiori Yoshimura
- Department of Anatomy, Tokyo Medical University, Shinjuku-Ku, Tokyo, Japan
| | - Takuya Omotehara
- Department of Anatomy, Tokyo Medical University, Shinjuku-Ku, Tokyo, Japan.
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Hiroki Nakata
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Ishikawa, Japan
| | - Lynn A Birch
- Department of Urology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Gail S Prins
- Department of Urology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Shinjuku-Ku, Tokyo, Japan
| |
Collapse
|
3
|
Anbarci DN, McKey J, Levic DS, Bagnat M, Capel B. Rediscovering the Rete Ovarii: a secreting auxiliary structure to the ovary. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.08.566085. [PMID: 37986754 PMCID: PMC10659334 DOI: 10.1101/2023.11.08.566085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The rete ovarii (RO) is an appendage of the ovary that has been given little attention. Although the RO appears in drawings of the ovary in early versions of Grays Anatomy, it disappeared from recent textbooks, and is often dismissed as a functionless vestige in the adult ovary. Using PAX8 immunostaining and confocal microscopy, we characterized the fetal development of the RO in the context of the ovary. The RO consists of three distinct regions that persist in adult life, the intraovarian rete (IOR), the extraovarian rete (EOR), and the connecting rete (CR). While the cells of the IOR appear to form solid cords within the ovary, the EOR rapidly develops into a convoluted tubular epithelium ending in a distal dilated tip. Cells of the EOR are ciliated and exhibit cellular trafficking capabilities. The CR, connecting the EOR to the IOR, gradually acquires tubular epithelial characteristics by birth. Using microinjections into the distal dilated tip of the EOR, we found that luminal contents flow towards the ovary. Mass spectrometry revealed that the EOR lumen contains secreted proteins potentially important for ovarian function. We show that the cells of the EOR are closely associated with vasculature and macrophages, and are contacted by neuronal projections, consistent with a role as a sensory appendage of the ovary. The direct proximity of the RO to the ovary and its integration with the extraovarian landscape suggest that it plays an important role in ovary development and homeostasis.
Collapse
|
4
|
Fan L, Liu B, Yao R, Gao X, Wang H, Jiang S, Zheng X, Chen H, Hou H, Liu Y, Hu Q. Nicotine-induced transcriptional changes and mitochondrial dysfunction in the ventral tegmental area revealed by single-nucleus transcriptomics. J Genet Genomics 2024; 51:1237-1251. [PMID: 39244085 DOI: 10.1016/j.jgg.2024.08.009] [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: 06/03/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Nicotine is widely recognized as the primary contributor to tobacco dependence. Previous studies have indicated that molecular and behavioral responses to nicotine are primarily mediated by ventral tegmental area (VTA) neurons, and accumulating evidence suggests that glia play prominent roles in nicotine addiction. However, VTA neurons and glia have yet to be characterized at the transcriptional level during the progression of nicotine self-administration. Here, a male mouse model of nicotine self-administration is established and the timing of three critical phases (pre-addiction, addicting, and post-addiction phase) is characterized. Single-nucleus RNA sequencing in the VTA at each phase is performed to comprehensively classify specific cell subtypes. Adaptive changes occurred during the addicting and post-addiction phases, with the addicting phase displaying highly dynamic neuroplasticity that profoundly impacts the transcription in each cell subtype. Furthermore, significant transcriptional changes in energy metabolism-related genes are observed, accompanied by notable structural alterations in neuronal mitochondria during the progression of nicotine self-administration. The results provide insights into mechanisms underlying the progression of nicotine addiction, serving as an important resource for identifying potential molecular targets for nicotine cessation.
Collapse
Affiliation(s)
- Lei Fan
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230000, China; University of Science and Technology of China, Hefei, Anhui 230000, China; Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China
| | - Boxin Liu
- BGI Genomics, BGI-Shenzhen, Guangdong 518018, China
| | - Ru Yao
- BGI Genomics, BGI-Shenzhen, Guangdong 518018, China
| | - Xia Gao
- Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China
| | - Hongjuan Wang
- Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China
| | - Sanjie Jiang
- BGI Genomics, BGI-Shenzhen, Guangdong 518018, China
| | | | - Huan Chen
- Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China.
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China.
| | - Yong Liu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230000, China; University of Science and Technology of China, Hefei, Anhui 230000, China.
| | - Qingyuan Hu
- Beijing Life Science Academy, Beijing 100000, China; Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing 100000, China; China National Tobacco Quality Supervision & Test Center, Zhengzhou, Henan 450000, China; Key Laboratory of Tobacco Biological Effects, Zhengzhou, Henan 450000, China.
| |
Collapse
|
5
|
Li MY, Lu M, Cao DM, Han Q, Ma XH, Wei CC, Zhang WJ. Characterization of Ucp1-iCre knockin mice reveals the recombination activity in male germ cells. Am J Physiol Endocrinol Metab 2024; 327:E544-E551. [PMID: 39230395 DOI: 10.1152/ajpendo.00128.2024] [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: 04/01/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024]
Abstract
Ucp1 promoter-driven Cre transgenic mice are useful in the manipulation of gene expression specifically in thermogenic adipose tissues. However, the wildly used Ucp1-Cre line was generated by random insertion into the genome and showed ectopic activity in some tissues beyond adipose tissues. Here, we characterized a knockin mouse line Ucp1-iCre generated by targeting IRES-Cre cassette immediately downstream the stop codon of the Ucp1 gene. The Cre insertion had little to no effect on uncoupling protein 1 (UCP1) levels in brown adipose tissue. Ucp1-iCre mice of both genders exhibited normal thermogenesis and cold tolerance. When crossed with Rosa-tdTomato reporter mice, Ucp1-iCre mice showed robust Cre activity in thermogenic adipose tissues. In addition, limited Cre activity was sparsely present in the ventromedial hypothalamus (VMH), choroid plexus, kidney, adrenal glands, ovary, and testis in Ucp1-iCre mice, albeit to a much lesser extent and with reduced intensity compared with the conventional Ucp1-Cre line. Single-cell transcriptome analysis revealed Ucp1 mRNA expression in male spermatocytes. Moreover, male Ucp1-iCre mice displayed a high frequency of Cre-mediated recombination in the germline, whereas no such effect was observed in female Ucp1-iCre mice. These findings suggest that Ucp1-iCre mice offer promising utility in the context of conditional gene manipulation in thermogenic adipose tissues, while also highlighting the need for caution in mouse mating and genotyping procedures.NEW & NOTEWORTHY Ucp1 promoter-driven Cre transgenic mice are useful in the manipulation of gene expression specifically in thermogenic adipose tissues. The widely used Ucp1-Cre mouse line (Ucp1-CreEvdr), which was generated using the bacterial artificial chromosome (BAC) strategy, exhibits major brown and white fat transcriptomic dysregulation and ectopic activity beyond adipose tissues. Here, we comprehensively validate Ucp1-iCre knockin mice, which serve as another optional model besides Ucp1-CreEvdr mice for specific genetic manipulation in thermogenic tissue.
