Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 446
Filter
1.
Med Sci (Paris) ; 40(8-9): 627-633, 2024.
Article in French | MEDLINE | ID: mdl-39303114

ABSTRACT

The sex of an individual is determined at the time of fertilization. The mother passes on one sex chromosome, the X chromosome, and the father transmits the second sex chromosome, X or Y. Thus, an XX embryo becomes a female, whereas an XY individual becomes a male. A process known as "primary sex determination" allows the bipotential gonad to become a testis or an ovary in XY and XX embryos, respectively. In 1990, the Sry gene, located on the Y chromosome, was found to be necessary and sufficient to induce the male developmental program. At this time, the scientific community thought that other genes involved in the process of sex determination would be rapidly identified. However, it took more than 30 years to identify the ovarian determining factor. This factor is one variant of WT1, denoted -KTS, which is required to induce ovarian development in XX mice and can prevent male development of the gonad when it is prematurely activated in XY embryos. Because the -KTS variant of WT1 acts very early during development, this discovery opens new avenues for research on ovarian development, as it happened for SRY for testis development. It will also lead to a better understanding of the regulatory gene networks implicated in many unresolved cases of sex development disorders.


Title: Détermination du sexe - Chaque chose en son temps ! Abstract: Le sexe de l'embryon est décidé au moment de la fécondation par la transmission paternelle du chromosome sexuel X ou Y, tandis que la mère fournit un de ses deux chromosomes X. La différenciation sexuelle débute par le processus de détermination du sexe, qui va permettre le développement de l'ébauche gonadique soit en testicule, chez l'embryon XY, soit en ovaire, chez l'embryon XX. Le gène Sry, localisé sur le chromosome Y, nécessaire et suffisant pour induire le programme de développement masculin, a été découvert en 1990, et la communauté scientifique pensait alors que les autres gènes impliqués dans le processus de détermination du sexe seraient rapidement identifiés. Il aura cependant fallu plus de 30 ans pour identifier le facteur déterminant la différenciation ovarienne, une isoforme de WT1 appelée -KTS. Cette protéine est nécessaire pour induire le développement de l'ovaire chez les souris XX, et peut empêcher le développement masculin lorsqu'elle est activée prématurément chez les embryons XY. L'isoforme -KTS de WT1 agissant très tôt au cours du développement, sa découverte ouvre de nouvelles perspectives de recherche sur le développement ovarien et permettra de mieux comprendre les réseaux de gènes impliqués dans certaines altérations du développement du sexe.


Subject(s)
Sex Determination Processes , Sex Determination Processes/physiology , Sex Determination Processes/genetics , Female , Male , Animals , Humans , Time Factors , Mice , Disorders of Sex Development/genetics , Ovary/physiology , Sex-Determining Region Y Protein/genetics
2.
J Biol Chem ; 300(9): 107683, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39168182

ABSTRACT

Y-chromosome-encoded master transcription factor SRY functions in the embryogenesis of therian mammals to initiate male development. Through interactions of its conserved high-mobility group box within a widened DNA minor groove, SRY and related Sox factors induce sharp bends at specific DNA target sites. Here, we present the crystal structure of the SRY high-mobility group domain bound to a DNA site containing consensus element 5'-ATTGTT. The structure contains three complexes in the asymmetric unit; in each complex, SRY forms 10 hydrogen bonds with minor-groove base atoms in 5'-CATTGT/ACAATG-3', shifting the recognition sequence by one base pair (italics). These nucleobase interactions involve conserved residues Arg7, Asn10, and Tyr74 on one side of intercalated Ile13 (the cantilever) and Arg20, Asn32, and Ser36 on the other. Unlike the less-bent NMR structure, DNA bend angles (69-84°) of the distinct box-DNA complexes are similar to those observed in homologous Sox domain-DNA structures. Electrophoretic studies indicate that respective substitutions of Asn32, Ser36, or Tyr74 by Ala exhibit slightly attenuated specific DNA-binding affinity and bend angles (70-73°) relative to WT (79°). By contrast, respective substitutions of Arg7, Asn10, or Arg20 by Ala markedly impaired DNA-binding affinity in association with much smaller DNA bend angles (53-65°). In a rodent cell-based model of the embryonic gonadal ridge, full-length SRY variants bearing these respective Ala substitutions exhibited significantly decreased transcriptional activation of SRY's principal target gene (Sox9). Together, our findings suggest that nucleobase-specific hydrogen bonds by SRY are critical for specific DNA binding, bending, and transcriptional activation.


