1
|
Wilhelm D, Perea-Gomez A, Newton A, Chaboissier MC. Gonadal sex determination in vertebrates: rethinking established mechanisms. Development 2025; 152:dev204592. [PMID: 40162719 DOI: 10.1242/dev.204592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Sex determination and differentiation are fundamental processes that are not only essential for fertility but also influence the development of many other organs, and hence, are important for species diversity and survival. In mammals, sex is determined by the inheritance of an X or a Y chromosome from the father. The Y chromosome harbours the testis-determining gene SRY, and it has long been thought that its absence is sufficient for ovarian development. Consequently, the ovarian pathway has been treated as a default pathway, in the sense that ovaries do not have or need a female-determining factor. Recently, a female-determining factor has been identified in mouse as the master regulator of ovarian development. Interestingly, this scenario was predicted as early as 1983. In this Review, we discuss the model predicted in 1983, how the mechanisms and genes currently known to be important for sex determination and differentiation in mammals have changed or supported this model, and finally, reflect on what these findings might mean for sex determination in other vertebrates.
Collapse
Affiliation(s)
- Dagmar Wilhelm
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Aitana Perea-Gomez
- Université Côte d'Azur, INSERM, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Axel Newton
- TIGRR Lab, The School of BioSciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | | |
Collapse
|
2
|
Bogdanov A, Sokolova M, Bakloushinskaya I. Specificity of Key Sex Determination Genes in a Mammal with Ovotestes: The European Mole Talpa europaea. Animals (Basel) 2024; 14:2180. [PMID: 39123706 PMCID: PMC11311037 DOI: 10.3390/ani14152180] [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: 04/29/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Here, for the first time, the structure of genes involved in sex determination in mammals (full Sry and partial Rspo1, Eif2s3x, and Eif2s3y) was analyzed for the European mole Talpa europaea with ovotestes in females. We confirmed male-specificity for Eif2s3y and Sry. Five exons were revealed for Rspo1 and the deep similarity with the structure of this gene in T. occidentalis was proved. The most intriguing result was obtained for the Sry gene, which, in placental mammals, initiates male development. We described two exons for this canonically single-exon gene: the first (initial) exon is only 15 bp while the second exon includes 450 bp. The exons are divided by an extended intron of about 1894 bp, including the fragment of the LINE retroposon. Moreover, in chromatogram fragments, which correspond to intron and DNA areas, flanking both exons, we revealed double peaks, similar to heterozygous nucleotide sites of autosomal genes. This may indicate the existence of two or more copies of the Sry gene. Proof of copies requires an additional in-depth study. We hypothesize that unusual structure and possible supernumerary copies of Sry may be involved in ovotestes formation.
Collapse
Affiliation(s)
- Alexey Bogdanov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.B.); (M.S.)
| | - Maria Sokolova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.B.); (M.S.)
- Biological Department, Lomonosov State University, 119234 Moscow, Russia
| | - Irina Bakloushinskaya
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (A.B.); (M.S.)
| |
Collapse
|
3
|
Ferrari MTM, Silva ESDN, Nishi MY, Batista RL, Mendonca BB, Domenice S. Testicular differentiation in 46,XX DSD: an overview of genetic causes. Front Endocrinol (Lausanne) 2024; 15:1385901. [PMID: 38721146 PMCID: PMC11076692 DOI: 10.3389/fendo.2024.1385901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/18/2024] [Indexed: 01/18/2025] Open
Abstract
In mammals, the development of male or female gonads from fetal bipotential gonads depends on intricate genetic networks. Changes in dosage or temporal expression of sex-determining genes can lead to differences of gonadal development. Two rare conditions are associated with disruptions in ovarian determination, including 46,XX testicular differences in sex development (DSD), in which the 46,XX gonads differentiate into testes, and 46,XX ovotesticular DSD, characterized by the coexistence of ovarian and testicular tissue in the same individual. Several mechanisms have been identified that may contribute to the development of testicular tissue in XX gonads. This includes translocation of SRY to the X chromosome or an autosome. In the absence of SRY, other genes associated with testis development may be overexpressed or there may be a reduction in the activity of pro-ovarian/antitesticular factors. However, it is important to note that a significant number of patients with these DSD conditions have not yet recognized a genetic diagnosis. This finding suggests that there are additional genetic pathways or epigenetic mechanisms that have yet to be identified. The text will provide an overview of the current understanding of the genetic factors contributing to 46,XX DSD, specifically focusing on testicular and ovotesticular DSD conditions. It will summarize the existing knowledge regarding the genetic causes of these differences. Furthermore, it will explore the potential involvement of other factors, such as epigenetic mechanisms, in developing these conditions.
Collapse
Affiliation(s)
- Maria Tereza Martins Ferrari
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Elinaelma Suelane do Nascimento Silva
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mirian Yumie Nishi
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Loch Batista
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Sosa E, Mumu SK, Alvarado CC, Wu QY, Roberson I, Espinoza A, Hsu FM, Saito K, Hunt TJ, Faith JE, Lowe MG, DiRusso JA, Clark AT. Reconstituted ovaries self-assemble without an ovarian surface epithelium. Stem Cell Reports 2023; 18:2190-2202. [PMID: 37890483 PMCID: PMC10679655 DOI: 10.1016/j.stemcr.2023.10.001] [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: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Three-dimensional (3D) stem cell models of the ovary have the potential to benefit women's reproductive health research. One such model, the reconstituted ovary (rOvary) self-assembles with pluripotent stem cell-derived germ cells creating a 3D ovarian mimic competent to support the differentiation of functional oocytes inside follicles. In this study, we evaluated the cellular composition of the rOvary revealing the capacity to generate multiple follicles surrounded by NR2F2+ stroma cells. However, the rOvary does not develop a surface epithelium, the source of second-wave pre-granulosa cells, or steroidogenic theca. Therefore, the rOvary models represent the self-assembly of activated follicles in a pre-pubertal ovary poised but not yet competent for hormone production.
Collapse
Affiliation(s)
- Enrique Sosa
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sinthia Kabir Mumu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christian C Alvarado
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qiu Ya Wu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Isaias Roberson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences - The Collaboratory, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Fei-Man Hsu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kaori Saito
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy J Hunt
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jared E Faith
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew G Lowe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan A DiRusso
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amander T Clark
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
5
|
Bird AD, Frost ER, Bagheri-Fam S, Croft BM, Ryan JM, Zhao L, Koopman P, Harley VR. Somatic FGFR2 is Required for Germ Cell Maintenance in the Mouse Ovary. Endocrinology 2023; 164:7036407. [PMID: 36786658 DOI: 10.1210/endocr/bqad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 02/15/2023]
Abstract
During sex determination in the mouse, fibroblast growth factor 9 signals through the fibroblast growth factor receptor 2c isoform (FGFR2c) to trigger Sertoli cell and testis development from 11.5 days post coitum (dpc). In the XX gonad, the FOXL2 and WNT4/RSPO1 pathways drive granulosa cell and ovarian development. The function of FGFR2 in the developing ovary, and whether FGFR2 is required in the testis after sex determination, is not clear. In fetal mouse gonads from 12.5 dpc, FGFR2 shows sexually dimorphic expression. In XX gonads, FGFR2c is coexpressed with FOXL2 in pregranulosa cells, whereas XY gonads show FGFR2b expression in germ cells. Deletion of Fgfr2c in XX mice led to a marked decrease/absence of germ cells by 13.5 dpc in the ovary. This indicates that FGFR2c in the somatic pregranulosa cells is required for the maintenance of germ cells. Surprisingly, on the Fgfr2c-/- background, the germ cell phenotype could be rescued by ablation of Foxl2, suggesting a novel mechanism whereby FGFR2 and FOXL2 act antagonistically during germ cell development. Consistent with low/absent FGFR2 expression in the Sertoli cells of 12.5 and 13.5 dpc XY gonads, XY AMH:Cre; Fgfr2flox/flox mice showed normal testis morphology and structures during fetal development and in adulthood. Thus, FGFR2 is not essential for maintaining Sertoli cell fate after sex determination. Combined, these data show that FGFR2 is not necessary for Sertoli cell function after sex determination but does play an important role in the ovary.
Collapse
Affiliation(s)
- Anthony D Bird
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, 3010, Australia
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
| | - Emily R Frost
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Stefan Bagheri-Fam
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Brittany M Croft
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Janelle M Ryan
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
| | - Liang Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Vincent R Harley
- Sex Development Laboratory, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, 3168, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3168, Australia
| |
Collapse
|
6
|
Singhania P, Ghosh A, Das D, Neogi S, Bhattacharjee R, Datta D. A Rare Differences of Sex Development: Male Sex Reversal Syndrome (NonSyndromic 46, XX with Negative Sex-Determining Region of Y Chromosome Gene). J Indian Assoc Pediatr Surg 2023; 28:154-159. [PMID: 37197249 PMCID: PMC10185029 DOI: 10.4103/jiaps.jiaps_109_22] [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: 08/12/2022] [Revised: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 05/19/2023] Open
Abstract
46, XX testicular differences of sex development (DSD) is a rare cause of DSD presenting as a phenotypical male with chromosomal sex of 46, XX. Sex-determining region of the Y chromosome (SRY)-positive 46, XX DSDs have a well-characterized pathogenetic mechanism, whereas in SRY-negative 46, XX DSDs, the pathogenesis is not clearly delineated. Herein, we present a case of a 3½-year-old child who presented with ambiguous genitalia and bilateral palpable gonads. On the basis of a karyotype and fluorescent in situ hybridization, we arrived at a diagnosis of SRY-negative 46, XX testicular DSD. Basal serum estradiol and human menopausal gonadotrophin stimulated estradiol levels and inhibin A blood levels were against the presence of any ovarian tissue. Imaging of the gonads showed bilateral normal-looking testis. A clinical exome sequencing revealed a heterozygous missense variant NR5A1:c275G>A (p. Arg92gln) located at exon 4 in the affected child. Protein structure analysis was further performed, and the variant was found to be highly conserved. Sanger's sequencing showed that the mother was heterozygous for the variant detected in the child. This case highlights the rarity of SRY-negative 46, XX testicular DSD with a unique variant. Largely under characterized, this group of DSDs needs to be reported and analyzed to add to the spectrum of presentation and genetic characteristics. Our case is expected to add to the database, knowledge, and approach to cases of 46, XX testicular DSD.
