Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 32(6): 1032-1047, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36282544

ABSTRACT

Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a congenital condition characterized by aplasia or hypoplasia of the uterus and vagina in women with a 46,XX karyotype. This condition can occur as type I when isolated or as type II when associated with extragenital anomalies including kidney and skeletal abnormalities. The genetic basis of MRKH syndrome remains unexplained and several candidate genes have been proposed to play a role in its etiology, including HNF1B, LHX1 and WNT4. Here, we conducted a microarray analysis of 13 women affected by MRKH syndrome, resulting in the identification of chromosomal changes, including the deletion at 17q12, which contains both HNF1B and LHX1. We focused on HNF1B for further investigation due to its known association with, but unknown etiological role in, MRKH syndrome. We ablated Hnf1b specifically in the epithelium of the Müllerian ducts in mice and found that this caused hypoplastic development of the uterus, as well as kidney anomalies, closely mirroring the MRKH type II phenotype. Using single-cell RNA sequencing of uterine tissue in the Hnf1b-ablated embryos, we analyzed the molecules and pathways downstream of Hnf1b, revealing a dysregulation of processes associated with cell proliferation, migration and differentiation. Thus, we establish that loss of Hnf1b function leads to an MRKH phenotype and generate the first mouse model of MRKH syndrome type II. Our results support the investigation of HNF1B in clinical genetic settings of MRKH syndrome and shed new light on the molecular mechanisms underlying this poorly understood condition in women's reproductive health.


Subject(s)
46, XX Disorders of Sex Development , Mullerian Ducts , Animals , Female , Mice , 46, XX Disorders of Sex Development/genetics , Cell Differentiation , Genomics , Hepatocyte Nuclear Factor 1-beta/genetics , Humans
2.
Annu Rev Genet ; 51: 265-285, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28853925

ABSTRACT

Sexual reproduction crucially depends on the production of sperm in males and oocytes in females. Both types of gamete arise from the same precursor, the germ cells. We review the events that characterize the development of germ cells during fetal life as they commit to, and prepare for, oogenesis or spermatogenesis. In females, fetal germ cells enter meiosis, whereas in males they delay meiosis and instead lose pluripotency, activate an irreversible program of prospermatogonial differentiation, and temporarily cease dividing. Both pathways involve sex-specific molecular signals from the somatic cells of the developing gonads and a suite of intrinsic receptors, signal transducers, transcription factors, RNA stability factors, and epigenetic modulators that act in complex, interconnected positive and negative regulatory networks. Understanding these networks is important in the contexts of the etiology, diagnosis, and treatment of infertility and gonadal cancers, and in efforts to augment human and animal fertility using stem cell approaches.


Subject(s)
Infertility, Female/genetics , Infertility, Male/genetics , Oogenesis/genetics , Sex Determination Processes , Sex Differentiation/genetics , Spermatogenesis/genetics , Animals , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Infertility, Female/metabolism , Infertility, Female/pathology , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Meiosis , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Ovum/cytology , Ovum/growth & development , Ovum/metabolism , Signal Transduction , Spermatozoa/cytology , Spermatozoa/growth & development , Spermatozoa/metabolism
3.
Hum Mol Genet ; 31(13): 2223-2235, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35134173