Subject(s)
DNA , Sex-Determining Region Y Protein , Humans , Sex-Determining Region Y Protein/metabolism , Sex-Determining Region Y Protein/chemistry , Sex-Determining Region Y Protein/genetics , DNA/metabolism , DNA/chemistry , Male , Crystallography, X-Ray , Protein Binding , Animals , Nucleic Acid Conformation , Hydrogen Bonding
3.
Zhonghua Nan Ke Xue ; 30(2): 118-122, 2024 Feb.
Article in Chinese | MEDLINE | ID: mdl-39177343

ABSTRACT

OBJECTIVE: To investigate the genetic and clinical characteristics of 46, XX testicular disorders of sex development (DSD). METHODS: We collected the clinical data on the patients with 46,XX testicular DSD diagnosed in the Center of Reproductive Medicine of the First Affiliated Hospital of Nanjing Medical University from January 2017 to January 2023, and analyzed their genetic and clinical characteristics and the SRY gene chromosomal location for those with SRY-positive. RESULTS: A total of 26 patients were included in this study, all with 46,XX and deletion of the AZFa, b and c regions, with a mean height of (168.3±5.9) cm, body weight of (64.0±7.5) kg, BMI of (22.66±2.79) kg/m2, left testis volume of (2.53±1.16) ml and right testis volume of (2.74±1.34) ml. The semen volume of the patients averaged 1.35 (0.18-2.78) ml, FSH (36.85±18.01) IU/L, LH (19.71±9.71) IU/L, and T (6.08±2.71) nmol/L. The SRY-negative patients had a higher incidence rate of development disorders in the reproductive system than the SRY-positive ones (5/6 vs 3/20, P = 0.004), but no statistically significant differences were observed in the other parameters. The SRY gene was localized at the end of Xp in 13 of the 14 SRY-positive cases, and at chromosome 15 in the other 1. CONCLUSION: 46,XX testicular DSD has some similarity and heterogeneity in genetics and clinical characteristics.


Subject(s)
Testis , Humans , Male , 46, XX Testicular Disorders of Sex Development/genetics , Sex-Determining Region Y Protein/genetics , Disorders of Sex Development/genetics , Karyotyping , Adult , Chromosome Deletion
4.
J Pediatr Endocrinol Metab ; 37(8): 750-755, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38973169

ABSTRACT

OBJECTIVES: 45,X/46,XY mosaicism is a rare condition with clinical and genetic heterogeneity and have a greatly increased risk of developing germ cell tumors. We describe a rare 45,X/46,XY Chinese girl with malignant tumors, especially focusing on the molecular genetics of gonadal tumor. CASE PRESENTATION: We report a phenotypically Turner-like Chinese adolescent girl who presented primary amenorrhea and a pelvic mass as the chief complaint, which finally demonstrated dysgerminoma replacing the left gonad and gonadoblastoma arising from right gonad respectively. Her chromosome karyotype was 45,X(4)/46,XY(46); Y-chromosome microdeletions in AZFb regions were found on gonadal DNA rather than peripheral blood lymphocyte (PBL) DNA, while no variants were found in the promoter and coding region of SRY gene in both PBL and gonadal tissues. She underwent bilateral gonadectomy; no recurrence or serious complications were identified after 3 years of follow-up. CONCLUSIONS: This case emphasizes the probable correlation between Y chromosome microdeletions in gonadal tissue and the severity of the phenotype in patients with 45,X/46,XY mosaicism and highlights the importance of clinical genetic testing at the chromosomal and molecular level.