Collapse
Affiliation(s)
- Pankaj Singhania
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, SSKM Hospital, Kolkata, West Bengal, India
| | - Arunava Ghosh
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, SSKM Hospital, Kolkata, West Bengal, India
| | - Debaditya Das
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, SSKM Hospital, Kolkata, West Bengal, India
| | - Subhasis Neogi
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, SSKM Hospital, Kolkata, West Bengal, India
| | - Rana Bhattacharjee
- Department of Endocrinology and Metabolism, Institute of Post Graduate Medical Education and Research, SSKM Hospital, Kolkata, West Bengal, India
| | - Dipanjana Datta
- Institute of Child Health, Kolkata, West Bengal, India
- Organization of Rare Disease, Bengaluru, Karnataka, India
| |
Collapse
|
7
|
Bunce C, Barske L, Zhang G, Capel B. Biased precursor ingression underlies the center-to-pole pattern of male sex determination in mouse. Development 2023; 150:297121. [PMID: 36912416 PMCID: PMC10112898 DOI: 10.1242/dev.201060] [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: 06/30/2022] [Accepted: 02/15/2023] [Indexed: 03/14/2023]
Abstract
During mammalian development, gonadal sex determination results from the commitment of bipotential supporting cells to Sertoli or granulosa cell fates. Typically, this decision is coordinated across the gonad to ensure commitment to a single organ fate. When unified commitment fails in an XY mouse, an ovotestis forms in which supporting cells in the center of the gonad typically develop as Sertoli cells, while supporting cells in the poles develop as granulosa cells. This central bias for Sertoli cell fate was thought to result from the initial expression of the drivers of Sertoli cell fate, SRY and/or SOX9, in the central domain, followed by paracrine expansion to the poles. However, we show here that the earliest cells expressing SRY and SOX9 are widely distributed across the gonad. In addition, Sertoli cell fate does not spread among supporting cells through paracrine relay. Instead, we uncover a center-biased pattern of supporting cell precursor ingression that occurs in both sexes and results in increased supporting cell density in the central domain. Our findings prompt a new model of gonad patterning in which a density-dependent organizing principle dominates Sertoli cell fate stabilization.
Collapse
Affiliation(s)
- Corey Bunce
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lindsey Barske
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Gloria Zhang
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
Clinton M, Zhao D. Avian Sex Determination: A Chicken and Egg Conundrum. Sex Dev 2023; 17:120-133. [PMID: 36796340 PMCID: PMC10659007 DOI: 10.1159/000529754] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Primary sex determination is the developmental process that results in the sexual differentiation of the gonads. Vertebrate sex determination is generally considered to follow the model based on the mammalian system, where a sex-specific master regulatory gene activates one of the two different gene networks that underlie testis and ovary differentiation. SUMMARY It is now known that, while many of the molecular components of these pathways are conserved across different vertebrates, a wide variety of different trigger factors are utilized to initiate primary sex determination. In birds, the male is the homogametic sex (ZZ), and significant differences exist between the avian system of sex determination and that of mammals. For example, DMRT1, FOXL2, and estrogen are key factors in gonadogenesis in birds, but none are essential for primary sex determination in mammals. KEY MESSAGE Gonadal sex determination in birds is thought to depend on a dosage-based mechanism involving expression of the Z-linked DMRT1 gene, and it may be that this "mechanism" is simply an extension of the cell autonomous sex identity associated with avian tissues, with no sex-specific trigger required.
Collapse
Affiliation(s)
- Michael Clinton
- Roslin Institute Chicken Embryology (RICE) Group, Gene Function and Development, The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Debiao Zhao
- Roslin Institute Chicken Embryology (RICE) Group, Gene Function and Development, The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| |
Collapse
|
9
|
Ogawa Y, Terao M, Tsuji-Hosokawa A, Tsuchiya I, Hasegawa M, Takada S. SOX9 and SRY binding sites on mouse mXYSRa/Enh13 enhancer redundantly regulate Sox9 expression to varying degrees. Hum Mol Genet 2023; 32:55-64. [PMID: 35921234 DOI: 10.1093/hmg/ddac184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023] Open
Abstract
Sox9 plays an essential role in mammalian testis formation. It has been reported that gene expression in the testes is regulated by enhancers. Among them, mXYSRa/Enh13-which is located at far upstream of the transcription start site-plays a critical role, wherein its deletion causes complete male-to-female sex reversal in mice. It has been proposed that the binding sites (BSs) of SOX9 and SRY, the latter of which is the sex determining gene on the Y chromosome, are associated with mXYSRa/Enh13. They function as an enhancer, whereby the sequences are evolutionarily conserved and in vivo binding of SOX9 and SRY to mXYSRa/Enh13 has been demonstrated previously. However, their precise in vivo functions have not been examined to date. To this end, this study generated mice with substitutions on the SOX9 and SRY BSs to reveal their in vivo functions. Homozygous mutants of SOX9 and SRY BS were indistinguishable from XY males, whereas double mutants had small testes, suggesting that these functions are redundant and that there is another functional sequence on mXYSRa/Enh13, since mXYSRa/Enh13 deletion mice are XY females. In addition, the majority of hemizygous mice with substitutions in SOX9 BS and SRY BS were female and male, respectively, suggesting that SOX9 BS contributes more to SRY BS for mXYSRa/Enh13 to function. The additive effect of SOX9 and SRY via these BSs was verified using an in vitro assay. In conclusion, SOX9 BS and SRY BS function redundantly in vivo, and at least one more functional sequence should exist in mXYSRa/Enh13.
Collapse
Affiliation(s)
- Yuya Ogawa
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.,Department of NCCHD, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Miho Terao
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Atsumi Tsuji-Hosokawa
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Iku Tsuchiya
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.,Department of NCCHD, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Midori Hasegawa
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.,Department of NCCHD, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.,Department of NCCHD, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
10
|
Rossitto M, Déjardin S, Rands CM, Le Gras S, Migale R, Rafiee MR, Neirijnck Y, Pruvost A, Nguyen AL, Bossis G, Cammas F, Le Gallic L, Wilhelm D, Lovell-Badge R, Boizet-Bonhoure B, Nef S, Poulat F. TRIM28-dependent SUMOylation protects the adult ovary from activation of the testicular pathway. Nat Commun 2022; 13:4412. [PMID: 35906245 PMCID: PMC9338040 DOI: 10.1038/s41467-022-32061-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/17/2022] [Indexed: 11/08/2022] Open
Abstract
Gonadal sexual fate in mammals is determined during embryonic development and must be actively maintained in adulthood. In the mouse ovary, oestrogen receptors and FOXL2 protect ovarian granulosa cells from transdifferentiation into Sertoli cells, their testicular counterpart. However, the mechanism underlying their protective effect is unknown. Here, we show that TRIM28 is required to prevent female-to-male sex reversal of the mouse ovary after birth. We found that upon loss of Trim28, ovarian granulosa cells transdifferentiate to Sertoli cells through an intermediate cell type, different from gonadal embryonic progenitors. TRIM28 is recruited on chromatin in the proximity of FOXL2 to maintain the ovarian pathway and to repress testicular-specific genes. The role of TRIM28 in ovarian maintenance depends on its E3-SUMO ligase activity that regulates the sex-specific SUMOylation profile of ovarian-specific genes. Our study identifies TRIM28 as a key factor in protecting the adult ovary from the testicular pathway.
Collapse
Affiliation(s)
- Moïra Rossitto
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Stephanie Déjardin
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
| | - Chris M Rands
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Stephanie Le Gras
- GenomEast platform, IGBMC, 1, rue Laurent Fries, 67404 ILLKIRCH Cedex, Illkirch-Graffenstaden, France
| | - Roberta Migale
- The Francis Crick Institute, 1 Midland Road, London, NW1 2 1AT, UK
| | | | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Alain Pruvost
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 91191, Gif-sur-Yvette, France
| | - Anvi Laetitia Nguyen
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 91191, Gif-sur-Yvette, France
| | - Guillaume Bossis
- Institut de Génétique Moléculaire de Montpellier (IGMM), University of Montpellier, CNRS, Montpellier, France
| | - Florence Cammas
- Institut de Recherche en Cancérologie de Montpellier, IRCM, INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Lionel Le Gallic
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | | | | | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva CMU, lab E09.2750.B 1, rue Michel-Servet CH 1211 Geneva 4, Geneva, Switzerland
| | - Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, 34396, Montpellier, France.
| |
Collapse
|
11
|
Rodriguez KF, Brown PR, Amato CM, Nicol B, Liu CF, Xu X, Yao HHC. Somatic cell fate maintenance in mouse fetal testes via autocrine/paracrine action of AMH and activin B. Nat Commun 2022; 13:4130. [PMID: 35840551 PMCID: PMC9287316 DOI: 10.1038/s41467-022-31486-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Fate determination and maintenance of fetal testes in most mammals occur cell autonomously as a result of the action of key transcription factors in Sertoli cells. However, the cases of freemartin, where an XX twin develops testis structures under the influence of an XY twin, imply that hormonal factor(s) from the XY embryo contribute to sex reversal of the XX twin. Here we show that in mouse XY embryos, Sertoli cell-derived anti-Mullerian hormone (AMH) and activin B together maintain Sertoli cell identity. Sertoli cells in the gonadal poles of XY embryos lacking both AMH and activin B transdifferentiate into their female counterpart granulosa cells, leading to ovotestis formation. The ovotestes remain to adulthood and produce both sperm and oocytes, although there are few of the former and the latter fail to mature. Finally, the ability of XY mice to masculinize ovaries is lost in the absence of these two factors. These results provide insight into fate maintenance of fetal testes through the action of putative freemartin factors.
Collapse
Affiliation(s)
- Karina F Rodriguez
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Paula R Brown
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Ciro M Amato
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Barbara Nicol
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Chia-Feng Liu
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xin Xu
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, NC, USA.
| |
Collapse
|
12
|
Ma X, Liu F, Chen Q, Sun W, Shen J, Wu K, Zheng Z, Huang J, Chen J, Qian G, Ge C. Foxl2 is required for the initiation of the female pathway in a temperature-dependent sex determination system in Trachemys scripta. Development 2022; 149:275948. [DOI: 10.1242/dev.200863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
KDM6B-mediated epigenetic modification of the testicular regulator Dmrt1 has previously been identified as the primary switch of the male pathway in a temperature-dependent sex-determination (TSD) system; however, the molecular network of the female pathway has not yet been established. Here, we have functionally characterized for the first time an upstream regulator of the female pathway, the forkhead transcription factor FOXL2, in Trachemys scripta, a turtle species with a TSD system. FOXL2 exhibited temperature-dependent female-specific expression patterns before the onset of gonadal differentiation and was preferentially localized in ovarian somatic cells. Foxl2 responded rapidly to temperature shifts and estrogen. Importantly, forced expression of Foxl2 at the male-producing temperature led to male-to-female sex reversal, as evidenced by the formation of an ovary-like structure, and upregulation of the ovarian regulators Cyp19a1 and R-spondin1. Additionally, knockdown of Foxl2 caused masculinization at the female-producing temperature, which was confirmed by loss of the female phenotype, development of seminiferous tubules, and elevated expression of Dmrt1 and Sox9. Collectively, we demonstrate that Foxl2 expression is necessary and sufficient to drive ovarian determination in T. scripta, suggesting a crucial role of Foxl2 in female sex determination in the TSD system.