ABSTRACT

The genetic regulation of ovarian development remains largely unclear. Indeed, in most cases of impaired ovarian development-such as 46,XX disorders of sex development (DSD) without SRY, and premature ovarian insufficiency (POI)-the genetic causes have not been identified, and the vast majority of disease-associated sequence variants could lie within non-coding regulatory sequences. In this study, we aimed to identify enhancers of five ovarian genes known to play key roles in early ovarian development, basing our analysis on the expression of enhancer derived transcripts (eRNAs), which are considered to characterize active enhancers. Temporal expression profile changes in mouse WT1-positive ovarian cells were obtained from cap analysis of gene expression at E13.5, E16.5 and P0. We compared the chronological expression profiles of ovarian-specific eRNA with expression profiles for each of the ovarian-specific genes, yielding two candidate sequences for enhancers of Wnt4 and Rspo1. Both sequences are conserved between mouse and human, and we confirmed their enhancer activities using transient expression assays in murine granulosa cells. Furthermore, by sequencing the region in patients with impaired ovarian development in 24 patients, such as POI, gonadal dysgenesis and 46,XX DSD, we identified rare single nucleotide variants in both sequences. Our results demonstrate that combined analysis of the temporal expression profiles of eRNA and mRNA of target genes presents a powerful tool for locating cis-element enhancers, and a means of identifying disease-associated sequence variants that lie within non-coding regulatory sequences, thus advancing an important unmet need in forward human genetics.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Animals , Enhancer Elements, Genetic/genetics , Female , Genetic Variation , Humans , Menopause, Premature/genetics , Mice , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , RNA/genetics , Time Factors
4.
Development ; 148(5)2021 03 09.
Article in English | MEDLINE | ID: mdl-33574039

ABSTRACT

In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Germ Cells/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , CRISPR-Cas Systems/genetics , Female , Fetal Development/genetics , Fetus/cytology , Fetus/metabolism , Gene Expression Regulation/drug effects , Germ Cells/cytology , Meiosis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis , Ovary/cytology , Ovary/metabolism , Promoter Regions, Genetic , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , Tretinoin/pharmacology
5.
Nucleic Acids Res ; 49(19): 10931-10955, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34570228

ABSTRACT

Few genetically dominant mutations involved in human disease have been fully explained at the molecular level. In cases where the mutant gene encodes a transcription factor, the dominant-negative mode of action of the mutant protein is particularly poorly understood. Here, we studied the genome-wide mechanism underlying a dominant-negative form of the SOX18 transcription factor (SOX18RaOp) responsible for both the classical mouse mutant Ragged Opossum and the human genetic disorder Hypotrichosis-lymphedema-telangiectasia-renal defect syndrome. Combining three single-molecule imaging assays in living cells together with genomics and proteomics analysis, we found that SOX18RaOp disrupts the system through an accumulation of molecular interferences which impair several functional properties of the wild-type SOX18 protein, including its target gene selection process. The dominant-negative effect is further amplified by poisoning the interactome of its wild-type counterpart, which perturbs regulatory nodes such as SOX7 and MEF2C. Our findings explain in unprecedented detail the multi-layered process that underpins the molecular aetiology of dominant-negative transcription factor function.


Subject(s)
Glomerulonephritis/genetics , Hypotrichosis/genetics , Lymphedema/genetics , SOXF Transcription Factors/genetics , Telangiectasis/genetics , Transcription, Genetic , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Gene Expression Regulation , Gene Regulatory Networks , Genes, Reporter , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Hypotrichosis/metabolism , Hypotrichosis/pathology , Luciferases/genetics , Luciferases/metabolism , Lymphedema/metabolism , Lymphedema/pathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mutation , SOXF Transcription Factors/metabolism , Single Molecule Imaging , Telangiectasis/metabolism , Telangiectasis/pathology
6.
Dev Dyn ; 251(2): 336-349, 2022 02.
Article in English | MEDLINE | ID: mdl-34174014

ABSTRACT

BACKGROUND: Lymphatic vascular development is regulated by well-characterized signaling and transcriptional pathways. These pathways regulate lymphatic endothelial cell (LEC) migration, motility, polarity, and morphogenesis. Canonical and non-canonical WNT signaling pathways are known to control LEC polarity and development of lymphatic vessels and valves. PKD1, encoding Polycystin-1, is the most commonly mutated gene in polycystic kidney disease but has also been shown to be essential in lymphatic vascular morphogenesis. The mechanism by which Pkd1 acts during lymphangiogenesis remains unclear. RESULTS: Here we find that loss of non-canonical WNT signaling components Wnt5a and Ryk phenocopy lymphatic defects seen in Pkd1 knockout mice. To investigate genetic interaction, we generated Pkd1;Wnt5a double knockout mice. Loss of Wnt5a suppressed phenotypes seen in the lymphatic vasculature of Pkd1-/- mice and Pkd1 deletion suppressed phenotypes observed in Wnt5a-/- mice. Thus, we report mutually suppressive roles for Pkd1 and Wnt5a, with developing lymphatic networks restored to a more wild type state in double mutant mice. This genetic interaction between Pkd1 and the non-canonical WNT signaling pathway ultimately controls LEC polarity and the morphogenesis of developing vessel networks. CONCLUSION: Our work suggests that Pkd1 acts at least in part by regulating non-canonical WNT signaling during the formation of lymphatic vascular networks.