Subject(s)
Dysgerminoma , Gonadoblastoma , Ovarian Neoplasms , Phenotype , Turner Syndrome , Humans , Female , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Gonadoblastoma/surgery , Dysgerminoma/genetics , Dysgerminoma/pathology , Dysgerminoma/surgery , Adolescent , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Turner Syndrome/genetics , Turner Syndrome/pathology , Turner Syndrome/complications , Mosaicism , Prognosis , Karyotype , Sex-Determining Region Y Protein/genetics , East Asian People
5.
Nat Commun ; 15(1): 3809, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714644

ABSTRACT

Mammalian sex determination is controlled by antagonistic gene cascades operating in embryonic undifferentiated gonads. The expression of the Y-linked gene SRY is sufficient to trigger the testicular pathway, whereas its absence in XX embryos leads to ovarian differentiation. Yet, the potential involvement of non-coding regulation in this process remains unclear. Here we show that the deletion of a single microRNA cluster, miR-17~92, induces complete primary male-to-female sex reversal in XY mice. Sry expression is delayed in XY knockout gonads, which develop as ovaries. Sertoli cell differentiation is reduced, delayed and unable to sustain testicular development. Pre-supporting cells in mutant gonads undergo a transient state of sex ambiguity which is subsequently resolved towards the ovarian fate. The miR-17~92 predicted target genes are upregulated, affecting the fine regulation of gene networks controlling gonad development. Thus, microRNAs emerge as key components for mammalian sex determination, controlling Sry expression timing and Sertoli cell differentiation.


Subject(s)
Cell Differentiation , MicroRNAs , Ovary , Sertoli Cells , Sex Determination Processes , Sex-Determining Region Y Protein , Testis , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Male , Sertoli Cells/metabolism , Sertoli Cells/cytology , Mice , Ovary/metabolism , Testis/metabolism , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Cell Differentiation/genetics , Sex Determination Processes/genetics , Gene Expression Regulation, Developmental , Mice, Knockout , Sex Differentiation/genetics , Disorders of Sex Development/genetics , Gonads/metabolism
6.
Nucleic Acids Res ; 52(12): 6763-6776, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38721783

ABSTRACT

The kinetics of protein-DNA recognition, along with its thermodynamic properties, including affinity and specificity, play a central role in shaping biological function. Protein-DNA recognition kinetics are characterized by two key elements: the time taken to locate the target site amid various nonspecific alternatives; and the kinetics involved in the recognition process, which may necessitate overcoming an energetic barrier. In this study, we developed a coarse-grained (CG) model to investigate interactions between a transcription factor called the sex-determining region Y (SRY) protein and DNA, in order to probe how DNA conformational changes affect SRY-DNA recognition and binding kinetics. We find that, not only does a requirement for such a conformational DNA transition correspond to a higher energetic barrier for binding and therefore slower kinetics, it may further impede the recognition kinetics by increasing unsuccessful binding events (skipping events) where the protein partially binds its DNA target site but fails to form the specific protein-DNA complex. Such skipping events impose the need for additional cycles protein search of nonspecific DNA sites, thus significantly extending the overall recognition time. Our results highlight a trade-off between the speed with which the protein scans nonspecific DNA and the rate at which the protein recognizes its specific target site. Finally, we examine molecular approaches potentially adopted by natural systems to enhance protein-DNA recognition despite its intrinsically slow kinetics.


Subject(s)
DNA , Nucleic Acid Conformation , Protein Binding , Thermodynamics , Kinetics , DNA/metabolism , DNA/chemistry , Sex-Determining Region Y Protein/metabolism , Sex-Determining Region Y Protein/chemistry , Sex-Determining Region Y Protein/genetics , Binding Sites , Models, Molecular , Protein Conformation , Molecular Dynamics Simulation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry
7.
Nat Commun ; 15(1): 2796, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38555298