Collapse
Affiliation(s)
- Xiaohui Ma
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
- College of Fisheries and Life Sciences, Shanghai Ocean University 2 , Shanghai 201306 , China
| | - Fang Liu
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
- College of Fisheries and Life Sciences, Shanghai Ocean University 2 , Shanghai 201306 , China
| | - Qiran Chen
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
- College of Marine Life Sciences, Ocean University of China 3 MOE Key Laboratory of Marine Genetics and Breeding , , Qingdao 266003 , China
| | - Wei Sun
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Jiadong Shen
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Kaiyue Wu
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Ziyan Zheng
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Jiaqi Huang
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Jiawen Chen
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Guoying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| | - Chutian Ge
- College of Biological and Environmental Sciences, Zhejiang Wanli University 1 , Ningbo 315100 , China
| |
Collapse
|
13
|
Windley SP, Mayère C, McGovern AE, Harvey NL, Nef S, Schwarz Q, Kumar S, Wilhelm D. Loss of NEDD4 causes complete XY gonadal sex reversal in mice. Cell Death Dis 2022; 13:75. [PMID: 35075134 PMCID: PMC8786929 DOI: 10.1038/s41419-022-04519-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/16/2021] [Accepted: 01/06/2022] [Indexed: 11/24/2022]
Abstract
Gonadogenesis is the process wherein two morphologically distinct organs, the testis and the ovary, arise from a common precursor. In mammals, maleness is driven by the expression of Sry. SRY subsequently upregulates the related family member Sox9 which is responsible for initiating testis differentiation while repressing factors critical to ovarian development such as FOXL2 and β-catenin. Here, we report a hitherto uncharacterised role for the ubiquitin-protein ligase NEDD4 in this process. XY Nedd4-deficient mice exhibit complete male-to-female gonadal sex reversal shown by the ectopic upregulation of Foxl2 expression at the time of gonadal sex determination as well as insufficient upregulation of Sox9. This sex reversal extends to germ cells with ectopic expression of SYCP3 in XY Nedd4-/- germ cells and significantly higher Sycp3 transcripts in XY and XX Nedd4-deficient mice when compared to both XY and XX controls. Further, Nedd4-/- mice exhibit reduced gonadal precursor cell formation and gonadal size as a result of reduced proliferation within the developing gonad as well as reduced Nr5a1 expression. Together, these results establish an essential role for NEDD4 in XY gonadal sex determination and development and suggest a potential role for NEDD4 in orchestrating these cell fate decisions through the suppression of the female pathway to ensure proper testis differentiation.
Collapse
Affiliation(s)
- Simon P Windley
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, 3010, Australia
| | - Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland
| | - Alice E McGovern
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, 3010, Australia
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia, Adelaide, 5001, Australia
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia, Adelaide, 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, 5001, Australia
| | - Dagmar Wilhelm
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
14
|
Syryn H, Van De Vijver K, Cools M. Ovotesticular Difference of Sex Development: Genetic Background, Histological Features, and Clinical Management. Horm Res Paediatr 2021; 96:180-189. [PMID: 34469891 DOI: 10.1159/000519323] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Ovotesticular disorder/difference of sex development (DSD) refers to the co-presence of testicular and ovarian tissue in one individual. Childhood management is challenging as there are many uncertainties regarding etiology, gonadal function, and gender outcome. SUMMARY Ovotesticular DSD should mainly be considered in 46,XX children with atypical genitalia and normal adrenal steroid profiles. Various underlying genetic mechanisms have been described. Histological assessment of ovotestes requires expert revision and has many pitfalls. Neonatal sex assignment is essential, but as gender outcome is unpredictable, this should be regarded as provisional until a stable gender identity has developed. Therefore, it is crucial not to perform any irreversible medical or surgical procedure in affected individuals until adolescents can give their full informed consent. Gonadal function mostly allows for spontaneous pubertal development; however, fertility is compromised, especially in boys. Specific long-term outcome data for ovotesticular DSD are lacking but can be extrapolated from studies in other DSD populations. Key Messages: Management of ovotesticular DSD has changed in recent years, prioritizing the child's future right for autonomy and self-determination. The benefits and pitfalls of this new approach have not been documented yet and require intensive monitoring on an international scale.
Collapse
Affiliation(s)
- Hannes Syryn
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | | | - Martine Cools
- Department of Internal Medicine and Pediatrics, Ghent University and Pediatric Endocrinology Service, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
15
|
Lecluze E, Rolland AD, Filis P, Evrard B, Leverrier-Penna S, Maamar MB, Coiffec I, Lavoué V, Fowler PA, Mazaud-Guittot S, Jégou B, Chalmel F. Dynamics of the transcriptional landscape during human fetal testis and ovary development. Hum Reprod 2021; 35:1099-1119. [PMID: 32412604 DOI: 10.1093/humrep/deaa041] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION Which transcriptional program triggers sex differentiation in bipotential gonads and downstream cellular events governing fetal testis and ovary development in humans? SUMMARY ANSWER The characterization of a dynamically regulated protein-coding and non-coding transcriptional landscape in developing human gonads of both sexes highlights a large number of potential key regulators that show an early sexually dimorphic expression pattern. WHAT IS KNOWN ALREADY Gonadal sex differentiation is orchestrated by a sexually dimorphic gene expression program in XX and XY developing fetal gonads. A comprehensive characterization of its non-coding counterpart offers promising perspectives for deciphering the molecular events underpinning gonad development and for a complete understanding of the etiology of disorders of sex development in humans. STUDY DESIGN, SIZE, DURATION To further investigate the protein-coding and non-coding transcriptional landscape during gonad differentiation, we used RNA-sequencing (RNA-seq) and characterized the RNA content of human fetal testis (N = 24) and ovaries (N = 24) from 6 to 17 postconceptional week (PCW), a key period in sex determination and gonad development. PARTICIPANTS/MATERIALS, SETTING, METHODS First trimester fetuses (6-12 PCW) and second trimester fetuses (13-14 and 17 PCW) were obtained from legally induced normally progressing terminations of pregnancy. Total RNA was extracted from whole human fetal gonads and sequenced as paired-end 2 × 50 base reads. Resulting sequences were mapped to the human genome, allowing for the assembly and quantification of corresponding transcripts. MAIN RESULTS AND THE ROLE OF CHANCE This RNA-seq analysis of human fetal testes and ovaries at seven key developmental stages led to the reconstruction of 22 080 transcripts differentially expressed during testicular and/or ovarian development. In addition to 8935 transcripts displaying sex-independent differential expression during gonad development, the comparison of testes and ovaries enabled the discrimination of 13 145 transcripts that show a sexually dimorphic expression profile. The latter include 1479 transcripts differentially expressed as early as 6 PCW, including 39 transcription factors, 40 long non-coding RNAs and 20 novel genes. Despite the use of stringent filtration criteria (expression cut-off of at least 1 fragment per kilobase of exon model per million reads mapped, fold change of at least 2 and false discovery rate adjusted P values of less than <1%), the possibility of assembly artifacts and of false-positive differentially expressed transcripts cannot be fully ruled out. LARGE-SCALE DATA Raw data files (fastq) and a searchable table (.xlss) containing information on genomic features and expression data for all refined transcripts have been submitted to the NCBI GEO under accession number GSE116278. LIMITATIONS, REASONS FOR CAUTION The intrinsic nature of this bulk analysis, i.e. the sequencing of transcripts from whole gonads, does not allow direct identification of the cellular origin(s) of the transcripts characterized. Potential cellular dilution effects (e.g. as a result of distinct proliferation rates in XX and XY gonads) may account for a few of the expression profiles identified as being sexually dimorphic. Finally, transcriptome alterations that would result from exposure to pre-abortive drugs cannot be completely excluded. Although we demonstrated the high quality of the sorted cell populations used for experimental validations using quantitative RT-PCR, it cannot be totally excluded that some germline expression may correspond to cell contamination by, for example, macrophages. WIDER IMPLICATIONS OF THE FINDINGS For the first time, this study has led to the identification of 1000 protein-coding and non-coding candidate genes showing an early, sexually dimorphic, expression pattern that have not previously been associated with sex differentiation. Collectively, these results increase our understanding of gonad development in humans, and contribute significantly to the identification of new candidate genes involved in fetal gonad differentiation. The results also provide a unique resource that may improve our understanding of the fetal origin of testicular and ovarian dysgenesis syndromes, including cryptorchidism and testicular cancers. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the French National Institute of Health and Medical Research (Inserm), the University of Rennes 1, the French School of Public Health (EHESP), the Swiss National Science Foundation [SNF n° CRS115_171007 to B.J.], the French National Research Agency [ANR n° 16-CE14-0017-02 and n° 18-CE14-0038-02 to F.C.], the Medical Research Council [MR/L010011/1 to P.A.F.] and the European Community's Seventh Framework Programme (FP7/2007-2013) [under grant agreement no 212885 to P.A.F.] and from the European Union's Horizon 2020 Research and Innovation Programme [under grant agreement no 825100 to P.A.F. and S.M.G.]. There are no competing interests related to this study.
Collapse
Affiliation(s)
- Estelle Lecluze
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Antoine D Rolland
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Panagiotis Filis
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Bertrand Evrard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Sabrina Leverrier-Penna
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.,Univ Poitiers, STIM, CNRS ERL7003, Poitiers Cedex 9, CNRS ERL7003, France
| | - Millissia Ben Maamar
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Isabelle Coiffec
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Vincent Lavoué
- Service Gynécologie et Obstétrique, CHU Rennes, F-35000 Rennes, France
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Séverine Mazaud-Guittot
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Bernard Jégou
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
16
|
Bagheri-Fam S, Combes AN, Ling CK, Wilhelm D. Heterozygous deletion of Sox9 in mouse mimics the gonadal sex reversal phenotype associated with campomelic dysplasia in humans. Hum Mol Genet 2020; 29:3781-3792. [PMID: 33305798 DOI: 10.1093/hmg/ddaa259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/11/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Heterozygous mutations in the human SOX9 gene cause the skeletal malformation syndrome campomelic dysplasia which in 75% of 46, XY individuals is associated with male-to-female sex reversal. Although studies in homozygous Sox9 knockout mouse models confirmed that SOX9 is critical for testis development, mice heterozygous for the Sox9-null allele were reported to develop normal testes. This led to the belief that the SOX9 dosage requirement for testis differentiation is different between humans, which often require both alleles, and mice, in which one allele is sufficient. However, in prior studies, gonadal phenotypes in heterozygous Sox9 XY mice were assessed only by either gross morphology, histological staining or analyzed on a mixed genetic background. In this study, we conditionally inactivated Sox9 in somatic cells of developing gonads using the Nr5a1-Cre mouse line on a pure C57BL/6 genetic background. Section and whole-mount immunofluorescence for testicular and ovarian markers showed that XY Sox9 heterozygous gonads developed as ovotestes. Quantitative droplet digital PCR confirmed a 50% reduction of Sox9 mRNA as well as partial sex reversal shown by an upregulation of ovarian genes. Our data show that haploinsufficiency of Sox9 can perturb testis development in mice, suggesting that mice may provide a more accurate model of human disorders/differences of sex development than previously thought.