Subject(s)
Lymphatic Vessels , Polycystic Kidney Diseases , Animals , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , Morphogenesis/genetics , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Protein Kinase C , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway/genetics , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
7.
Hum Mutat ; 43(3): 362-379, 2022 03.
Article in English | MEDLINE | ID: mdl-34918413

ABSTRACT

SRY is the Y-chromosomal gene that determines male sex development in humans and most other mammals. After three decades of study, we still lack a detailed understanding of which domains of the SRY protein are required to engage the pathway of gene activity leading to testis development. Some insight has been gained from the study of genetic variations underlying differences/disorders of sex determination (DSD), but the lack of a system of experimentally generating SRY mutations and studying their consequences in vivo has limited progress in the field. To address this issue, we generated a mouse model carrying a human SRY transgene able to drive testis determination in XX mice. Using CRISPR-Cas9 gene editing, we generated novel genetic modifications in each of SRY's three domains (N-terminal, HMG box, and C-terminal) and performed a detailed analysis of their molecular and cellular effects on embryonic testis development. Our results provide new functional insights unique to human SRY and present a versatile and powerful system in which to functionally analyze variations of SRY including known and novel pathogenic variants found in DSD.


Subject(s)
Gene Editing , Mice, Transgenic , Sex-Determining Region Y Protein , Testis , Animals , Humans , Male , Mice , Protein Domains , Sex-Determining Region Y Protein/genetics , Testis/metabolism
8.
Hum Mol Genet ; 29(13): 2148-2161, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32452519

ABSTRACT

In mice, male sex determination depends on FGF9 signalling via FGFR2c in the bipotential gonads to maintain the expression of the key testis gene SOX9. In humans, however, while FGFR2 mutations have been linked to 46,XY disorders of sex development (DSD), the role of FGF9 is unresolved. The only reported pathogenic mutations in human FGF9, FGF9S99N and FGF9R62G, are dominant and result in craniosynostosis (fusion of cranial sutures) or multiple synostoses (fusion of limb joints). Whether these synostosis-causing FGF9 mutations impact upon gonadal development and DSD etiology has not been explored. We therefore examined embryonic gonads in the well-characterized Fgf9 missense mouse mutants, Fgf9S99N and Fgf9N143T, which phenocopy the skeletal defects of FGF9S99N and FGF9R62G variants, respectively. XY Fgf9S99N/S99N and XY Fgf9N143T/N143T fetal mouse gonads showed severely disorganized testis cords and partial XY sex reversal at 12.5 days post coitum (dpc), suggesting loss of FGF9 function. By 15.5 dpc, testis development in both mutants had partly recovered. Mitotic analysis in vivo and in vitro suggested that the testicular phenotypes in these mutants arise in part through reduced proliferation of the gonadal supporting cells. These data raise the possibility that human FGF9 mutations causative for dominant skeletal conditions can also lead to loss of FGF9 function in the developing testis, at least in mice. Our data suggest that, in humans, testis development is largely tolerant of deleterious FGF9 mutations which lead to skeletal defects, thus offering an explanation as to why XY DSDs are rare in patients with pathogenic FGF9 variants.