ABSTRACT

The Y-linked SRY gene initiates mammalian testis-determination. However, how the expression of SRY is regulated remains elusive. Here, we demonstrate that a conserved steroidogenic factor-1 (SF-1)/NR5A1 binding enhancer is required for appropriate SRY expression to initiate testis-determination in humans. Comparative sequence analysis of SRY 5' regions in mammals identified an evolutionary conserved SF-1/NR5A1-binding motif within a 250 bp region of open chromatin located 5 kilobases upstream of the SRY transcription start site. Genomic analysis of 46,XY individuals with disrupted testis-determination, including a large multigenerational family, identified unique single-base substitutions of highly conserved residues within the SF-1/NR5A1-binding element. In silico modelling and in vitro assays demonstrate the enhancer properties of the NR5A1 motif. Deletion of this hemizygous element by genome-editing, in a novel in vitro cellular model recapitulating human Sertoli cell formation, resulted in a significant reduction in expression of SRY. Therefore, human NR5A1 acts as a regulatory switch between testis and ovary development by upregulating SRY expression, a role that may predate the eutherian radiation. We show that disruption of an enhancer can phenocopy variants in the coding regions of SRY that cause human testis dysgenesis. Since disease causing variants in enhancers are currently rare, the regulation of gene expression in testis-determination offers a paradigm to define enhancer activity in a key developmental process.


Subject(s)
Gonadal Dysgenesis , Testis , Animals , Female , Humans , Male , Cell Line , Mammals/genetics , Regulatory Sequences, Nucleic Acid , Sertoli Cells/metabolism , Sex-Determining Region Y Protein/genetics , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism
8.
Nucleic Acids Res ; 52(10): 5514-5528, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38499491

ABSTRACT

Male development in mammals depends on the activity of the two SOX gene: Sry and Sox9, in the embryonic testis. As deletion of Enhancer 13 (Enh13) of the Sox9 gene results in XY male-to-female sex reversal, we explored the critical elements necessary for its function and hence, for testis and male development. Here, we demonstrate that while microdeletions of individual transcription factor binding sites (TFBS) in Enh13 lead to normal testicular development, combined microdeletions of just two SRY/SOX binding motifs can alone fully abolish Enh13 activity leading to XY male-to-female sex reversal. This suggests that for proper male development to occur, these few nucleotides of non-coding DNA must be intact. Interestingly, we show that depending on the nature of these TFBS mutations, dramatically different phenotypic outcomes can occur, providing a molecular explanation for the distinct clinical outcomes observed in patients harboring different variants in the same enhancer.


Subject(s)
Enhancer Elements, Genetic , Sex Determination Processes , Animals , Female , Humans , Male , Mice , Binding Sites/genetics , Enhancer Elements, Genetic/genetics , Sequence Deletion , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Testis/growth & development , Phenotype
9.
J Hepatol ; 80(6): 928-940, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38336346

ABSTRACT

BACKGROUND & AIMS: Men are more prone to develop and die from liver fibrosis than women. In this study, we aim to investigate how sex-determining region Y gene (SRY) in hepatocytes promotes liver fibrosis. METHODS: Hepatocyte-specific Sry knock-in (KI), Sry knockout (KO), and Sry KI with platelet-derived growth factor receptor α (Pdgfrα) KO mice were generated. Liver fibrosis was induced in mice by bile duct ligation for 2 weeks or carbon tetrachloride treatment for 6 weeks. In addition, primary hepatocytes, hepatic stellate cells (HSCs), and immortalized cell lines were used for in vitro studies and mechanistic investigation. RESULTS: Compared to females, the severity of toxin- or cholestasis-induced liver fibrosis is similarly increased in castrated and uncastrated male mice. Among all Y chromosome-encoded genes, SRY was the most significantly upregulated and consistently increased gene in fibrotic/cirrhotic livers in male patients and in mouse models. Sry KI mice developed exacerbated liver fibrosis, whereas Sry KO mice had alleviated liver fibrosis, compared to age- and sex-matched control mice after bile duct ligation or administration of carbon tetrachloride. Mechanistically, both our in vivo and in vitro studies illustrated that SRY in hepatocytes can transcriptionally regulate Pdgfrα expression, and promote HMGB1 (high mobility group box 1) release and subsequent HSC activation. Pdgfrα KO or treatment with the SRY inhibitor DAX1 in Sry KI mice abolished SRY-induced HMGB1 secretion and liver fibrosis. CONCLUSIONS: SRY is a strong pro-fibrotic factor and accounts for the sex disparity observed in liver fibrosis, suggesting its critical role as a potentially sex-specific therapeutic target for prevention and treatment of the disease. IMPACT AND IMPLICATION: We identified that a male-specific gene, sex-determining region Y gene (SRY), is a strong pro-fibrotic gene that accounts for the sex disparity observed in liver fibrosis. As such, SRY might be an appropriate target for surveillance and treatment of liver fibrosis in a sex-specific manner. Additionally, SRY might be a key player in the sexual dimorphism observed in hepatic pathophysiology more generally.