Collapse
Affiliation(s)
- Stefan Bagheri-Fam
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexander N Combes
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC 3052, Australia
| | - Cheuk K Ling
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Dagmar Wilhelm
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
17
|
Nagahama Y, Chakraborty T, Paul-Prasanth B, Ohta K, Nakamura M. Sex determination, gonadal sex differentiation, and plasticity in vertebrate species. Physiol Rev 2020; 101:1237-1308. [PMID: 33180655 DOI: 10.1152/physrev.00044.2019] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A diverse array of sex determination (SD) mechanisms, encompassing environmental to genetic, have been found to exist among vertebrates, covering a spectrum from fixed SD mechanisms (mammals) to functional sex change in fishes (sequential hermaphroditic fishes). A major landmark in vertebrate SD was the discovery of the SRY gene in 1990. Since that time, many attempts to clone an SRY ortholog from nonmammalian vertebrates remained unsuccessful, until 2002, when DMY/dmrt1by was discovered as the SD gene of a small fish, medaka. Surprisingly, however, DMY/dmrt1by was found in only 2 species among more than 20 species of medaka, suggesting a large diversity of SD genes among vertebrates. Considerable progress has been made over the last 3 decades, such that it is now possible to formulate reasonable paradigms of how SD and gonadal sex differentiation may work in some model vertebrate species. This review outlines our current understanding of vertebrate SD and gonadal sex differentiation, with a focus on the molecular and cellular mechanisms involved. An impressive number of genes and factors have been discovered that play important roles in testicular and ovarian differentiation. An antagonism between the male and female pathway genes exists in gonads during both sex differentiation and, surprisingly, even as adults, suggesting that, in addition to sex-changing fishes, gonochoristic vertebrates including mice maintain some degree of gonadal sexual plasticity into adulthood. Importantly, a review of various SD mechanisms among vertebrates suggests that this is the ideal biological event that can make us understand the evolutionary conundrums underlying speciation and species diversity.
Collapse
Affiliation(s)
- Yoshitaka Nagahama
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Faculty of Biological Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Tapas Chakraborty
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan.,Karatsu Satellite of Aqua-Bioresource Innovation Center, Kyushu University, Karatsu, Japan
| | - Bindhu Paul-Prasanth
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidapeetham, Kochi, Kerala, India
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan
| | - Masaru Nakamura
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Research Center, Okinawa Churashima Foundation, Okinawa, Japan
| |
Collapse
|
18
|
Molecular identification and expression analysis of foxl2 and sox9b in Oryzias celebensis. AQUACULTURE AND FISHERIES 2020. [DOI: 10.1016/j.aaf.2020.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Richardson N, Gillot I, Gregoire EP, Youssef SA, de Rooij D, de Bruin A, De Cian MC, Chaboissier MC. Sox8 and Sox9 act redundantly for ovarian-to-testicular fate reprogramming in the absence of R-spondin1 in mouse sex reversals. eLife 2020; 9:53972. [PMID: 32450947 PMCID: PMC7250573 DOI: 10.7554/elife.53972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/20/2020] [Indexed: 12/17/2022] Open
Abstract
In mammals, testicular differentiation is initiated by transcription factors SRY and SOX9 in XY gonads, and ovarian differentiation involves R-spondin1 (RSPO1) mediated activation of WNT/β-catenin signaling in XX gonads. Accordingly, the absence of RSPO1/Rspo1 in XX humans and mice leads to testicular differentiation and female-to-male sex reversal in a manner that does not requireSry or Sox9 in mice. Here we show that an alternate testis-differentiating factor exists and that this factor is Sox8. Specifically, genetic ablation of Sox8 and Sox9 prevents ovarian-to-testicular reprogramming observed in XX Rspo1 loss-of-function mice. Consequently, Rspo1 Sox8 Sox9 triple mutant gonads developed as atrophied ovaries. Thus, SOX8 alone can compensate for the loss of SOX9 for Sertoli cell differentiation during female-to-male sex reversal. In humans, mice and other mammals, genetic sex is determined by the combination of sex chromosomes that each individual inherits. Individuals with two X chromosomes (XX) are said to be chromosomally female, while individuals with one X and one Y chromosome (XY) are chromosomally males. One of the major differences between XX and XY individuals is that they have different types of gonads (the organs that make egg cells or sperm). In mice, for example, before males are born, a gene called Sox9 triggers a cascade of events that result in the gonads developing into testes. In females, on the other hand, another gene called Rspo1 stimulates the gonads to develop into ovaries. Loss of Sox9 in XY embryos, or Rspo1 in XX embryos, leads to mice developing physical characteristics that do not match their genetic sex, a phenomenon known as sex reversal. For example, in XX female mice lacking Rspo1, cells in the gonads reprogram into testis cells known as Sertoli cells just before birth and form male structures known as testis cords. The gonads of female mice missing both Sox9 and Rspo1 (referred to as “double mutants”) also develop Sertoli cells and testis cords, suggesting another gene may compensate for the loss of Sox9. Previous studies suggest that a gene known as Sox8, which is closely related to Sox9, may be able to drive sex reversal in female mice. However, it was not clear whether Sox8 is able to stimulate testis to form in female mice in the absence of Sox9. To address this question, Richardson et al. studied mutant female mice lacking Rspo1, Sox8 and Sox9, known as “triple mutants”. Just before birth, the gonads in the triple mutant mice showed some characteristics of sex reversal but lacked the Sertoli cells found in the double mutant mice. After the mice were born, the gonads of the triple mutant mice developed as rudimentary ovaries without testis cords, unlike the more testis-like gonads found in the double mutant mice. The findings of Richardson et al. show that Sox8 is able to trigger sex reversal in female mice in the absence of Rspo1 and Sox9. Differences in sexual development in humans affect the appearance of individuals and often cause infertility. Identifying Sox8 and other similar genes in mice may one day help to diagnose people with such conditions and lead to the development of new therapies.
Collapse
Affiliation(s)
| | | | | | - Sameh A Youssef
- Department of Pathobiology, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department Pediatrics, Divisions Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Dirk de Rooij
- Department of Biology, Faculty of Science, Division of Developmental Biology, Reproductive Biology Group, Utrecht University, Utrecht, Netherlands
| | - Alain de Bruin
- Department of Pathobiology, Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department Pediatrics, Divisions Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | |
Collapse
|
20
|
Ortega EA, Salvador Q, Fernandez M, Ward MA. Alterations of sex determination pathways in the genital ridges of males with limited Y chromosome genes†. Biol Reprod 2020; 100:810-823. [PMID: 30285093 DOI: 10.1093/biolre/ioy218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/09/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022] Open
Abstract
We previously demonstrated that in the mouse only two Y chromosome genes are required for a male to produce an offspring with the help of assisted reproduction technologies (ART): testis determinant Sry and spermatogonial proliferation factor Eif2s3y. Subsequently, we have shown that the function of these genes can be replaced by transgenic overexpression of their homologs, autosomally encoded Sox9 and X-chromosome encoded Eif2s3x. Males with Y chromosome contribution limited to two (XEif2s3yOSry), one (XEif2s3yOSox9 and XOSry,Eif2s3x), and no genes (XOSox9,Eif2s3x) produced haploid germ cells and sired offspring after ART. However, despite successful assisted reproductive outcome, they had smaller testes and displayed abnormal development of the seminiferous epithelium and testicular interstitium. Here we explored whether these testicular defects originated from altered pro-testis and pro-ovary factor signaling in genital ridges at the time of sex determination. Timed pregnancies were generated to obtain transgenic XEif2s3yOSry, XEif2s3yOSox9, XOSry,Eif2s3x, XOSox9,Eif2s3x, and wild-type XX and XY fetuses at 12.5 days post coitum. Dissected genital ridges were assessed for their morphology and anatomy, and expression of pro-testis and pro-ovary transcripts. All transgenic males displayed incomplete masculinization of gonadal shape, impaired development of testicular cords and gonadal vasculature, and decreased expression of factors promoting male pathway. Fetal gonad masculinization was more effective when sex determination was driven by the Sry transgene, in the presence of Y chromosome genes, and to a lesser extent a double dosage of X genes. The study adds to the understanding of the role of Y chromosome genes and their homologs during sex determination.
Collapse
Affiliation(s)
- Eglė A Ortega
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Quinci Salvador
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Mayumi Fernandez
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Monika A Ward
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
21
|
Manning J, Windley SP, Sandow JJ, Shah SS, Western P, Wilhelm D, Kumar S. Identification of novel interacting partners of the NEDD4 ubiquitin ligase in mouse testis. J Proteomics 2020; 223:103830. [PMID: 32450490 DOI: 10.1016/j.jprot.2020.103830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 04/14/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
Abstract
Posttranslational modification by ubiquitination targets proteins for degradation, recycling, stabilization or altered trafficking, and as such can alter cellular signaling pathways. The substrate specificity of this multistep process is controlled by ubiquitin ligases, including those of the HECT domain-containing NEDD4 family. In the testis, ubiquitination of many proteins contributes to organ development and maturation of spermatozoa and NEDD4 is known to be important in the control of spermatogonial stem cell homeostasis. However, a comprehensive understanding of NEDD4 substrates in testis development is lacking. Here we demonstrate high expression of Nedd4 in somatic cells of the mouse testis and in the murine Leydig cell-like cell line TM3. Immunoprecipitation of NEDD4 tagged with GFP at either the amino or carboxyl terminus was subjected to proteomic analysis for interacting proteins. We identified a substantial list of potential interaction partners, including known NEDD4 substrates, proteins involved in ubiquitination and proteins important for testis development and spermatogenesis. We confirmed the interaction of NEDD4 with a subset of these putative interacting proteins, validating the integrity of the dataset. These potential interactors may be further explored to reveal important roles of NEDD4-mediated ubiquitination in the testis. SIGNIFICANCE: Ubiquitination is important for testis development and function, and NEDD4 is known to ubiquitinate various proteins to affect cellular signaling and development, including those implicated in spermatogenesis. However, substrates of NEDD4 that are important during testis development remain to be identified. Here we report NEDD4 expression in the developing testis and TM3 testicular cell line. This study identifies a substantial list of NEDD4 interacting proteins in the TM3 testicular cell line, with validation of some of these interactions. Hence, this provides novel NEDD4 targets that may contribute to testis development and function that may be further explored.
Collapse
Affiliation(s)
- JantinaA Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia.
| | - Simon P Windley
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jarrod J Sandow
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Sonia S Shah
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Patrick Western
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3800, Australia
| | - Dagmar Wilhelm
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia.
| |
Collapse
|
22
|
Tang F, Richardson N, Albina A, Chaboissier MC, Perea-Gomez A. Mouse Gonad Development in the Absence of the Pro-Ovary Factor WNT4 and the Pro-Testis Factor SOX9. Cells 2020; 9:cells9051103. [PMID: 32365547 PMCID: PMC7291083 DOI: 10.3390/cells9051103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/03/2022] Open
Abstract
The transcription factors SRY and SOX9 and RSPO1/WNT4/β-Catenin signaling act as antagonistic pathways to drive testis and ovary development respectively, from a common gonadal primordium in mouse embryos. In this work, we took advantage of a double knockout mouse model to study gonadal development when Sox9 and Wnt4 are both mutated. We show that the XX gonad mutant for Wnt4 or for both Wnt4 and Sox9 develop as ovotestes, demonstrating that ectopic SOX9 function is not required for the partial female-to-male sex reversal caused by a Wnt4 mutation. Sox9 deletion in XY gonads leads to ovarian development accompanied by ectopic WNT/β-catenin signaling. In XY Sox9 mutant gonads, SRY-positive supporting precursors adopt a female-like identity and develop as pre-granulosa-like cells. This phenotype cannot be fully prevented by the deletion of Wnt4 or Rspo1, indicating that SOX9 is required for the early determination of the male supporting cell identity independently of repressing RSPO1/WNT4/β-Catenin signaling. However, in XY Sox9 Wnt4 double mutant gonads, pre-granulosa cells are not maintained, as they prematurely differentiate as mature granulosa cells and then trans-differentiate into Sertoli-like cells. Together, our results reveal the dynamics of the specific and independent actions of SOX9 and WNT4 during gonadal differentiation: SOX9 is essential in the testis for early specification of male-supporting cells whereas WNT4 functions in the ovary to maintain female-supporting cell identity and inhibit male-specific vascular and steroidogenic cell differentiation.