Collapse
Affiliation(s)
- Meng-Yue Li
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Ming Lu
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Dong-Mei Cao
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Qing Han
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Xian-Hua Ma
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Chun-Chun Wei
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| | - Weiping J Zhang
- National Key Laboratory of Immunity and Inflammation, Department of Pathophysiology, Obesity and Diabetes Center, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Kulibin AY, Malolina EA. Thyroid Hormone Regulates Postnatal Development of the Rete Testis in Mice. Endocrinology 2024; 165:bqae125. [PMID: 39279445 DOI: 10.1210/endocr/bqae125] [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: 03/27/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
Thyroid hormone regulates the rate of testis maturation in mammals. Manipulations of thyroid hormone levels in neonatal animals affect various aspects of testis biology. However, there have been no studies examining the effects of thyroid hormone on the rete testis (RT). Here, we used animal models of neonatal hyperthyroidism (injections of triiodothyronine, or T3) and hypothyroidism (goitrogen 6-propyl-2-thiouracil [PTU] treatment) and found that higher levels of thyroid hormone accelerate RT development, while lower levels of thyroid hormone delay it. T3 and PTU treatments influence RT size, proliferation of RT cells, and expression of DMRT1 and androgen receptor in the RT. T3 supplementation accelerates RT development in an organ testicular culture, which indicates the local action of thyroid hormone. Additionally, it was found that follicle-stimulating hormone could be involved in the regulation both of RT proliferation and RT size. The fact that RT cells in a cell culture do not respond to T3 suggests indirect action of thyroid hormone on the RT in vivo or the loss of the responsiveness to the hormone in vitro.
Collapse
Affiliation(s)
- Andrey Yu Kulibin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow 119334, Russian Federation
| | - Ekaterina A Malolina
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow 119334, Russian Federation
| |
Collapse
|
7
|
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
|
8
|
Ridnik M, Abberbock E, Alipov V, Lhermann SZ, Kaufman S, Lubman M, Poulat F, Gonen N. Two redundant transcription factor binding sites in a single enhancer are essential for mammalian sex determination. Nucleic Acids Res 2024; 52:5514-5528. [PMID: 38499491 PMCID: PMC11162780 DOI: 10.1093/nar/gkae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/25/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024] Open
Abstract
Male development in mammals depends on the activity of the two SOX gene: Sry and Sox9, in the embryonic testis. As deletion of Enhancer 13 (Enh13) of the Sox9 gene results in XY male-to-female sex reversal, we explored the critical elements necessary for its function and hence, for testis and male development. Here, we demonstrate that while microdeletions of individual transcription factor binding sites (TFBS) in Enh13 lead to normal testicular development, combined microdeletions of just two SRY/SOX binding motifs can alone fully abolish Enh13 activity leading to XY male-to-female sex reversal. This suggests that for proper male development to occur, these few nucleotides of non-coding DNA must be intact. Interestingly, we show that depending on the nature of these TFBS mutations, dramatically different phenotypic outcomes can occur, providing a molecular explanation for the distinct clinical outcomes observed in patients harboring different variants in the same enhancer.
Collapse
Affiliation(s)
- Meshi Ridnik
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Elisheva Abberbock
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Veronica Alipov
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Shelly Ziv Lhermann
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Shoham Kaufman
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Maor Lubman
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Francis Poulat
- Group “Development and Pathology of the Gonad”. Department of Genetics, Cell Biology and Development, Institute of Human Genetics, CNRS-University of Montpellier UMR9002, Montpellier, France
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
9
|
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
|
10
|
Li J, Zhang X, Wang X, Wang Z, Li X, Zheng J, Li J, Xu G, Sun C, Yi G, Yang N. Single-nucleus transcriptional and chromatin accessible profiles reveal critical cell types and molecular architecture underlying chicken sex determination. J Adv Res 2024:S2090-1232(24)00185-1. [PMID: 38734369 DOI: 10.1016/j.jare.2024.05.007] [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: 09/09/2023] [Revised: 01/23/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
INTRODUCTION Understanding the sex determination mechanisms in birds has great significance for the biological sciences and production in the poultry industry. Sex determination in chickens is a complex process that involves fate decisions of supporting cells such as granulosa or Sertoli cells. However, a systematic understanding of the genetic regulation and cell commitment process underlying sex determination in chickens is still lacking. OBJECTIVES We aimed to dissect the molecular characteristics associated with sex determination in the gonads of chicken embryos. METHODS Single-nucleus RNA-seq (snRNA-seq) and ATAC-seq (snATAC-seq) analysis were conducted on the gonads of female and male chickens at embryonic day 3.5 (E3.5), E4.5, and E5.5. RESULTS Here, we provided a time-course transcriptional and chromatin accessible profiling of gonads during chicken sex determination at single-cell resolution. We uncovered differences in cell composition and developmental trajectories between female and male gonads and found that the divergence of transcription and accessibility in gonadal cells first emerged at E5.5. Furthermore, we revealed key cell-type-specific transcription factors (TFs) and regulatory networks that drive lineage commitment. Sex determination signaling pathways, dominated by BMP signaling, are preferentially activated in males during gonadal development. Further pseudotime analysis of the supporting cells indicated that granulosa cells were regulated mainly by the TEAD gene family and that Sertoli cells were driven by the DMRT1 regulons. Cross-species analysis suggested high conservation of both cell types and cell-lineage-specific TFs across the six vertebrates. CONCLUSIONS Overall, our study will contribute to accelerating the development of sex manipulation technology in the poultry industry and the application of chickens as a unique model for studying cell fate decisions.