Subject(s)
Hepatic Stellate Cells , Hepatocytes , Liver Cirrhosis , Sex-Determining Region Y Protein , Animals , Female , Male , Mice , Carbon Tetrachloride/toxicity , Carbon Tetrachloride/adverse effects , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/physiopathology , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice, Knockout , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sex Characteristics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism
10.
Prenat Diagn ; 44(5): 580-585, 2024 05.
Article in English | MEDLINE | ID: mdl-38204192

ABSTRACT

BACKGROUND: Generally, the translocation of SRY onto one of the X chromosomes leads to 46, XX testicular disorders of sex development, a relatively rare condition characterized by the presence of testicular tissue with a 46, XX karyotype. Three prenatal cases of unbalanced X; Y translocation carrying SRY were identified in this study. METHODS: Structural variants were confirmed using single nucleotide polymorphism array and chromosomal karyotyping. X chromosome inactivation (XCI) was also analyzed. Detailed clinical features of the three cases were collected. RESULTS: We identified two fetuses with maternal inherited unbalanced X; Y translocations carrying SRY and skewed XCI presenting with normal female external genitalia, and one fetus with de novo 46, XX (SRY+) and random XCI manifested male phenotypic external genitalia. CONCLUSION: This study reports that cases with unbalanced X; Y translocations carrying SRY manifested a normal female external genitalia in a prenatal setting. We speculate that the skewed XCI mediates the silence of SRY. In addition, our study emphasizes that combining clinical findings with pedigree analysis is critical for estimating the prognosis of fetuses with sex chromosome abnormalities.


Subject(s)
Chromosomes, Human, X , Chromosomes, Human, Y , Translocation, Genetic , Adult , Female , Humans , Male , Pregnancy , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Cytogenetic Analysis , Karyotyping , Prenatal Diagnosis/methods , Sex Chromosome Aberrations , Sex-Determining Region Y Protein/genetics , X Chromosome Inactivation/genetics
11.
DNA Cell Biol ; 43(1): 12-25, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38170186

ABSTRACT

The male sex-determining gene, sex-determining region on the Y chromosome (SRY), is expressed in adult testicular germ cells; however, its role in regulating spermatogenesis remains unclear. The role of SRY in the postmeiotic gene expression was investigated by determining the effect of SRY on the promoter of the haploid-specific Protamine 1 (PRM1) gene, which harbors five distinct SRY-binding motifs. In a luciferase reporter assay system, SRY upregulates PRM1 promoter activity in vitro in a dose-dependent manner. Through a gel-shift assay involving a 31-bp DNA fragment encompassing the SRY element within the PRM1 promoter, the third SRY-binding site on the sense strand (-373/-367) was identified as crucial for PRM1 promoter activation. This assay was extended to analyze 9 SRY variants found in the testicular DNA of 44 azoospermia patients. The findings suggest that SRY regulates PRM1 promoter activity by directly binding to its specific motif within the PRM1 promoter.