Collapse
|
23
|
Bagheri-Fam S, Chen H, Wilson S, Ayers K, Hughes J, Sloan-Bena F, Calvel P, Robevska G, Puisac B, Kusz-Zamelczyk K, Gimelli S, Spik A, Jaruzelska J, Warenik-Szymankiewicz A, Faradz S, Nef S, Pié J, Thomas P, Sinclair A, Wilhelm D. The gene encoding the ketogenic enzyme HMGCS2 displays a unique expression during gonad development in mice. PLoS One 2020; 15:e0227411. [PMID: 31910233 PMCID: PMC6946174 DOI: 10.1371/journal.pone.0227411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 12/18/2019] [Indexed: 11/26/2022] Open
Abstract
Disorders/differences of sex development (DSD) cause profound psychological and reproductive consequences for the affected individuals, however, most are still unexplained at the molecular level. Here, we present a novel gene, 3-hydroxy-3-methylglutaryl coenzyme A synthase 2 (HMGCS2), encoding a metabolic enzyme in the liver important for energy production from fatty acids, that shows an unusual expression pattern in developing fetal mouse gonads. Shortly after gonadal sex determination it is up-regulated in the developing testes following a very similar spatial and temporal pattern as the male-determining gene Sry in Sertoli cells before switching to ovarian enriched expression. To test if Hmgcs2 is important for gonad development in mammals, we pursued two lines of investigations. Firstly, we generated Hmgcs2-null mice using CRISPR/Cas9 and found that these mice had gonads that developed normally even on a sensitized background. Secondly, we screened 46,XY DSD patients with gonadal dysgenesis and identified two unrelated patients with a deletion and a deleterious missense variant in HMGCS2 respectively. However, both variants were heterozygous, suggesting that HMGCS2 might not be the causative gene. Analysis of a larger number of patients in the future might shed more light into the possible association of HMGCS2 with human gonadal development.
Collapse
Affiliation(s)
- Stefan Bagheri-Fam
- Department of Anatomy & Neuroscience, The University of Melbourne, Melbourne, Australia
| | - Huijun Chen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Sean Wilson
- Murdoch Children’s Research Institute, Melbourne, Australia
| | - Katie Ayers
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - James Hughes
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | | | - Pierre Calvel
- Department of Genetics, Medicine & Development, University of Geneva, Geneva, Switzerland
| | - Gorjana Robevska
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Beatriz Puisac
- Unit of Clinical Genetics and Functional Genomics, Department of Pharmacology-Physiology, School of Medicine, University of Zaragoza, CIBERER-GCV02 and ISS-Aragon, Zaragoza, Spain
| | | | - Stefania Gimelli
- Service of Genetic Medicine, University Geneva Hospitals, Geneva, Switzerland
| | - Anna Spik
- Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | | | | | - Sultana Faradz
- Center for Biomedical Research Faculty of Medicine Diponegoro University (FMDU), Semarang, Indonesia
| | - Serge Nef
- Service of Genetic Medicine, University Geneva Hospitals, Geneva, Switzerland
| | - Juan Pié
- Unit of Clinical Genetics and Functional Genomics, Department of Pharmacology-Physiology, School of Medicine, University of Zaragoza, CIBERER-GCV02 and ISS-Aragon, Zaragoza, Spain
| | - Paul Thomas
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Andrew Sinclair
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Dagmar Wilhelm
- Department of Anatomy & Neuroscience, The University of Melbourne, Melbourne, Australia
- * E-mail:
| |
Collapse
|
24
|
Mäkelä JA, Koskenniemi JJ, Virtanen HE, Toppari J. Testis Development. Endocr Rev 2019; 40:857-905. [PMID: 30590466 DOI: 10.1210/er.2018-00140] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022]
Abstract
Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades, and these are described along with the description of molecular regulation of testis development.
Collapse
Affiliation(s)
- Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jaakko J Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Helena E Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
25
|
Fang X, Ni N, Gao Y, Vincent DF, Bartholin L, Li Q. A novel mouse model of testicular granulosa cell tumors. Mol Hum Reprod 2019; 24:343-356. [PMID: 29788434 DOI: 10.1093/molehr/gay023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/17/2018] [Indexed: 12/27/2022] Open
Abstract
STUDY QUESTION What is the role of dysregulated transforming growth factor beta (TGFB) signaling in the development of sex cord-stromal tumors in the testis? SUMMARY ANSWER Overactivation of TGFB signaling results in the development of testicular tumors resembling granulosa cell tumors (GrCTs). WHAT IS KNOWN ALREADY In an earlier study, we demonstrated that constitutively active TGFB receptor 1 (TGFBR1) in ovarian somatic cells promotes the development of ovarian GrCTs. However, the consequence of dysregulation of TGFB signaling in the pathobiology of the testis, remains poorly defined. STUDY DESIGN, SIZE, DURATION To identify the impact of dysregulation of TGFB signaling on the testis, we generated mice with constitutive activation of TGFBR1 using anti-Mullerian hormone receptor type 2 (Amhr2)-Cre recombinase. The effect of constitutively active TGFBR1 on testis development and the timeline of testicular tumor formation were examined. We further investigated the molecular features of testicular tumors and determined the expression of beta-catenin (CTNNB1) known to be involved in testicular GrCT development. PARTICIPANTS/MATERIALS, SETTING, METHODS Male mice with constitutive activation of TGFBR1 were examined at various developmental stages (i.e. from 1 week up to 6 months) along with controls. Testis samples were collected and processed for histological and molecular analyses, including haematoxylin and eosin (H and E) staining, real-time PCR, immunohistochemistry, immunofluorescence and western blotting. Immunostaining/immunoblotting and real-time PCR experiments were performed using at least three animals per genotype. Data are presented as mean ± SEM. Statistical significance was determined using unpaired two-tail t-test and reported when P value is <0.05. MAIN RESULTS AND THE ROLE OF CHANCE Mice harboring constitutively active TGFBR1 in the testes developed tumors resembling testicular GrCTs, a rare type of tumors in the testis. The formation of testicular tumors led to altered cell proliferation, loss of germ cells and defective spermatogenesis. Immunohistochemically, these tumors were positive for inhibin alpha (INHA), forkhead box O1 (FOXO1), and more importantly, forkhead box L2 (FOXL2), a protein specifically expressed in the ovary and required for normal granulosa cell differentiation and function. Consistent with the immunohistochemical findings, FOXL2 proteins were only detectable in testes of TGFBR1-CAAcre mice but not those of controls by western blotting, suggesting potential alteration of Sertoli cell fate. To explore mechanisms underlying the tumor-promoting effect of TGFBR1 overactivation, we examined the expression of CTNNB1. The results revealed increased expression of CTNNB1 in testicular tumors in TGFBR1-CAAcre mice. Collectively, this study uncovered tumorigenic function of enhanced TGFB signaling in the testis. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study was performed using mice, and the direct relevance of the experimental paradigm and findings to human testicular GrCTs awaits further investigation. Of note, constitutive activation of TGFBR1 was employed to enhance TGFB/SMAD signaling activity and may not be interpreted as the genetic cause of the disease. WIDER IMPLICATIONS OF THE FINDINGS This mouse model may prove to be a useful addition to the mouse genetics toolkit for GrCT research. Our finding that dysregulation of TGFB signaling results in the development of testicular GrCTs supports a common origin between Sertoli cells and granulosa cells, and highlights the paramount importance of balanced TGFB signaling in reproduction and development. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the National Institutes of Health grant R03HD082416 from the Eunice Kennedy Shriver National Institute of Child Health & Human Development and the New Faculty Start-up Funds from Texas A&M University awarded to Q.L. The authors declare no competing interest.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Yang Gao
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - David F Vincent
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, UK
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
26
|
Nicol B, Grimm SA, Gruzdev A, Scott GJ, Ray MK, Yao HHC. Genome-wide identification of FOXL2 binding and characterization of FOXL2 feminizing action in the fetal gonads. Hum Mol Genet 2019; 27:4273-4287. [PMID: 30212841 DOI: 10.1093/hmg/ddy312] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
The identity of the gonads is determined by which fate, ovarian granulosa cell or testicular Sertoli cell, the bipotential somatic cell precursors choose to follow. In most vertebrates, the conserved transcription factor FOXL2 contributes to the fate of granulosa cells. To understand FOXL2 functions during gonad differentiation, we performed genome-wide analysis of FOXL2 chromatin occupancy in fetal ovaries and established a genetic mouse model that forces Foxl2 expression in the fetal testis. When FOXL2 was ectopically expressed in the somatic cell precursors in the fetal testis, FOXL2 was sufficient to repress Sertoli cell differentiation, ultimately resulting in partial testis-to-ovary sex-reversal. Combining genome-wide analysis of FOXL2 binding in the fetal ovary with transcriptomic analyses of our Foxl2 gain-of-function and previously published Foxl2 loss-of-function models, we identified potential pathways responsible for the feminizing action of FOXL2. Finally, comparison of FOXL2 genome-wide occupancy in the fetal ovary with testis-determining factor SOX9 genome-wide occupancy in the fetal testis revealed extensive overlaps, implying that antagonistic signals between FOXL2 and SOX9 occur at the chromatin level.
Collapse
Affiliation(s)
- Barbara Nicol
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Greg J Scott
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Manas K Ray
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
27
|
Abstract
Mammalian sex determination is triggered by activation of the mammalian sex-determining gene, Sry, in a spatially and temporally controlled manner. Because reduced or delayed Sry expression results in male-to-female sex reversal, male development is highly dependent on the accurate transcription of Sry. SRY dysregulation is a potential cause of human disorders of sex development (DSD). In addition to changes in DNA sequences, gene expression is regulated by epigenetic mechanisms. Epigenetic regulation ensures spatial and temporal accuracy of the expression of developmentally regulated genes. Epigenetic regulation such as histone tail modification, DNA methylation, chromatin remodeling, and non-coding RNA regulation engages several biological processes in multicellular organisms. In recent years, it has been revealed that various types of epigenetic regulation are involved in accurate gonadal differentiation in mammals. In particular, histone modification plays an integral part in sex determination, which is the first step of gonadal differentiation. Here, we focus on the findings on the epigenetic modifications that regulate Sry expression. Finally, we discuss the role of metabolism that potentially alters the epigenetic state in response to environmental cues.