Subject(s)
Fibroblast Growth Factor 9/genetics , Ovotesticular Disorders of Sex Development/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Synostosis/genetics , Animals , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/genetics , Gonads/growth & development , Gonads/pathology , Humans , Male , Mice , Mutation, Missense/genetics , Ovotesticular Disorders of Sex Development/pathology , SOX9 Transcription Factor/genetics , Sex Determination Processes/genetics , Sexual Development/genetics
9.
J Cell Sci ; 132(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30877223

ABSTRACT

The nuclear receptor NR5A1 is equally expressed and required for development of the gonadal primordia of both sexes, but, after sex determination, it is upregulated in XY testes and downregulated in XX ovaries. We have recently demonstrated, in mice, that this downregulation is mediated by forkhead box L2 (FOXL2) and hypothesized that adequate suppression of Nr5a1 is essential for normal ovarian development. Further, analysis of human patients with disorders/differences of sex development suggests that overexpression of NR5A1 can result in XX (ovo)testicular development. Here, we tested the role of Nr5a1 by overexpression in fetal gonads using a Wt1-BAC (bacterial artificial chromosome) transgene system. Enforced Nr5a1 expression compromised ovarian development in 46,XX mice, resulting in late-onset infertility, but did not induce (ovo)testis differentiation. The phenotype was similar to that of XX mice lacking Notch signaling. The expression level of Notch2 was significantly reduced in Nr5a1 transgenic mice, and the ovarian phenotype was almost completely rescued by in utero treatment with a NOTCH2 agonist. We conclude that suppression of Nr5a1 during the fetal period optimizes ovarian development by fine-tuning Notch signaling.


Subject(s)
Ovary/physiology , Receptor, Notch2/physiology , Sexual Development , Steroidogenic Factor 1/physiology , Testis/physiology , Animals , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Transgenic , Phenotype , Receptor, Notch2/genetics , Signal Transduction , Steroidogenic Factor 1/genetics
10.
Genes Dev ; 27(22): 2409-26, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24240231

ABSTRACT

Development of testes in the mammalian embryo requires the formation and assembly of several cell types that allow these organs to achieve their roles in male reproduction and endocrine regulation. Testis development is unusual in that several cell types such as Sertoli, Leydig, and spermatogonial cells arise from bipotential precursors present in the precursor tissue, the genital ridge. These cell types do not differentiate independently but depend on signals from Sertoli cells that differentiate under the influence of transcription factors SRY and SOX9. While these steps are becoming better understood, the origins and roles of many testicular cell types and structures-including peritubular myoid cells, the tunica albuginea, the arterial and venous blood vasculature, lymphatic vessels, macrophages, and nerve cells-have remained unclear. This review synthesizes current knowledge of how the architecture of the testis unfolds and highlights the questions that remain to be explored, thus providing a roadmap for future studies that may help illuminate the causes of XY disorders of sex development, infertility, and testicular cancers.


Subject(s)
Cell Differentiation , Testis/cytology , Testis/embryology , Animals , Humans , Macrophages/metabolism , Male , Sertoli Cells/cytology , Testis/blood supply , Testis/innervation
11.
PLoS Genet ; 13(1): e1006520, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28045957

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.


Subject(s)
Enhancer Elements, Genetic , SOX9 Transcription Factor/genetics , Sex Determination Processes/genetics , Testis/metabolism , Alleles , Animals , Female , Gene Deletion , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , SOX9 Transcription Factor/metabolism , Testis/growth & development
12.
PLoS Genet ; 13(2): e1006584, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28146551

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pgen.1006520.].

13.
Development ; 143(10): 1633-7, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27190031

ABSTRACT

On Thursday 9 May 1991, the world awoke to front-page news of a breakthrough in biological research. From Washington to Wollongong, newspapers, radio and TV were abuzz with the story of a transgenic mouse in London called Randy. Why was this mouse so special? The mouse in question was a chromosomal female (XX) made male by the presence of a transgene containing the Y chromosome gene Sry This sex-reversal provided clear experimental proof that Sry was the elusive mammalian sex-determining gene. Twenty-five years on, we reflect on what this discovery meant for our understanding of how males and females arise and what remains to be understood.