Subject(s)
Testis , Y Chromosome , Humans , Male , DNA/metabolism , Protamines/genetics , Protamines/metabolism , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Testis/metabolism , Y Chromosome/metabolism
12.
Genes (Basel) ; 15(1)2024 01 16.
Article in English | MEDLINE | ID: mdl-38254992

ABSTRACT

The translocation of the testis-determining factor, the SRY gene, from the Y to the X chromosome is a rare event that causes abnormalities in gonadal development. In all cases of males and females carrying this translocation, disorder of sex development is reported. In our study, we described a peculiar pedigree with the first evidence of four healthy females from three generations who are carriers of the newly identified t(X;Y)(q28;p11.2)(SRY+) translocation with no evidence of ambiguous genitalia or other SRY-dependent alterations. Our study was a consequence of a Non-Invasive Prenatal Test (NIPT) showing a sexual chromosomal abnormality (XXY) followed by a chorionic villus analysis suggesting a normal karyotype 46,XX and t(X;Y) translocation detected by FISH. Here, we (i) demonstrated the inheritance of the translocation in the maternal lineage via karyotyping and FISH analysis; (ii) characterised the structural rearrangement via chromosomal microarray; and (iii) demonstrated, via Click-iT® EdU Imaging assay, that there was an absolute preferential inactivation of the der(X) chromosome responsible for the lack of SRY expression. Overall, our study provides valuable genetic and molecular information that may lead personal and medical decisions.


Subject(s)
Chromosomes, Human, X , Genes, sry , Male , Pregnancy , Humans , Female , Sex-Determining Region Y Protein/genetics , Chromosomes, Human, X/genetics , Chromosome Aberrations , Karyotyping , Translocation, Genetic/genetics
13.
Science ; 382(6670): 600-606, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917714

ABSTRACT

Sex determination in mammals depends on the differentiation of the supporting lineage of the gonads into Sertoli or pregranulosa cells that govern testis and ovary development, respectively. Although the Y-linked testis-determining gene Sry has been identified, the ovarian-determining factor remains unknown. In this study, we identified -KTS, a major, alternatively spliced isoform of the Wilms tumor suppressor WT1, as a key determinant of female sex determination. Loss of -KTS variants blocked gonadal differentiation in mice, whereas increased expression, as found in Frasier syndrome, induced precocious differentiation of ovaries independently of their genetic sex. In XY embryos, this antagonized Sry expression, resulting in male-to-female sex reversal. Our results identify -KTS as an ovarian-determining factor and demonstrate that its time of activation is critical in gonadal sex differentiation.


Subject(s)
Ovary , Sex Determination Processes , WT1 Proteins , Animals , Female , Male , Mice , Ovary/growth & development , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Testis/growth & development , WT1 Proteins/genetics , WT1 Proteins/metabolism , Protein Isoforms
14.
Proc Natl Acad Sci U S A ; 120(20): e2221499120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155872

ABSTRACT

In mammals, male and female gonads initially develop from bipotential progenitor cells, which can differentiate into either testicular or ovarian cells. The decision to adopt a testicular or ovarian fate relies on robust genetic forces, i.e., activation of the testis-determining gene Sry, as well as a delicate balance of expression levels for pro-testis and pro-ovary factors. Recently, epigenetic regulation has been found to be a key element in activation of Sry. Nevertheless, the mechanism by which epigenetic regulation controls the expression balance of pro-testis and pro-ovary factors remains unclear. Chromodomain Y-like protein (CDYL) is a reader protein for repressive histone H3 methylation marks. We found that a subpopulation of Cdyl-deficient mice exhibited XY sex reversal. Gene expression analysis revealed that the testis-promoting gene Sox9 was downregulated in XY Cdyl-deficient gonads during the sex determination period without affecting Sry expression. Instead, we found that the ovary-promoting gene Wnt4 was derepressed in XY Cdyl-deficient gonads prior to and during the sex-determination period. Wnt4 heterozygous deficiency restored SOX9 expression in Cdyl-deficient XY gonads, indicating that derepressed Wnt4 is a cause of the repression of Sox9. We found that CDYL directly bound to the Wnt4 promoter and maintained its H3K27me3 levels during the sex-determination period. These findings indicate that CDYL reinforces male gonadal sex determination by repressing the ovary-promoting pathway in mice.