Collapse
Affiliation(s)
- Jianbo Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Xiuan Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Xiqiong Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Zhen Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xingzheng Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Jiangxia Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Junying Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Guiyun Xu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Congjiao Sun
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China.
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China.
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
11
|
Migale R, Neumann M, Mitter R, Rafiee MR, Wood S, Olsen J, Lovell-Badge R. FOXL2 interaction with different binding partners regulates the dynamics of ovarian development. SCIENCE ADVANCES 2024; 10:eadl0788. [PMID: 38517962 PMCID: PMC10959415 DOI: 10.1126/sciadv.adl0788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
The transcription factor FOXL2 is required in ovarian somatic cells for female fertility. Differential timing of Foxl2 deletion, in embryonic versus adult mouse ovary, leads to distinctive outcomes, suggesting different roles across development. Here, we comprehensively investigated FOXL2's role through a multi-omics approach to characterize gene expression dynamics and chromatin accessibility changes, coupled with genome-wide identification of FOXL2 targets and on-chromatin interacting partners in somatic cells across ovarian development. We found that FOXL2 regulates more targets postnatally, through interaction with factors regulating primordial follicle formation and steroidogenesis. Deletion of one interactor, ubiquitin-specific protease 7 (Usp7), results in impairment of somatic cell differentiation, germ cell nest breakdown, and ovarian development, leading to sterility. Our datasets constitute a comprehensive resource for exploration of the molecular mechanisms of ovarian development and causes of female infertility.
Collapse
Affiliation(s)
- Roberta Migale
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Michelle Neumann
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Richard Mitter
- Bioinformatics core, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mahmoud-Reza Rafiee
- RNA Networks Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sophie Wood
- Genetic Modification Service, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jessica Olsen
- Genetic Modification Service, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
12
|
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: 0] [Impact Index Per Article: 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
|
13
|
Stévant I, Gonen N, Poulat F. Transposable elements acquire time- and sex-specific transcriptional and epigenetic signatures along mouse fetal gonad development. Front Cell Dev Biol 2024; 11:1327410. [PMID: 38283992 PMCID: PMC10811072 DOI: 10.3389/fcell.2023.1327410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/30/2024] Open
Abstract
Gonadal sex determination in mice is a complex and dynamic process, which is crucial for the development of functional reproductive organs. The expression of genes involved in this process is regulated by a variety of genetic and epigenetic mechanisms. Recently, there has been increasing evidence that transposable elements (TEs), which are a class of mobile genetic elements, play a significant role in regulating gene expression during embryogenesis and organ development. In this study, we aimed to investigate the involvement of TEs in the regulation of gene expression during mouse embryonic gonadal development. Through bioinformatics analysis, we aimed to identify and characterize specific TEs that operate as regulatory elements for sex-specific genes, as well as their potential mechanisms of regulation. We identified TE loci expressed in a time- and sex-specific manner along fetal gonad development that correlate positively and negatively with nearby gene expression, suggesting that their expression is integrated to the gonadal regulatory network. Moreover, chromatin accessibility and histone post-transcriptional modification analyses in differentiating supporting cells revealed that TEs are acquiring a sex-specific signature for promoter-, enhancer-, and silencer-like elements, with some of them being proximal to critical sex-determining genes. Altogether, our study introduces TEs as the new potential players in the gene regulatory network that controls gonadal development in mammals.
Collapse
Affiliation(s)
- Isabelle Stévant
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, Montpellier, France
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, Montpellier, France
| |
Collapse
|
14
|
Souali-Crespo S, Condrea D, Vernet N, Féret B, Klopfenstein M, Grandgirard E, Alunni V, Cerciat M, Jung M, Mayere C, Nef S, Mark M, Chalmel F, Ghyselinck NB. Loss of NR5A1 in mouse Sertoli cells after sex determination changes cellular identity and induces cell death by anoikis. Development 2023; 150:dev201710. [PMID: 38078651 PMCID: PMC10753587 DOI: 10.1242/dev.201710] [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: 02/17/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023]
Abstract
To investigate the role of the nuclear receptor NR5A1 in the testis after sex determination, we analyzed mice lacking NR5A1 in Sertoli cells (SCs) from embryonic day (E) 13.5 onwards. Ablation of Nr5a1 impaired the expression of genes characteristic of SC identity (e.g. Sox9 and Amh), caused SC death from E14.5 onwards through a Trp53-independent mechanism related to anoikis, and induced disorganization of the testis cords. Together, these effects caused germ cells to enter meiosis and die. Single-cell RNA-sequencing experiments revealed that NR5A1-deficient SCs changed their molecular identity: some acquired a 'pre-granulosa-like' cell identity, whereas other reverted to a 'supporting progenitor-like' cell identity, most of them being 'intersex' because they expressed both testicular and ovarian genes. Fetal Leydig cells (LCs) did not display significant changes, indicating that SCs are not required beyond E14.5 for their emergence or maintenance. In contrast, adult LCs were absent from postnatal testes. In addition, adult mutant males displayed persistence of Müllerian duct derivatives, decreased anogenital distance and reduced penis length, which could be explained by the loss of AMH and testosterone synthesis due to SC failure.
Collapse
Affiliation(s)
- Sirine Souali-Crespo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Diana Condrea
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Nadège Vernet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Betty Féret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Muriel Klopfenstein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Erwan Grandgirard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Imaging Center, IGBMC, F-67404 Illkirch Cedex, France
| | - Violaine Alunni
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- GenomEast Platform, France Génomique consortium, IGBMC, 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Marie Cerciat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- GenomEast Platform, France Génomique consortium, IGBMC, 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Matthieu Jung
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- GenomEast Platform, France Génomique consortium, IGBMC, 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Chloé Mayere
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland
| | - Manuel Mark
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), F-67000 Strasbourg, France
| | - Frédéric Chalmel
- Univ Rennes, EHESP, Inserm, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Norbert B. Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| |
Collapse
|
15
|
Chen B, Pei D. Genetic clues to reprogramming power and formation of mouse oocyte. Curr Opin Genet Dev 2023; 83:102110. [PMID: 37722148 DOI: 10.1016/j.gde.2023.102110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/05/2023] [Accepted: 07/29/2023] [Indexed: 09/20/2023]
Abstract
Oocyte features the unique capacity to reprogram not only sperm but also somatic nuclei to totipotency, yet the scarcity of oocytes has hindered the exploration and application of their reprogramming ability. In the meanwhile, the formation of oocytes, which involves extensive intracellular alterations and interactions, has also attracted tremendous interest. This review discusses developmental principles and regulatory mechanisms associated with ooplasm reprogramming and oocyte formation from a genetic perspective, with knowledge derived from mouse models. We also discuss future directions, especially to address the lack of insight into the regulatory networks that shape the identity of female germ cells or drive transitions in their developmental programs.