Collapse
Affiliation(s)
- Shingo Miyawaki
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Makoto Tachibana
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
28
|
Nef S, Stévant I, Greenfield A. Characterizing the bipotential mammalian gonad. Curr Top Dev Biol 2019; 134:167-194. [DOI: 10.1016/bs.ctdb.2019.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
29
|
Zhao L, Wang C, Lehman ML, He M, An J, Svingen T, Spiller CM, Ng ET, Nelson CC, Koopman P. Transcriptomic analysis of mRNA expression and alternative splicing during mouse sex determination. Mol Cell Endocrinol 2018; 478:84-96. [PMID: 30053582 DOI: 10.1016/j.mce.2018.07.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/23/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022]
Abstract
Mammalian sex determination hinges on sexually dimorphic transcriptional programs in developing fetal gonads. A comprehensive view of these programs is crucial for understanding the normal development of fetal testes and ovaries and the etiology of human disorders of sex development (DSDs), many of which remain unexplained. Using strand-specific RNA-sequencing, we characterized the mouse fetal gonadal transcriptome from 10.5 to 13.5 days post coitum, a key time window in sex determination and gonad development. Our dataset benefits from a greater sensitivity, accuracy and dynamic range compared to microarray studies, allows global dynamics and sex-specificity of gene expression to be assessed, and provides a window to non-transcriptional events such as alternative splicing. Spliceomic analysis uncovered female-specific regulation of Lef1 splicing, which may contribute to the enhanced WNT signaling activity in XX gonads. We provide a user-friendly visualization tool for the complete transcriptomic and spliceomic dataset as a resource for the field.
Collapse
Affiliation(s)
- Liang Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Chenwei Wang
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - Melanie L Lehman
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - Mingyu He
- Longsoft, Brisbane, Queensland, 4109, Australia
| | - Jiyuan An
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - Terje Svingen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Cassy M Spiller
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ee Ting Ng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, 4102, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
30
|
Fu A, Oberholtzer SM, Bagheri-Fam S, Rastetter RH, Holdreith C, Caceres VL, John SV, Shaw SA, Krentz KJ, Zhang X, Hui CC, Wilhelm D, Jorgensen JS. Dynamic expression patterns of Irx3 and Irx5 during germline nest breakdown and primordial follicle formation promote follicle survival in mouse ovaries. PLoS Genet 2018; 14:e1007488. [PMID: 30071018 PMCID: PMC6071956 DOI: 10.1371/journal.pgen.1007488] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/13/2018] [Indexed: 01/12/2023] Open
Abstract
Women and other mammalian females are born with a finite supply of oocytes that determine their reproductive lifespan. During fetal development, individual oocytes are enclosed by a protective layer of granulosa cells to form primordial follicles that will grow, mature, and eventually release the oocyte for potential fertilization. Despite the knowledge that follicles are dysfunctional and will die without granulosa cell-oocyte interactions, the mechanisms by which these cells establish communication is unknown. We previously identified that two members of the Iroquois homeobox transcription factor gene family, Irx3 and Irx5, are expressed within developing ovaries but not testes. Deletion of both factors (Irx3-Irx5EGFP/Irx3-Irx5EGFP) disrupted granulosa cell-oocyte contact during early follicle development leading to oocyte death. Thus, we hypothesized that Irx3 and Irx5 are required to develop cell-cell communication networks to maintain follicle integrity and female fertility. A series of Irx3 and Irx5 mutant mouse models were generated to assess roles for each factor. While both Irx3 and Irx5 single mutant females were subfertile, their breeding outcomes and ovary histology indicated distinct causes. Careful analysis of Irx3- and Irx5-reporter mice linked the cause of this disparity to dynamic spatio-temporal changes in their expression patterns. Both factors marked the progenitor pre-granulosa cell population in fetal ovaries. At the critical phase of germline nest breakdown and primordial follicle formation however, Irx3 and Irx5 transitioned to oocyte- and granulosa cell-specific expression respectively. Further investigation into the cause of follicle death in Irx3-Irx5EGFP/Irx3-Irx5EGFP ovaries uncovered specific defects in both granulosa cells and oocytes. Granulosa cell defects included poor contributions to basement membrane deposition and mis-localization of gap junction proteins. Granulosa cells and oocytes both presented fewer cell projections resulting in compromised cell-cell communication. Altogether, we conclude that Irx3 and Irx5 first work together to define the pregranulosa cell population of germline nests. During primordial follicle formation, they transition to oocyte- and granulosa cell-specific expression patterns where they cooperate in neighboring cells to build the foundation for follicle integrity. This foundation is left as their legacy of the essential oocyte-granulosa cell communication network that ensures and ultimately optimizes the integrity of the ovarian reserve and therefore, the female reproductive lifespan.
Collapse
Affiliation(s)
- Anqi Fu
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Sydney M. Oberholtzer
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Stefan Bagheri-Fam
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - Raphael H. Rastetter
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Claire Holdreith
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Valeria L. Caceres
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Steven V. John
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Sarah A. Shaw
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Kathleen J. Krentz
- Genome Editing and Animal Models Core, Biotechnology Center, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - Xiaoyun Zhang
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Chi-chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Dagmar Wilhelm
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kuroki S, Tachibana M. Epigenetic regulation of mammalian sex determination. Mol Cell Endocrinol 2018; 468:31-38. [PMID: 29248548 DOI: 10.1016/j.mce.2017.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 11/22/2022]
Abstract
Regulation of gene expression without changing the DNA sequence is governed by epigenetic mechanisms. Epigenetic regulation is important for changing chromatin structure in response to environmental cues as well as maintaining chromatin structure after cell division. The epigenetic machinery can reversibly change chromatin function, allowing a wide variety of biological processes in multicellular organisms to be controlled. Epigenetic regulation ensures spatial and temporal accuracy of the expression of developmentally regulated genes. So far, few studies have focused on the relationship between epigenetic regulation and mammalian sex development, despite this being an interesting area of research. Sex development consists of three sequential stages: the undifferentiated stage, gonadal differentiation into testes or ovaries, and differentiation of internal and external genitalia. Some genetic studies have revealed that epigenetic regulation is required for proper gonadal differentiation in mice. Particularly, the epigenetic machinery plays an integral part in sex determination, which is the first step of gonadal differentiation. Mammalian sex determination is triggered by activation of the mammalian sex-determining gene, Sry, in a spatially and temporally accurate manner. Several studies have demonstrated that expression of Sry is controlled not only by specific transcription factors but also by the epigenetic machinery. Here, we focus on the epigenetic regulation of Sry expression.
Collapse
Affiliation(s)
- Shunsuke Kuroki
- Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima 770-8503, Japan
| | - Makoto Tachibana
- Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima 770-8503, Japan.
| |
Collapse
|
32
|
Barseghyan H, Symon A, Zadikyan M, Almalvez M, Segura EE, Eskin A, Bramble MS, Arboleda VA, Baxter R, Nelson SF, Délot EC, Harley V, Vilain E. Identification of novel candidate genes for 46,XY disorders of sex development (DSD) using a C57BL/6J-Y POS mouse model. Biol Sex Differ 2018; 9:8. [PMID: 29378665 PMCID: PMC5789682 DOI: 10.1186/s13293-018-0167-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/19/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Disorders of sex development (DSD) have an estimated frequency of 0.5% of live births encompassing a variety of urogenital anomalies ranging from mild hypospadias to a discrepancy between sex chromosomes and external genitalia. In order to identify the underlying genetic etiology, we had performed exome sequencing in a subset of DSD cases with 46,XY karyotype and were able to identify the causative genetic variant in 35% of cases. While the genetic etiology was not ascertained in more than half of the cases, a large number of variants of unknown clinical significance (VUS) were identified in those exomes. METHODS To investigate the relevance of these VUS in regards to the patient's phenotype, we utilized a mouse model in which the presence of a Y chromosome from the poschiavinus strain (Y POS ) on a C57BL/6J (B6) background results in XY undervirilization and sex reversal, a phenotype characteristic to a large subset of human 46,XY DSD cases. We assessed gene expression differences between B6-Y B6 and undervirilized B6-Y POS gonads at E11.5 and identified 515 differentially expressed genes (308 underexpressed and 207 overexpressed in B6-Y POS males). RESULTS We identified 15 novel candidate genes potentially involved in 46,XY DSD pathogenesis by filtering the list of human VUS-carrying genes provided by exome sequencing with the list of differentially expressed genes from B6-Y POS mouse model. Additionally, we identified that 7 of the 15 candidate genes were significantly underexpressed in the XY gonads of mice with suppressed Sox9 expression in Sertoli cells suggesting that some of the candidate genes may be downstream of a well-known sex determining gene, Sox9. CONCLUSION The use of a DSD-specific animal model improves variant interpretation by correlating human sequence variants with transcriptome variation.
Collapse
Affiliation(s)
- Hayk Barseghyan
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Health System, Washington, DC, 20010 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Aleisha Symon
- Department of Brain and Gender, Hudson Institute of Medical Research, Clayton, VIC 3168 Australia
| | - Mariam Zadikyan
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Miguel Almalvez
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Health System, Washington, DC, 20010 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Eva E. Segura
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Ascia Eskin
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Matthew S. Bramble
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Health System, Washington, DC, 20010 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Valerie A. Arboleda
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Ruth Baxter
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Stanley F. Nelson
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Emmanuèle C. Délot
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Health System, Washington, DC, 20010 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| | - Vincent Harley
- Department of Brain and Gender, Hudson Institute of Medical Research, Clayton, VIC 3168 Australia
| | - Eric Vilain
- Center for Genetic Medicine Research, Children’s Research Institute, Children’s National Health System, Washington, DC, 20010 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
33
|
Carré GA, Siggers P, Xipolita M, Brindle P, Lutz B, Wells S, Greenfield A. Loss of p300 and CBP disrupts histone acetylation at the mouse Sry promoter and causes XY gonadal sex reversal. Hum Mol Genet 2018; 27:190-198. [PMID: 29145650 PMCID: PMC5886154 DOI: 10.1093/hmg/ddx398] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 01/08/2023] Open
Abstract
CREB-binding protein (CBP, CREBBP, KAT3A) and its closely related paralogue p300 (EP300, KAT3B), together termed p300/CBP, are histone/lysine acetyl-transferases that control gene expression by modifying chromatin-associated proteins. Here, we report roles for both of these chromatin-modifying enzymes in mouse sex determination, the process by which the embryonic gonad develops into a testis or an ovary. By targeting gene ablation to embryonic gonadal somatic cells using an inducible Cre line, we show that gonads lacking either gene exhibit major abnormalities of XY gonad development at 14.5 dpc, including partial sex reversal. Embryos lacking three out of four functional copies of p300/Cbp exhibit complete XY gonadal sex reversal and have greatly reduced expression of the key testis-determining genes Sry and Sox9. An analysis of histone acetylation at the Sry promoter in mutant gonads at 11.5 dpc shows a reduction in levels of the positive histone mark H3K27Ac. Our data suggest a role for CBP/p300 in testis determination mediated by control of histone acetylation at the Sry locus and reveal a novel element in the epigenetic control of Sry and mammalian sex determination. They also suggest possible novel causes of human disorders of sex development (DSD).