Subject(s)
Epigenesis, Genetic , Sex Determination Processes , Animals , Female , Male , Mice , Gene Expression Regulation, Developmental , Gonads/metabolism , Mammals/genetics , Ovary/metabolism , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Testis/metabolism , Wnt4 Protein/genetics , Wnt4 Protein/metabolism
15.
J Vet Med Sci ; 85(4): 507-514, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-36858585

ABSTRACT

The expression of sex determining region of the Y chromosome (Sry) in the fetal gonads is important for male development. In a mouse model of disorders of sex development (C57BL/6 (B6)-XYPOS), the gonadal phenotype and the timing of Sry expression differ due to differences among B6 substrains as the genetic background. Since differences in Sry expression among B6 substrains have been speculated, the present study examined Sry expression in B6J, B6JJmsSlc, and B6NCrl mice. These substrains differed in the number of Sry-expressing cells in the gonads of embryonic mice at each developmental stage, with B6NCrl having more than the other strains. The substrains differed also in the number of Sry-expressing cells between the left and right gonads, with B6J and B6NCrl, but not B6JJmsSlc, showing left gonad-dominant Sry expression. Substrain differences existed also in the distribution of Sry-expressing cells in the medial and lateral directions of gonads. In addition, in the left gonad-dominant Sry-expressing substrains B6J and B6NCrl, the medial and central regions of the left gonad had more Sry-expressing cells than those of the right gonad. Substrains of B6 mice have not always been considered in sex differentiation studies. In the present study, however, we observed substrain differences in the number of Sry-expressing cells, left-right distribution, and medial/lateral distribution during the early stages of gonadal development in B6 mice. Therefore, future studies on sex differentiation in B6 mice should consider substrain differences.


Subject(s)
Gonads , Y Chromosome , Mice , Male , Animals , Mice, Inbred C57BL , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Y Chromosome/genetics , Y Chromosome/metabolism , Sex Differentiation/genetics , Testis/metabolism
16.
Development ; 150(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36912416

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.


Subject(s)
Gonads , Sex Determination Processes , Female , Mice , Male , Animals , Gonads/metabolism , Sertoli Cells/metabolism , Cell Differentiation , Embryonic Development , SOX9 Transcription Factor/metabolism , Testis/metabolism , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Mammals/metabolism
17.
Anim Genet ; 54(2): 93-103, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36504456

ABSTRACT

Swyer syndrome is where an individual has the karyotype of a typical male yet is phenotypically a female. The lack of a (functional) SRY gene located on the Y-chromosome is implicated in some cases of the Swyer syndrome, although many Swyer individuals with an apparently fully functional SRY gene have also been documented. The present study undertook whole genome sequence analyses of eight cattle with suspected Swyer syndrome and compared their genome to that of both a control male and female. Sequence analyses coupled with female phenotypes confirmed that all eight individuals had the 60,XY sex reversal Swyer syndrome. Seven of the eight Swyer syndrome individuals had a deletion on the Y chromosome encompassing the SRY gene (i.e., SRY-). The eighth individual had no obvious mutation in the SRY gene (SRY+) or indeed in any reported gene associated with sex reversal in mammals; a necropsy was performed on this individual. No testicles were detected during the necropsy. Histological examination of the reproductive tract revealed an immature uterine body and horns with inactive glandular tissue of normal histological appearance; both gonads were elongated, a characteristic of most reported cases of Swyer in mammals. The flanking sequence of 11 single nucleotide polymorphisms within 10 kb of the SRY gene are provided to help diagnose some cases of Swyer syndrome. These single nucleotide polymorphisms will not, however, detect all cases of Swyer syndrome since, as evidenced from the present study (and other studies), some individuals with the Swyer condition still contain the SRY gene (i.e., SRY+).


Subject(s)
Cattle Diseases , Gonadal Dysgenesis, 46,XY , Male , Cattle/genetics , Female , Animals , Gonadal Dysgenesis, 46,XY/genetics , Mutation , Genes, sry , Y Chromosome/genetics , Testis , Sex-Determining Region Y Protein/genetics , Mammals/genetics , Cattle Diseases/genetics
18.
Hum Mol Genet ; 32(1): 55-64, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35921234

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.