Collapse
|
16
|
Malolina EA, Galiakberova AA, Mun VV, Sabirov MS, Dashinimaev EB, Kulibin AY. A comparative analysis of genes differentially expressed between rete testis cells and Sertoli cells of the mouse testis. Sci Rep 2023; 13:20896. [PMID: 38017073 PMCID: PMC10684643 DOI: 10.1038/s41598-023-48149-7] [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: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023] Open
Abstract
The rete testis (RT) is a region of the mammalian testis that plays an important role in testicular physiology. The RT epithelium consists of cells sharing some well-known gene markers with supporting Sertoli cells (SCs). However, little is known about the differences in gene expression between these two cell populations. Here, we used fluorescence-activated cell sorting (FACS) to obtain pure cultures of neonatal RT cells and SCs and identified differentially expressed genes (DEGs) between these cell types using RNA sequencing (RNA-seq). We then compared our data with the RNA-seq data of other studies that examined RT cells and SCs of mice of different ages and generated a list of DEGs permanently upregulated in RT cells throughout testis development and in culture, which included 86 genes, and a list of 79 DEGs permanently upregulated in SCs. The analysis of studies on DMRT1 function revealed that nearly half of the permanent DEGs could be regulated by this SC upregulated transcription factor. We suggest that useful cell lineage markers and candidate genes for the specification of both RT cells and SCs may be present among these permanent DEGs.
Collapse
Affiliation(s)
- Ekaterina A Malolina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia.
| | - Adelya A Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Valery V Mun
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Marat S Sabirov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Erdem B Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Andrey Yu Kulibin
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| |
Collapse
|
17
|
Anbarci DN, O’Rourke R, Xiang Y, Peters DT, Capel B, McKey J. Transcriptome analysis of the mouse fetal and adult rete ovarii and surrounding tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.06.565717. [PMID: 37986846 PMCID: PMC10659311 DOI: 10.1101/2023.11.06.565717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The rete ovarii (RO) is an epithelial structure that arises during fetal development in close proximity to the ovary and persists throughout adulthood in mice. However, the functional significance of the RO remains elusive, and it has been absent from recent discussions of female reproductive anatomy. The RO comprises three distinct regions: the intraovarian rete (IOR) within the ovary, the extraovarian rete (EOR) in the periovarian tissue, and the connecting rete (CR) linking the EOR and IOR. We hypothesize that the RO plays a pivotal role in maintaining ovarian homeostasis and responding to physiological changes. To uncover the nature and function of RO cells, we conducted transcriptome analysis, encompassing bulk, single-cell, and nucleus-level sequencing of both fetal and adult RO tissues using the Pax8-rtTA; Tre-H2B-GFP mouse line, where all RO regions express nuclear GFP. This study presents three datasets, which highlight RO-specific gene expression signatures and reveal differences in gene expression across the three RO regions during development and in adulthood. The integration and rigorous validation of these datasets will advance our understanding of the RO's roles in ovarian development, female maturation, and adult female fertility.
Collapse
Affiliation(s)
- Dilara N. Anbarci
- Department of Cell Biology, Duke University Medical Center, Durham NC 27710
| | - Rebecca O’Rourke
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO 80045
| | - Yu Xiang
- Department of Cell Biology, Duke University Medical Center, Durham NC 27710
| | - Derek T. Peters
- Department of Cell Biology, Duke University Medical Center, Durham NC 27710
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham NC 27710
| | - Jennifer McKey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora CO 80045
| |
Collapse
|
18
|
Gregoire EP, De Cian MC, Migale R, Perea-Gomez A, Schaub S, Bellido-Carreras N, Stévant I, Mayère C, Neirijnck Y, Loubat A, Rivaud P, Sopena ML, Lachambre S, Linssen MM, Hohenstein P, Lovell-Badge R, Nef S, Chalmel F, Schedl A, Chaboissier MC. The -KTS splice variant of WT1 is essential for ovarian determination in mice. Science 2023; 382:600-606. [PMID: 37917714 PMCID: PMC7615308 DOI: 10.1126/science.add8831] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/29/2023] [Indexed: 11/04/2023]
Abstract
Sex determination in mammals depends on the differentiation of the supporting lineage of the gonads into Sertoli or pregranulosa cells that govern testis and ovary development, respectively. Although the Y-linked testis-determining gene Sry has been identified, the ovarian-determining factor remains unknown. In this study, we identified -KTS, a major, alternatively spliced isoform of the Wilms tumor suppressor WT1, as a key determinant of female sex determination. Loss of -KTS variants blocked gonadal differentiation in mice, whereas increased expression, as found in Frasier syndrome, induced precocious differentiation of ovaries independently of their genetic sex. In XY embryos, this antagonized Sry expression, resulting in male-to-female sex reversal. Our results identify -KTS as an ovarian-determining factor and demonstrate that its time of activation is critical in gonadal sex differentiation.