Collapse
Affiliation(s)
- Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Marilena Xipolita
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Paul Brindle
- Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center Mainz, 55128 Mainz, Germany
| | - Sara Wells
- Mary Lyon Centre, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell Institute, Oxfordshire OX11 0RD, UK
| |
Collapse
|
34
|
Bagheri-Fam S, Bird AD, Zhao L, Ryan JM, Yong M, Wilhelm D, Koopman P, Eswarakumar VP, Harley VR. Testis Determination Requires a Specific FGFR2 Isoform to Repress FOXL2. Endocrinology 2017; 158:3832-3843. [PMID: 28938467 PMCID: PMC5695826 DOI: 10.1210/en.2017-00674] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/05/2017] [Indexed: 02/03/2023]
Abstract
Male sex determination in mammals relies on sex determining region Y-mediated upregulation of sex determining region-box 9 (SOX9) expression in XY gonads, whereas Wnt family member (WNT)/R-spondin 1 signaling and forkhead box L2 (FOXL2) drive female sex determination in XX gonads. Fibroblast growth factor (FGF) 9 signaling ensures sustained SOX9 expression through repression of one of the ovarian pathways (WNT signaling), whereas the significance of FGF-mediated repression of the FOXL2 pathway has not been studied. Previously, we demonstrated that FGFR2 is the receptor for FGF9 in the XY gonad. Whether a specific isoform (FGFR2b or FGFR2c) is required was puzzling. Here, we show that FGFR2c is required for male sex determination. Initially, in developing mouse embryos at 12.5 to 13.5 days postcoitum (dpc), XY Fgfr2c-/- gonads appear as ovotestes, with SOX9 and FOXL2 expression predominantly localized to the posterior and anterior gonadal poles, respectively. However, by 15.5 dpc, XY Fgfr2c-/- gonads show complete male-to-female sex reversal, evident by the lack of SOX9 and ectopic expression of FOXL2 throughout the gonads. Furthermore, ablation of the Foxl2 gene leads to partial or complete rescue of gonadal sex reversal in XY Fgfr2c-/- mice. Together with previous findings, our data suggest that testis determination involves FGFR2c-mediated repression of both the WNT4- and FOXL2-driven ovarian-determining pathways.
Collapse
Affiliation(s)
- Stefan Bagheri-Fam
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Victoria 3168, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Anthony D. Bird
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Victoria 3168, Australia
| | - Liang Zhao
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Janelle M. Ryan
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Victoria 3168, Australia
| | - Meiyun Yong
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Victoria 3168, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Dagmar Wilhelm
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Veraragavan P. Eswarakumar
- Department of Orthopaedics and Rehabilitation, Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Vincent R. Harley
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Monash Medical Centre, Melbourne, Victoria 3168, Australia
| |
Collapse
|
35
|
Johansson HKL, Svingen T, Fowler PA, Vinggaard AM, Boberg J. Environmental influences on ovarian dysgenesis - developmental windows sensitive to chemical exposures. Nat Rev Endocrinol 2017; 13:400-414. [PMID: 28450750 DOI: 10.1038/nrendo.2017.36] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A woman's reproductive health and ability to have children directly affect numerous aspects of her life, from personal well-being and socioeconomic standing, to morbidity and lifespan. In turn, reproductive health depends on the development of correctly functioning ovaries, a process that starts early during fetal life. Early disruption to ovarian programming can have long-lasting consequences, potentially manifesting as disease much later in adulthood. A growing body of evidence suggests that exposure to chemicals early in life, including endocrine-disrupting chemicals, can cause a range of disorders later in life, such as those described in the ovarian dysgenesis syndrome hypothesis. In this Review, we discuss four specific time windows during which the ovary is particularly sensitive to disruption by exogenous insults: gonadal sex determination, meiotic division, follicle assembly and the first wave of follicle recruitment. To date, most evidence points towards the germ cell lineage being the most vulnerable to chemical exposure, particularly meiotic division and follicle assembly. Environmental chemicals and pharmaceuticals, such as bisphenols or mild analgesics (including paracetamol), can also affect the somatic cell lineages. This Review summarizes our current knowledge pertaining to environmental chemicals and pharmaceuticals, and their potential contributions to the development of ovarian dysgenesis syndrome. We also highlight knowledge gaps that need addressing to safeguard female reproductive health.
Collapse
Affiliation(s)
- Hanna Katarina Lilith Johansson
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Anne Marie Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Julie Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
36
|
Zhao L, Arsenault M, Ng ET, Longmuss E, Chau TCY, Hartwig S, Koopman P. SOX4 regulates gonad morphogenesis and promotes male germ cell differentiation in mice. Dev Biol 2017; 423:46-56. [PMID: 28118982 DOI: 10.1016/j.ydbio.2017.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
The group C SOX transcription factors SOX4, -11 and -12 play important and mutually overlapping roles in development of a number of organs. Here, we examined the role of SoxC genes during gonadal development in mice. All three genes were expressed in developing gonads of both sexes, predominantly in somatic cells, with Sox4 being most strongly expressed. Sox4 deficiency resulted in elongation of both ovaries and testes, and an increased number of testis cords. While female germ cells entered meiosis normally, male germ cells showed reduced levels of differentiation markers Nanos2 and Dnmt3l and increased levels of pluripotency genes Cripto and Nanog, suggesting that SOX4 may normally act to restrict the pluripotency period of male germ cells and ensure their proper differentiation. Finally, our data reveal that SOX4 (and, to a lesser extent, SOX11 and -12) repressed transcription of the sex-determining gene Sox9 via an upstream testis-specific enhancer core (TESCO) element in fetal gonads, raising the possibility that SOXC proteins may function as transcriptional repressors in a context-dependent manner.
Collapse
Affiliation(s)
- Liang Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Michel Arsenault
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island,550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Ee Ting Ng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Enya Longmuss
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Tevin Chui-Ying Chau
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sunny Hartwig
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island,550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
37
|
Gonen N, Quinn A, O’Neill HC, Koopman P, Lovell-Badge R. Normal Levels of Sox9 Expression in the Developing Mouse Testis Depend on the TES/TESCO Enhancer, but This Does Not Act Alone. PLoS Genet 2017; 13:e1006520. [PMID: 28045957 PMCID: PMC5207396 DOI: 10.1371/journal.pgen.1006520] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/02/2016] [Indexed: 01/18/2023] Open
Abstract
During mouse sex determination, transient expression of the Y-linked gene Sry up-regulates its direct target gene Sox9, via a 3.2 kb testis specific enhancer of Sox9 (TES), which includes a core 1.4 kb element, TESCO. SOX9 activity leads to differentiation of Sertoli cells, rather than granulosa cells from the bipotential supporting cell precursor lineage. Here, we present functional analysis of TES/TESCO, using CRISPR/Cas9 genome editing in mice. Deletion of TESCO or TES reduced Sox9 expression levels in XY fetal gonads to 60 or 45% respectively relative to wild type gonads, and reduced expression of the SOX9 target Amh. Although human patients heterozygous for null mutations in SOX9, which are assumed to have 50% of normal expression, often show XY female sex reversal, mice deleted for one copy of Sox9 do not. Consistent with this, we did not observe sex reversal in either TESCO-/- or TES-/- XY embryos or adult mice. However, embryos carrying both a conditional Sox9 null allele and the TES deletion developed ovotestes. Quantitative analysis of these revealed levels of 23% expression of Sox9 compared to wild type, and a significant increase in the expression of the granulosa cell marker Foxl2. This indicates that the threshold in mice where sex reversal begins to be seen is about half that of the ~50% levels predicted in humans. Our results demonstrate that TES/TESCO is a crucial enhancer regulating Sox9 expression in the gonad, but point to the existence of additional enhancers that act redundantly.
Collapse
Affiliation(s)
- Nitzan Gonen
- The Francis Crick Institute, Midland Road, London, United Kingdom
| | - Alexander Quinn
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Helen C. O’Neill
- The Francis Crick Institute, Midland Road, London, United Kingdom
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | |
Collapse
|
38
|
Pannetier M, Chassot AA, Chaboissier MC, Pailhoux E. Involvement of FOXL2 and RSPO1 in Ovarian Determination, Development, and Maintenance in Mammals. Sex Dev 2016; 10:167-184. [PMID: 27649556 DOI: 10.1159/000448667] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Indexed: 11/19/2022] Open
Abstract
In mammals, sex determination is a process through which the gonad is committed to differentiate into a testis or an ovary. This process relies on a delicate balance between genetic pathways that promote one fate and inhibit the other. Once the gonad is committed to the female pathway, ovarian differentiation begins and, depending on the species, is completed during gestation or shortly after birth. During this step, granulosa cell precursors, steroidogenic cells, and primordial germ cells start to express female-specific markers in a sex-dimorphic manner. The germ cells then arrest at prophase I of meiosis and, together with somatic cells, assemble into functional structures. This organization gives the ovary its definitive morphology and functionality during folliculogenesis. Until now, 2 main genetic cascades have been shown to be involved in female sex differentiation. The first is driven by FOXL2, a transcription factor that also plays a crucial role in folliculogenesis and ovarian fate maintenance in adults. The other operates through the WNT/CTNNB1 canonical pathway and is regulated primarily by R-spondin1. Here, we discuss the roles of FOXL2 and RSPO1/WNT/ CTNNB1 during ovarian development and homeostasis in different models, such as humans, goats, and rodents.
Collapse
Affiliation(s)
- Maëlle Pannetier
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | | | | | | |
Collapse
|
39
|
Racca JD, Chen YS, Yang Y, Phillips NB, Weiss MA. Human Sex Determination at the Edge of Ambiguity: INHERITED XY SEX REVERSAL DUE TO ENHANCED UBIQUITINATION AND PROTEASOMAL DEGRADATION OF A MASTER TRANSCRIPTION FACTOR. J Biol Chem 2016; 291:22173-22195. [PMID: 27576690 DOI: 10.1074/jbc.m116.741959] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Indexed: 12/15/2022] Open
Abstract
A general problem is posed by analysis of transcriptional thresholds governing cell fate decisions in metazoan development. A model is provided by testis determination in therian mammals. Its key step, Sertoli cell differentiation in the embryonic gonadal ridge, is initiated by SRY, a Y-encoded architectural transcription factor. Mutations in human SRY cause gonadal dysgenesis leading to XY female development (Swyer syndrome). Here, we have characterized an inherited mutation compatible with either male or female somatic phenotypes as observed in an XY father and XY daughter, respectively. The mutation (a crevice-forming substitution at a conserved back surface of the SRY high mobility group box) markedly destabilizes the domain but preserves specific DNA affinity and induced DNA bend angle. On transient transfection of diverse human and rodent cell lines, the variant SRY exhibited accelerated proteasomal degradation (relative to wild type) associated with increased ubiquitination; in vitro susceptibility to ubiquitin-independent ("default") cleavage by the 20S core proteasome was unchanged. The variant's gene regulatory activity (as assessed in a cellular model of the rat embryonic XY gonadal ridge) was reduced by 2-fold relative to wild-type SRY at similar levels of mRNA expression. Chemical proteasome inhibition restored native-like SRY expression and transcriptional activity in association with restored occupancy of a sex-specific enhancer element in principal downstream gene Sox9, demonstrating that the variant SRY exhibits essentially native activity on a per molecule basis. Our findings define a novel mechanism of impaired organogenesis, accelerated ubiquitin-directed proteasomal degradation of a master transcription factor leading to a developmental decision poised at the edge of ambiguity.