Subject(s)
Gonadal Dysgenesis, 46,XY , Regulatory Sequences, Nucleic Acid , Animals , Female , Male , Mice , Binding Sites , Mammals/metabolism , Sex Determination Processes , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Testis/metabolism , Genes, sry
19.
Life Sci Alliance ; 6(2)2023 02.
Article in English | MEDLINE | ID: mdl-36414375

ABSTRACT

Sry on the Y chromosome is the master switch of sex determination in mammals. It has been well established that Sry encodes a transcription factor that is transiently expressed in somatic cells of the male gonad, leading to the formation of testes. In the testis of adult mice, Sry is expressed as a circular RNA (circRNA) transcript. However, the physiological function of Sry circRNA (circSRY) remains unknown since its discovery in 1993. Here we show that circSRY is mainly expressed in the spermatocytes, but not in mature sperm or somatic cells of the testis. Loss of circSRY led to germ cell apoptosis and the reduction of sperm count in the epididymis. The level of γH2AX was decreased, and failure of XY body formation was noted in circSRY KO germ cells. Further study demonstrated that circSRY directly bound to miR-138-5p in spermatocytes, and in vitro assay suggested that circSRY regulates H2AX mRNA through sponging miR-138-5p. Our study demonstrates that, besides determining sex, Sry also plays an important role in spermatogenesis as a circRNA.


Subject(s)
MicroRNAs , RNA, Circular , Male , Mice , Animals , Sex-Determining Region Y Protein/genetics , RNA, Circular/genetics , Semen , Spermatogenesis/genetics , Germ Cells , MicroRNAs/genetics , Mammals/genetics
20.
Cytogenet Genome Res ; 163(5-6): 307-316, 2023.
Article in English | MEDLINE | ID: mdl-38246151

ABSTRACT

INTRODUCTION: Testis differentiation is initiated by the SRY gene on the Y chromosome in mammalian species. However, the Amami spiny rat, Tokudaia osimensis, lacks both the Y chromosome and the Sry gene and acquired a unique Sox9 regulatory mechanism via a male-specific duplication upstream of Sox9, without Sry. In general mammalian species, the SRY protein binds to a testis-specific enhancer to promote SOX9 gene expression. Several enhancers located upstream of Sox9/SOX9 have been reported in mice and humans. In particular, the binding of SRY to the highly conserved enhancer Enh13 is thought to be a common mechanism underlying testis differentiation and sex determination in mammals. METHODS: Sequences of T. osimensis homologues of three Sox9 enhancers that were previously reported in mice, Enh8, Enh14, and Enh13, were determined. We performed in vitro assays to confirm enhancer activity involved in Sox9 regulation in T. osimensis. RESULTS: T. osimensis Enh13 showed enhancer activity when co-transfected with NR5A1 and SOX9. Mouse Enh13 was activated by NR5A1 and SRY; however, T. osimensis Enh13 did not respond to SRY, even though the binding sites of SRY and NR5A1 were conserved. To identify the key sequence that is present in mouse but absent from T. osimensis, we performed reporter gene assays using vectors in which partial sequences of T. osimensis Enh13 were replaced with mouse sequences. For T. osimensis Enh13 in which the second half (approximately 430 bp) was replaced with the corresponding mouse sequence, activity in response to NR5A1 and SRY was recovered. Further, reporter assays revealed that multiple regions in the second half of the mouse Enh13 sequence are required for the response to NR5A1 and SRY. The latter 49 bp was particularly important and contained four binding sites for three transcription factors, POU2F1, HOXA3, and GATA1. CONCLUSION: We showed that there are unknown sequences responsible for the interaction between NR5A1 and SRY and mEnh13 based on comparative analyses of Sry-dependent and Sry-independent species. Our comparative analyses revealed new molecular mechanisms underlying mammalian sex determination.


Subject(s)
Enhancer Elements, Genetic , SOX9 Transcription Factor , Sex-Determining Region Y Protein , Animals , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Mice , Male , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Rats , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testis/metabolism , Base Sequence
SELECTION OF CITATIONS
SEARCH DETAIL