Collapse
Affiliation(s)
- Elodie P Gregoire
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Marie-Cécile De Cian
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Roberta Migale
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Aitana Perea-Gomez
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Sébastien Schaub
- Sorbonne Université, CNRS, Development Biology Laboratory (LBDV), 06234 Villefranche sur Mer, France
| | | | - 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
| | - 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
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Agnès Loubat
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Paul Rivaud
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, 35000 Rennes, France
| | | | - Simon Lachambre
- Infinity, Inserm, CNRS, University Toulouse III, 31024 Toulouse, France
| | - Margot M. Linssen
- Central Animal and Transgenic Facility and Dept. Human Genetics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Peter Hohenstein
- Central Animal and Transgenic Facility and Dept. Human Genetics, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | | | - 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
| | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, 35000 Rennes, France
| | - Andreas Schedl
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | | |
Collapse
|
19
|
Dujardin E, André M, Dewaele A, Mandon-Pépin B, Poulat F, Frambourg A, Thépot D, Jouneau L, Jolivet G, Pailhoux E, Pannetier M. DMRT1 is a testis-determining gene in rabbits and is also essential for female fertility. eLife 2023; 12:RP89284. [PMID: 37847154 PMCID: PMC10581690 DOI: 10.7554/elife.89284] [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: 10/18/2023] Open
Abstract
DMRT1 is the testis-determining factor in several species of vertebrates, but its involvement in mammalian testes differentiation, where SRY is the testis-determining gene, remains ambiguous. So far, DMRT1 loss-of-function has been described in two mammalian species and induces different phenotypes: Disorders of Sex Development (46, XY DSD) in men and male infertility in mice. We thus abolished DMRT1 expression by CRISPR/Cas9 in a third species of mammal, the rabbit. First, we observed that gonads from XY DMRT1-/- rabbit fetuses differentiated like ovaries, highlighting that DMRT1 is involved in testis determination. In addition to SRY, DMRT1 is required in the supporting cells to increase the expression of the SOX9 gene, which heads the testicular genetic cascade. Second, we highlighted another function of DMRT1 in the germline since XX and XY DMRT1-/- ovaries did not undergo meiosis and folliculogenesis. XX DMRT1-/- adult females were sterile, showing that DMRT1 is also crucial for female fertility. To conclude, these phenotypes indicate an evolutionary continuum between non-mammalian vertebrates such as birds and non-rodent mammals. Furthermore, our data support the potential involvement of DMRT1 mutations in different human pathologies, such as 46, XY DSD as well as male and female infertility.
Collapse
Affiliation(s)
- Emilie Dujardin
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Marjolaine André
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Aurélie Dewaele
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Béatrice Mandon-Pépin
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier; 34396MontpellierFrance
| | - Anne Frambourg
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Dominique Thépot
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Geneviève Jolivet
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Eric Pailhoux
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| | - Maëlle Pannetier
- Université Paris-Saclay, UVSQ, INRAE, BREED; 78350Jouy-en-JosasFrance
- École Nationale Vétérinaire d'Alfort, BREED; 94700Maisons-AlfortFrance
| |
Collapse
|
20
|
Whiley PAF, Luu MCM, O’Donnell L, Handelsman DJ, Loveland KL. Testis exposure to unopposed/elevated activin A in utero affects somatic and germ cells and alters steroid levels mimicking phthalate exposure. Front Endocrinol (Lausanne) 2023; 14:1234712. [PMID: 37727456 PMCID: PMC10505732 DOI: 10.3389/fendo.2023.1234712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/26/2023] [Indexed: 09/21/2023] Open
Abstract
Correct fetal testis development underpins adult male fertility, and TGFβ superfamily ligands control key aspects of this process. Transcripts encoding one such ligand, activin A, are upregulated in testes after sex determination and remain high until after birth. Testis development requires activin signalling; mice lacking activin A (Inhba KO) display altered somatic and germ cell proliferation, disrupted cord elongation and altered steroid synthesis. In human pregnancies with pre-eclampsia, the foetus is inappropriately exposed to elevated activin A. To learn how this affects testis development, we examined mice lacking the potent activin inhibitor, inhibin, (Inha KO) at E13.5, E15.5 and PND0. At E13.5, testes appeared similar in WT and KO littermates, however E15.5 Inha KO testes displayed two germline phenotypes: (1) multinucleated germ cells within cords, and (2) germ cells outside of cords, both of which are documented following in utero exposure to endocrine disrupting phthalates in rodents. Quantitation of Sertoli and germ cells in Inha KO (modelling elevated activin A) and Inhba KO (low activin A) testes using immunofluorescence demonstrated activin A bioactivity determines the Sertoli/germ cell ratio. The 50% reduction in gonocytes in Inha KO testes at birth indicates unopposed activin A has a profound impact on embryonic germ cells. Whole testis RNAseq on Inha KO mice revealed most transcripts affected at E13.5 were present in Leydig cells and associated with steroid biosynthesis/metabolism. In agreement, androstenedione (A4), testosterone (T), and the A4:T ratio were reduced in Inha KO testes at E17.5, confirming unopposed activin A disrupts testicular steroid production. E15.5 testes cultured with either activin A and/or mono-2-ethylhexyl phthalate (MEHP) generated common histological and transcriptional outcomes affecting germline and Leydig cells, recapitulating the phenotype observed in Inha KO testes. Cultures with activin A and MEHP together provided evidence of common targets. Lastly, this study extends previous work focussed on the Inhba KO model to produce a signature of activin A bioactivity in the fetal testis. These outcomes show the potential for elevated activin A signalling to replicate some aspects of fetal phthalate exposure prior to the masculinization programming window, influencing fetal testis growth and increasing the risk of testicular dysgenesis.
Collapse
Affiliation(s)
- Penny A. F. Whiley
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Michael C. M. Luu
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Liza O’Donnell
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | | | - Kate L. Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
Hala D. The use of in silico extreme pathway (ExPa) analysis to identify conserved reproductive transcriptional-regulatory networks in humans, mice, and zebrafish. Syst Biol Reprod Med 2023; 69:271-287. [PMID: 37023256 PMCID: PMC10461611 DOI: 10.1080/19396368.2023.2188996] [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: 01/03/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 04/08/2023]
Abstract
Vertebrate sex determination and differentiation are coordinated by the activations and maintenance of reproductive transcriptional-regulatory networks (TRNs). There is considerable interest in studying the conserved design principles and functions of reproductive TRNs given that their intricate regulation is susceptible to disruption by gene mutations or exposures to exogenous endocrine disrupting chemicals (or EDCs). In this manuscript, the Boolean rules describing reproductive TRNs in humans, mice, and zebrafish, were represented as a pseudo-stoichiometric matrix model. This model mathematically described the interactions of 35 transcription factors with 21 sex determination and differentiation genes across the three species. The in silico approach of Extreme Pathway (ExPa) analysis was used to predict the extent of TRN gene activations subject to the species-specific transcriptomics data, from across various developmental life-stages. A goal of this work was to identify conserved and functional reproductive TRNs across the three species. ExPa analyses predicted the sex differentiation genes, DHH, DMRT1, and AR, to be highly active in male humans, mice, and zebrafish. Whereas FOXL2 was the most active gene in female humans and mice; and CYP19A1A in female zebrafish. These results agree with the expectation that regardless of a lack of sex determination genes in zebrafish, the TRNs responsible for canalizing male vs. female sexual differentiation are conserved with mammalian taxa. ExPa analysis therefore provides a framework with which to study the TRNs that influence the development of sexual phenotypes. And the in silico predicted conservation of sex differentiation TRNs between mammals and zebrafish identifies the piscine species as an effective in vivo model to study mammalian reproductive systems under normal or perturbed pathologies.