Collapse
Affiliation(s)
- Joseph D Racca
- From the Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yen-Shan Chen
- From the Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yanwu Yang
- From the Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nelson B Phillips
- From the Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- From the Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
40
|
Baetens D, Stoop H, Peelman F, Todeschini AL, Rosseel T, Coppieters F, Veitia RA, Looijenga LHJ, De Baere E, Cools M. NR5A1 is a novel disease gene for 46,XX testicular and ovotesticular disorders of sex development. Genet Med 2016; 19:367-376. [PMID: 27490115 PMCID: PMC5392598 DOI: 10.1038/gim.2016.118] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/11/2016] [Indexed: 01/10/2023] Open
Abstract
PURPOSE We aimed to identify the genetic cause in a cohort of 11 unrelated cases and two sisters with 46,XX SRY-negative (ovo)testicular disorders of sex development (DSD). METHODS Whole-exome sequencing (n = 9), targeted resequencing (n = 4), and haplotyping were performed. Immunohistochemistry of sex-specific markers was performed on patients' gonads. The consequences of mutation were investigated using luciferase assays, localization studies, and RNA-seq. RESULTS We identified a novel heterozygous NR5A1 mutation, c.274C>T p.(Arg92Trp), in three unrelated patients. The Arg92 residue is highly conserved and located in the Ftz-F1 region, probably involved in DNA-binding specificity and stability. There were no consistent changes in transcriptional activation or subcellular localization. Transcriptomics in patient-derived lymphocytes showed upregulation of MAMLD1, a direct NR5A1 target previously associated with 46,XY DSD. In gonads of affected individuals, ovarian FOXL2 and testicular SRY-independent SOX9 expression observed. CONCLUSIONS We propose NR5A1, previously associated with 46,XY DSD and 46,XX primary ovarian insufficiency, as a novel gene for 46,XX (ovo)testicular DSD. We hypothesize that p.(Arg92Trp) results in decreased inhibition of the male developmental pathway through downregulation of female antitestis genes, thereby tipping the balance toward testicular differentiation in 46,XX individuals. In conclusion, our study supports a role for NR5A1 in testis differentiation in the XX gonad.Genet Med 19 4, 367-376.
Collapse
Affiliation(s)
- Dorien Baetens
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Hans Stoop
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank Peelman
- Flanders Institute for Biotechnology (VIB), Department of Medical Protein Research, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Anne-Laure Todeschini
- Molecular Oncology and Pathology, Institut Jacques Monod, France; Université Paris Diderot, Paris VII, France
| | - Toon Rosseel
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Frauke Coppieters
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Reiner A Veitia
- Molecular Oncology and Pathology, Institut Jacques Monod, France; Université Paris Diderot, Paris VII, France
| | - Leendert H J Looijenga
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Martine Cools
- Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Ghent, Belgium
| |
Collapse
|
41
|
Warr N, Siggers P, Carré GA, Wells S, Greenfield A. Genetic Analyses Reveal Functions for MAP2K3 and MAP2K6 in Mouse Testis Determination. Biol Reprod 2016; 94:103. [PMID: 27009039 PMCID: PMC5842889 DOI: 10.1095/biolreprod.115.138057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/08/2016] [Indexed: 01/27/2023] Open
Abstract
Testis determination in mammals is initiated by expression of SRY in somatic cells of the embryonic gonad. Genetic analyses in the mouse have revealed a requirement for mitogen-activated protein kinase (MAPK) signaling in testis determination: targeted loss of the kinases MAP3K4 and p38 MAPK causes complete XY embryonic gonadal sex reversal. These kinases occupy positions at the top and bottom level, respectively, in the canonical threetier MAPK-signaling cascade: MAP3K, MAP2K, MAPK. To date, no role in sex determination has been attributed to a MAP2K, although such a function is predicted to exist. Here, we report roles for the kinases MAP2K3 and MAP2K6 in testis determination. C57BL/6J (B6) embryos lacking MAP2K3 exhibited no significant abnormalities of testis development, whilst those lacking MAP2K6 exhibited a minor delay in testis determination. Compound mutants lacking three out of four functional alleles at the two loci also exhibited delayed testis determination and transient ovotestis formation as a consequence, suggestive of partially redundant roles for these kinases in testis determination. Early lethality of double-knockout embryos precludes analysis of sexual development. To reveal their roles in testis determination more clearly, we generated Map2k mutant B6 embryos using a weaker Sry allele (SryAKR). Loss of Map2k3 on this highly sensitized background exacerbates ovotestis development, whilst loss of Map2k6 results in complete XY gonadal sex reversal associated with reduction of Sry expression at 11.25 days postcoitum. Our data suggest that MAP2K6 functions in mouse testis determination, via positive effects on Sry, and also indicate a minor role for MAP2K3.
Collapse
Affiliation(s)
- Nick Warr
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Sara Wells
- The Mary Lyon Centre, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| |
Collapse
|
42
|
Abstract
Current knowledge on gonadal development and sex determination is the product of many decades of research involving a variety of scientific methods from different biological disciplines such as histology, genetics, biochemistry, and molecular biology. The earliest embryological investigations, followed by the invention of microscopy and staining methods, were based on histological examinations. The most robust development of histological staining techniques occurred in the second half of the nineteenth century and resulted in structural descriptions of gonadogenesis. These first studies on gonadal development were conducted on domesticated animals; however, currently the mouse is the most extensively studied species. The next key point in the study of gonadogenesis was the advancement of methods allowing for the in vitro culture of fetal gonads. For instance, this led to the description of the origin of cell lines forming the gonads. Protein detection using antibodies and immunolabeling methods and the use of reporter genes were also invaluable for developmental studies, enabling the visualization of the formation of gonadal structure. Recently, genetic and molecular biology techniques, especially gene expression analysis, have revolutionized studies on gonadogenesis and have provided insight into the molecular mechanisms that govern this process. The successive invention of new methods is reflected in the progress of research on gonadal development.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| |
Collapse
|
43
|
Carré GA, Greenfield A. The Gonadal Supporting Cell Lineage and Mammalian Sex Determination: The Differentiation of Sertoli and Granulosa Cells. Results Probl Cell Differ 2016; 58:47-66. [PMID: 27300175 DOI: 10.1007/978-3-319-31973-5_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The supporting cell lineage plays a crucial role in nurturing the development of germ cells in the adult gonad. Sertoli cells in the testis support the progression of spermatogonial stem cells through meiosis to the production of motile spermatozoa. Granulosa cells, meanwhile, are a critical component of the ovarian follicle that produces the mature oocyte. It is a distinctive feature of the embryonic gonad that at least some of the supporting cells are derived from a single sexually bipotential precursor lineage. It is the commitment of this somatic lineage to either the Sertoli or granulosa cell fate that defines sex determination. In this chapter we review what is known about the key molecules responsible for this lineage decision in the developing mammalian gonads, relying primarily on data from studies of mice and humans. We focus on recent advances in our understanding of the mutually antagonistic interactions of testis- and ovary-determining pathways and their complexity as revealed by genetic analyses. For the sake of simplicity, we will deal with supporting cells in testis and ovary development in separate sections, but numerous points of contact exist between these accounts of gonadogenesis in male and female embryos, primarily due to the aforementioned mutual antagonisms. The final section will offer a brief synthesis of these organ-specific overviews and a summary of the key themes that emerge in this review of supporting cell differentiation in mammalian sex determination.
Collapse
Affiliation(s)
- Gwenn-Aël Carré
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, OX11 0RD, UK
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
44
|
Understanding sex determination in the mouse: genetics, epigenetics and the story of mutual antagonisms. J Genet 2015; 94:585-90. [DOI: 10.1007/s12041-015-0565-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
45
|
|
46
|
Lin YT, Capel B. Cell fate commitment during mammalian sex determination. Curr Opin Genet Dev 2015; 32:144-52. [PMID: 25841206 DOI: 10.1016/j.gde.2015.03.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/24/2015] [Accepted: 03/05/2015] [Indexed: 01/10/2023]
Abstract
The gonads form bilaterally as bipotential organs that can develop as testes or ovaries. All secondary sex characteristics that we associate with 'maleness' or 'femaleness' depend on whether testes or ovaries form. The fate of the gonads depends on a cell fate decision that occurs in a somatic cell referred to as the 'supporting cell lineage'. Once supporting cell progenitors commit to Sertoli (male) or granulosa (female) fate, they propagate this decision to the other cells within the organ. In this review, we will describe what is known about the bipotential state of somatic and germ cell lineages in the gonad and the transcriptional and antagonistic signaling networks that lead to commitment, propagation, and maintenance of testis or ovary fate.
Collapse
Affiliation(s)
- Yi-Tzu Lin
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
47
|
Wainwright EN, Svingen T, Ng ET, Wicking C, Koopman P. Primary cilia function regulates the length of the embryonic trunk axis and urogenital field in mice. Dev Biol 2014; 395:342-54. [DOI: 10.1016/j.ydbio.2014.08.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/20/2014] [Accepted: 08/27/2014] [Indexed: 01/06/2023]
|
48
|
Rastetter RH, Bernard P, Palmer JS, Chassot AA, Chen H, Western PS, Ramsay RG, Chaboissier MC, Wilhelm D. Marker genes identify three somatic cell types in the fetal mouse ovary. Dev Biol 2014; 394:242-52. [DOI: 10.1016/j.ydbio.2014.08.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 12/28/2022]
|
49
|
Regulation of male sex determination: genital ridge formation and Sry activation in mice. Cell Mol Life Sci 2014; 71:4781-802. [PMID: 25139092 PMCID: PMC4233110 DOI: 10.1007/s00018-014-1703-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 11/27/2022]
Abstract
Sex determination is essential for the sexual reproduction to generate the next generation by the formation of functional male or female gametes. In mammals, primary sex determination is commenced by the presence or absence of the Y chromosome, which controls the fate of the gonadal primordium. The somatic precursor of gonads, the genital ridge is formed at the mid-gestation stage and gives rise to one of two organs, a testis or an ovary. The fate of the genital ridge, which is governed by the differentiation of somatic cells into Sertoli cells in the testes or granulosa cells in the ovaries, further determines the sex of an individual and their germ cells. Mutation studies in human patients with disorders of sex development and mouse models have revealed factors that are involved in mammalian sex determination. In most of mammals, a single genetic trigger, the Y-linked gene Sry (sex determination region on Y chromosome), regulates testicular differentiation. Despite identification of Sry in 1990, precise mechanisms underlying the sex determination of bipotential genital ridges are still largely unknown. Here, we review the recent progress that has provided new insights into the mechanisms underlying genital ridge formation as well as the regulation of Sry expression and its functions in male sex determination of mice.
Collapse
|
50
|
Larney C, Bailey TL, Koopman P. Switching on sex: transcriptional regulation of the testis-determining gene Sry. Development 2014; 141:2195-205. [PMID: 24866114 DOI: 10.1242/dev.107052] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mammalian sex determination hinges on the development of ovaries or testes, with testis fate being triggered by the expression of the transcription factor sex-determining region Y (Sry). Reduced or delayed Sry expression impairs testis development, highlighting the importance of its accurate spatiotemporal regulation and implying a potential role for SRY dysregulation in human intersex disorders. Several epigenetic modifiers, transcription factors and kinases are implicated in regulating Sry transcription, but it remains unclear whether or how this farrago of factors acts co-ordinately. Here we review our current understanding of Sry regulation and provide a model that assembles all known regulators into three modules, each converging on a single transcription factor that binds to the Sry promoter. We also discuss potential future avenues for discovering the cis-elements and trans-factors required for Sry regulation.
Collapse
Affiliation(s)
- Christian Larney
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Timothy L Bailey
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|