Collapse
Affiliation(s)
- David Hala
- Department of Marine Biology, Texas A&M University at Galveston, TX, USA
| |
Collapse
|
22
|
Okashita N, Maeda R, Tachibana M. CDYL reinforces male gonadal sex determination through epigenetically repressing Wnt4 transcription in mice. Proc Natl Acad Sci U S A 2023; 120:e2221499120. [PMID: 37155872 PMCID: PMC10193937 DOI: 10.1073/pnas.2221499120] [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: 12/19/2022] [Accepted: 04/01/2023] [Indexed: 05/10/2023] Open
Abstract
In mammals, male and female gonads initially develop from bipotential progenitor cells, which can differentiate into either testicular or ovarian cells. The decision to adopt a testicular or ovarian fate relies on robust genetic forces, i.e., activation of the testis-determining gene Sry, as well as a delicate balance of expression levels for pro-testis and pro-ovary factors. Recently, epigenetic regulation has been found to be a key element in activation of Sry. Nevertheless, the mechanism by which epigenetic regulation controls the expression balance of pro-testis and pro-ovary factors remains unclear. Chromodomain Y-like protein (CDYL) is a reader protein for repressive histone H3 methylation marks. We found that a subpopulation of Cdyl-deficient mice exhibited XY sex reversal. Gene expression analysis revealed that the testis-promoting gene Sox9 was downregulated in XY Cdyl-deficient gonads during the sex determination period without affecting Sry expression. Instead, we found that the ovary-promoting gene Wnt4 was derepressed in XY Cdyl-deficient gonads prior to and during the sex-determination period. Wnt4 heterozygous deficiency restored SOX9 expression in Cdyl-deficient XY gonads, indicating that derepressed Wnt4 is a cause of the repression of Sox9. We found that CDYL directly bound to the Wnt4 promoter and maintained its H3K27me3 levels during the sex-determination period. These findings indicate that CDYL reinforces male gonadal sex determination by repressing the ovary-promoting pathway in mice.
Collapse
Affiliation(s)
- Naoki Okashita
- Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Ryo Maeda
- Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| | - Makoto Tachibana
- Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
23
|
Zhang X, Li J, Chen S, Yang N, Zheng J. Overview of Avian Sex Reversal. Int J Mol Sci 2023; 24:ijms24098284. [PMID: 37175998 PMCID: PMC10179413 DOI: 10.3390/ijms24098284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Sex determination and differentiation are processes by which a bipotential gonad adopts either a testicular or ovarian cell fate, and secondary sexual characteristics adopt either male or female developmental patterns. In birds, although genetic factors control the sex determination program, sex differentiation is sensitive to hormones, which can induce sex reversal when disturbed. Although these sex-reversed birds can form phenotypes opposite to their genotypes, none can experience complete sex reversal or produce offspring under natural conditions. Promising evidence indicates that the incomplete sex reversal is associated with cell autonomous sex identity (CASI) of avian cells, which is controlled by genetic factors. However, studies cannot clearly describe the regulatory mechanism of avian CASI and sex development at present, and these factors require further exploration. In spite of this, the abundant findings of avian sex research have provided theoretical bases for the progress of gender control technologies, which are being improved through interdisciplinary co-operation and will ultimately be employed in poultry production. In this review, we provide an overview of avian sex determination and differentiation and comprehensively summarize the research progress on sex reversal in birds, especially chickens. Importantly, we describe key issues faced by applying gender control systems in poultry production and chronologically summarize the development of avian sex control methods. In conclusion, this review provides unique perspectives for avian sex studies and helps scientists develop more advanced systems for sex regulation in birds.
Collapse
Affiliation(s)
- Xiuan Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Jianbo Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Sirui Chen
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Jiangxia Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| |
Collapse
|
24
|
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
|
25
|
Gonen N, Eozenou C, Mitter R, Elzaiat M, Stévant I, Aviram R, Bernardo AS, Chervova A, Wankanit S, Frachon E, Commère PH, Brailly-Tabard S, Valon L, Barrio Cano L, Levayer R, Mazen I, Gobaa S, Smith JC, McElreavey K, Lovell-Badge R, Bashamboo A. In vitro cellular reprogramming to model gonad development and its disorders. SCIENCE ADVANCES 2023; 9:eabn9793. [PMID: 36598988 PMCID: PMC9812383 DOI: 10.1126/sciadv.abn9793] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 12/02/2022] [Indexed: 05/28/2023]
Abstract
During embryonic development, mutually antagonistic signaling cascades determine gonadal fate toward a testicular or ovarian identity. Errors in this process result in disorders of sex development (DSDs), characterized by discordance between chromosomal, gonadal, and anatomical sex. The absence of an appropriate, accessible in vitro system is a major obstacle in understanding mechanisms of sex-determination/DSDs. Here, we describe protocols for differentiation of mouse and human pluripotent cells toward gonadal progenitors. Transcriptomic analysis reveals that the in vitro-derived murine gonadal cells are equivalent to embryonic day 11.5 in vivo progenitors. Using similar conditions, Sertoli-like cells derived from 46,XY human induced pluripotent stem cells (hiPSCs) exhibit sustained expression of testis-specific genes, secrete anti-Müllerian hormone, migrate, and form tubular structures. Cells derived from 46,XY DSD female hiPSCs, carrying an NR5A1 variant, show aberrant gene expression and absence of tubule formation. CRISPR-Cas9-mediated variant correction rescued the phenotype. This is a robust tool to understand mechanisms of sex determination and model DSDs.
Collapse
Affiliation(s)
- Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caroline Eozenou
- Institut Pasteur, Université de Paris, CNRS UMR3738, Human Developmental Genetics, F-75015 Paris, France
| | - Richard Mitter
- Bioinformatics Core, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maëva Elzaiat
- Institut Pasteur, Université de Paris, CNRS UMR3738, Human Developmental Genetics, F-75015 Paris, France
| | - Isabelle Stévant
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Rona Aviram
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Andreia Sofia Bernardo
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Almira Chervova
- Department of Stem Cell and Developmental Biology, Institut Pasteur, Paris 75724, France
| | - Somboon Wankanit
- Institut Pasteur, Université de Paris, CNRS UMR3738, Human Developmental Genetics, F-75015 Paris, France
| | - Emmanuel Frachon
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, F-75015 Paris, France
| | - Pierre-Henri Commère
- Cytometry and Biomarkers, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F-75015 Paris, France
| | - Sylvie Brailly-Tabard
- Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Molecular Genetics, Pharmacogenetics, and Hormonology, Le Kremlin-Bicêtre, France
| | - Léo Valon
- Institut Pasteur, Université de Paris, CNRS UMR3738, Cell Death and Epithelial Homeostasis, F-75015 Paris, France
| | - Laura Barrio Cano
- Cytometry and Biomarkers, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, F-75015 Paris, France
| | - Romain Levayer
- Institut Pasteur, Université de Paris, CNRS UMR3738, Cell Death and Epithelial Homeostasis, F-75015 Paris, France
| | - Inas Mazen
- Genetics Department, National Research Center, Cairo, Egypt
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, F-75015 Paris, France
| | - James C. Smith
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kenneth McElreavey
- Institut Pasteur, Université de Paris, CNRS UMR3738, Human Developmental Genetics, F-75015 Paris, France
| | | | - Anu Bashamboo
- Institut Pasteur, Université de Paris, CNRS UMR3738, Human Developmental Genetics, F-75015 Paris, France
| |
Collapse
|
26
|
Suzuki T. Overview of single-cell RNA sequencing analysis and its application to spermatogenesis research. Reprod Med Biol 2023; 22:e12502. [PMID: 36726594 PMCID: PMC9884325 DOI: 10.1002/rmb2.12502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/18/2022] [Accepted: 01/10/2023] [Indexed: 01/30/2023] Open
Abstract
Background Single-cell transcriptomics allows parallel analysis of multiple cell types in tissues. Because testes comprise somatic cells and germ cells at various stages of spermatogenesis, single-cell RNA sequencing is a powerful tool for investigating the complex process of spermatogenesis. However, single-cell RNA sequencing analysis needs extensive knowledge of experimental technologies and bioinformatics, making it difficult for many, particularly experimental biologists and clinicians, to use it. Methods Aiming to make single-cell RNA sequencing analysis familiar, this review article presents an overview of experimental and computational methods for single-cell RNA sequencing analysis with a history of transcriptomics. In addition, combining the PubMed search and manual curation, this review also provides a summary of recent novel insights into human and mouse spermatogenesis obtained using single-cell RNA sequencing analyses. Main Findings Single-cell RNA sequencing identified mesenchymal cells and type II innate lymphoid cells as novel testicular cell types in the adult mouse testes, as well as detailed subtypes of germ cells. This review outlines recent discoveries into germ cell development and subtypes, somatic cell development, and cell-cell interactions. Conclusion The findings on spermatogenesis obtained using single-cell RNA sequencing may contribute to a deeper understanding of spermatogenesis and provide new directions for male fertility therapy.
Collapse
Affiliation(s)
- Takahiro Suzuki
- RIKEN Center for Integrated Medical Science (IMS)Yokohama CityKanagawaJapan
- Graduate School of Medical Life ScienceYokohama City UniversityYokohama CityKanagawaJapan
| |
Collapse
|
27
|
Uchida A, Imaimatsu K, Suzuki H, Han X, Ushioda H, Uemura M, Imura-Kishi K, Hiramatsu R, Takase HM, Hirate Y, Ogura A, Kanai-Azuma M, Kudo A, Kanai Y. SOX17-positive rete testis epithelium is required for Sertoli valve formation and normal spermiogenesis in the male mouse. Nat Commun 2022; 13:7860. [PMID: 36543770 PMCID: PMC9772346 DOI: 10.1038/s41467-022-35465-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Seminiferous tubules (STs) in the mammalian testes are connected to the rete testis (RT) via a Sertoli valve (SV). Spermatozoa produced in the STs are released into the tubular luminal fluid and passively transported through the SV into the RT. However, the physiological functions of the RT and SV remain unclear. Here, we identified the expression of Sox17 in RT epithelia. The SV valve was disrupted before puberty in RT-specific Sox17 conditional knockout (Sox17-cKO) male mice. This induced a backflow of RT fluid into the STs, which caused aberrant detachment of immature spermatids. RT of Sox17-cKO mice had reduced expression levels of various growth factor genes, which presumably support SV formation. When transplanted next to the Sox17+ RT, Sertoli cells of Sox17-cKO mice reconstructed the SV and supported proper spermiogenesis in the STs. This study highlights the novel and unexpected modulatory roles of the RT in SV valve formation and spermatogenesis in mouse testes, as a downstream action of Sox17.
Collapse
Affiliation(s)
- Aya Uchida
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan ,grid.7597.c0000000094465255Bioresource Engineering Division, RIKEN BioResouce Research Center, Tsukuba, Ibaraki Japan
| | - Kenya Imaimatsu
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Honoka Suzuki
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Xiao Han
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Hiroki Ushioda
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Mami Uemura
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Kasane Imura-Kishi
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Ryuji Hiramatsu
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| | - Hinako M. Takase
- grid.265073.50000 0001 1014 9130Center for Experimental Animals, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo Japan
| | - Yoshikazu Hirate
- grid.265073.50000 0001 1014 9130Center for Experimental Animals, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo Japan
| | - Atsuo Ogura
- grid.7597.c0000000094465255Bioresource Engineering Division, RIKEN BioResouce Research Center, Tsukuba, Ibaraki Japan
| | - Masami Kanai-Azuma
- grid.265073.50000 0001 1014 9130Center for Experimental Animals, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo Japan
| | - Akihiko Kudo
- grid.411205.30000 0000 9340 2869Department of Microscopic Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo Japan
| | - Yoshiakira Kanai
- grid.26999.3d0000 0001 2151 536XDepartment of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo Japan
| |
Collapse
|
28
|
Lorenzi V, Vento‐Tormo R, Garcia‐Alonso L. Human gonadal development, cell by cell. Clin Transl Med 2022; 12:e1123. [PMID: 36513876 PMCID: PMC9747682 DOI: 10.1002/ctm2.1123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Valentina Lorenzi
- Cellular GeneticsWellcome Sanger InstituteHinxtonCambridgeshireUK
- European Molecular Biology LaboratoryEuropean Bioinformatics Institute (EMBL‐EBI)HinxtonCambridgeshireUK
| | | | | |
Collapse
|
29
|
Reconstitution of reproductive organ system that produces functional oocytes. Curr Opin Genet Dev 2022; 77:101982. [PMID: 36179583 DOI: 10.1016/j.gde.2022.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 01/27/2023]
Abstract
Reproductive organs have unique developmental and functional properties that enable them to manage both germ cell development and the endocrine system in a sex-dependent manner. Proper reconstitution of the reproductive organs, therefore, will contribute to a deeper understanding of the mechanisms underlying germ cell development and sex-determination. However, reproductive organs have not yet been systematically reconstituted from pluripotent stem cells. This is largely due to technical problems in the reconstitution of the germ cell and somatic cell lineages, which have very different developmental trajectories. Accordingly, faithful construction of reproductive organoids requires that the reconstitution and evaluation of these two different cell lineages be performed separately. Here, we update the state-of-the-art in the reconstitution of reproductive organoids that produce functional oocytes.
Collapse
|
30
|
Estermann MA, Smith CA. Fadrozole-mediated sex reversal in the embryonic chicken gonad involves a PAX2 positive undifferentiated supporting cell state. Front Cell Dev Biol 2022; 10:1042759. [PMID: 36438569 PMCID: PMC9684329 DOI: 10.3389/fcell.2022.1042759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/28/2022] [Indexed: 09/08/2024] Open
Abstract
Gonadal sex differentiation among vertebrates involves divergent fates of a common group of progenitor cells present in both presumptive ovaries and testes. The first cell type to differentiate gives rise to pre-Sertoli cells in the testis, and pre-follicular cells in the ovary. These cells derive from a common lineage of so-called "supporting cells". In birds and other egg-laying vertebrates, locally synthesised estrogen has a central role in ovarian development and influences the fate of these supporting cells. Manipulation of estrogen levels during embryonic development induces gonadal sex reversal, providing an experimental setting to evaluate the process of gonadal sex differentiation. Recently, we identified PAX2 as a novel marker of the undifferentiated supporting cell lineage in the chicken embryo, expressed in both sexes prior to overt gonadal sex differentiation. PAX2 expression is downregulated at the onset of gonadal sex differentiation in both males and females. The analysis of this undifferentiated supporting cell marker, together with Sertoli (male) and pre-granulosa (female) will enhance our understanding of supporting cell differentiation. Here we characterized the supporting cells differentiation process and identified undifferentiated supporting cells in estrogen-mediated sex reversal experiments. Female embryos treated with the aromatase inhibitor fadrozole developed into ovotestis, containing pre-granulosa cells, Sertoli cells and PAX2 positive undifferentiated supporting cells. In contrast, male embryos treated with 17β-estradiol showed no PAX2+ undifferentiated gonadal supporting cells. Fadrozole time-course as well as multiple dose analysis suggests that supporting cell transdifferentiation involves a dedifferentiation event into a PAX2+ undifferentiated supporting cell state, followed by a redifferentiation towards the opposite sex lineage.
Collapse
Affiliation(s)
| | - Craig A. Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Imaimatsu K, Uchida A, Hiramatsu R, Kanai Y. Gonadal Sex Differentiation and Ovarian Organogenesis along the Cortical-Medullary Axis in Mammals. Int J Mol Sci 2022; 23:13373. [PMID: 36362161 PMCID: PMC9655463 DOI: 10.3390/ijms232113373] [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] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 09/20/2023] Open
Abstract
In most mammals, the sex of the gonads is based on the fate of the supporting cell lineages, which arises from the proliferation of coelomic epithelium (CE) that surfaces on the bipotential genital ridge in both XY and XX embryos. Recent genetic studies and single-cell transcriptome analyses in mice have revealed the cellular and molecular events in the two-wave proliferation of the CE that produce the supporting cells. This proliferation contributes to the formation of the primary sex cords in the medullary region of both the testis and the ovary at the early phase of gonadal sex differentiation, as well as to that of the secondary sex cords in the cortical region of the ovary at the perinatal stage. To support gametogenesis, the testis forms seminiferous tubules in the medullary region, whereas the ovary forms follicles mainly in the cortical region. The medullary region in the ovary exhibits morphological and functional diversity among mammalian species that ranges from ovary-like to testis-like characteristics. This review focuses on the mechanism of gonadal sex differentiation along the cortical-medullary axis and compares the features of the cortical and medullary regions of the ovary in mammalian species.
Collapse
Affiliation(s)
- Kenya Imaimatsu
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Aya Uchida
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
- RIKEN BioResouce Research Center, Tsukuba 305-0074, Japan
| | - Ryuji Hiramatsu
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| |
Collapse
|
32
|
McKey J, Anbarci DN, Bunce C, Ontiveros AE, Behringer RR, Capel B. Integration of mouse ovary morphogenesis with developmental dynamics of the oviduct, ovarian ligaments, and rete ovarii. eLife 2022; 11:e81088. [PMID: 36165446 PMCID: PMC9621696 DOI: 10.7554/elife.81088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/26/2022] [Indexed: 01/29/2023] Open
Abstract
Morphogenetic events during the development of the fetal ovary are crucial to the establishment of female fertility. However, the effects of structural rearrangements of the ovary and surrounding reproductive tissues on ovary morphogenesis remain largely uncharacterized. Using tissue clearing and lightsheet microscopy, we found that ovary folding correlated with regionalization into cortex and medulla. Relocation of the oviduct to the ventral aspect of the ovary led to ovary encapsulation, and mutual attachment of the ovary and oviduct to the cranial suspensory ligament likely triggered ovary folding. During this process, the rete ovarii (RO) elaborated into a convoluted tubular structure extending from the ovary into the ovarian capsule. Using genetic mouse models in which the oviduct and RO are perturbed, we found the oviduct is required for ovary encapsulation. This study reveals novel relationships among the ovary and surrounding tissues and paves the way for functional investigation of the relationship between architecture and differentiation of the mammalian ovary.
Collapse
Affiliation(s)
- Jennifer McKey
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Dilara N Anbarci
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Corey Bunce
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| | - Alejandra E Ontiveros
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Richard R Behringer
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
33
|
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
|