Subject(s)
Sex Determination Processes , Animals , Female , Male , Mice , Mice, Transgenic , Sex-Determining Region Y Protein/genetics
14.
Hum Mutat ; 39(12): 1861-1874, 2018 12.
Article in English | MEDLINE | ID: mdl-30067310

ABSTRACT

Nuclear receptor subfamily 5 group A member 1/Steroidogenic factor 1 (NR5A1; SF-1; Ad4BP) mutations cause 46,XY disorders of sex development (DSD), with phenotypes ranging from developmentally mild (e.g., hypospadias) to severe (e.g., complete gonadal dysgenesis). The molecular mechanism underlying this spectrum is unclear. During sex determination, SF-1 regulates SOX9 (SRY [sex determining region Y]-box 9) expression. We hypothesized that SF-1 mutations in 46,XY DSD patients affect SOX9 expression via the Testis-specific Enhancer of Sox9 core element, TESCO. Our objective was to assess the ability of 20 SF-1 mutants found in 46,XY DSD patients to activate TESCO. Patient DNA was sequenced for SF-1 mutations and mutant SF-1 proteins were examined for transcriptional activity, protein expression, sub-cellular localization and in silico structural defects. Fifteen of the 20 mutants showed reduced SF-1 activation on TESCO, 11 with atypical sub-cellular localization. Fourteen SF-1 mutants were predicted in silico to alter DNA, ligand or cofactor interactions. Our study may implicate aberrant SF-1-mediated transcriptional regulation of SOX9 in 46,XY DSDs.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Enhancer Elements, Genetic , Mutation , SOX9 Transcription Factor/genetics , Steroidogenic Factor 1/genetics , Adolescent , Adult , Child , Child, Preschool , Computer Simulation , Gene Expression Regulation , HEK293 Cells , Humans , Infant , Infant, Newborn , Ligands , Male , Protein Binding , Sequence Analysis, DNA/methods , Steroidogenic Factor 1/chemistry , Steroidogenic Factor 1/metabolism
15.
Dev Biol ; 423(1): 46-56, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28118982

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.


Subject(s)
Cell Differentiation , Germ Cells/cytology , Germ Cells/metabolism , Morphogenesis , SOXC Transcription Factors/metabolism , Testis/growth & development , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Female , Fetus/cytology , Gene Expression Regulation, Developmental , Male , Mice , SOXC Transcription Factors/genetics , Sex-Determining Region Y Protein/metabolism , Spermatogenesis , Testis/cytology
16.
J Cell Physiol ; 233(4): 3262-3273, 2018 04.
Article in English | MEDLINE | ID: mdl-28884822

ABSTRACT

RNA-binding proteins (RBP) are important facilitators of post-transcriptional gene regulation. We have previously established that nuclear overexpression of the RBP Musashi-2 (MSI2) during male germ cell maturation is detrimental to sperm cell development and fertility. Herein we determine the genes and pathways impacted by the upregulation of Msi2. Microarray analysis and qPCR confirmed differential gene expression in factors fundamental to the cell cycle, cellular proliferation, and cell death. Similarly, comparative protein expression analysis via iTRAQ, immunoblot, and immunolocalization, identified differential expression and localization of important regulators of transcription, translation, RNA processing, and spermatogenesis. Specifically, the testis-expressed transcription factor, Tbx1, and the piRNA regulator of gamete development, Piwil1, were both found to be targeted for translational repression by MSI2. This study provides key evidence to support a fundamental role for MSI2 in post-transcriptional regulation during male gamete development.


Subject(s)
Argonaute Proteins/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis , T-Box Domain Proteins/metabolism , Animals , Argonaute Proteins/genetics , Gene Expression Regulation , Male , Mice, Transgenic , Models, Biological , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spermatids/metabolism , Spermatogenesis/genetics , T-Box Domain Proteins/genetics
17.
Development ; 142(6): 1083-8, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25725066

ABSTRACT

Genes related to Dmrt1, which encodes a DNA-binding DM domain transcription factor, act as triggers for primary sex determination in a broad range of metazoan species. However, this role is fulfilled in mammals by Sry, a newly evolved gene on the Y chromosome, such that Dmrt1 has become dispensable for primary sex determination and instead maintains Sertoli cell phenotype in postnatal testes. Here, we report that enforced expression of Dmrt1 in XX mouse fetal gonads using a Wt1-BAC transgene system is sufficient to drive testicular differentiation and male secondary sex development. XX transgenic fetal gonads showed typical testicular size and vasculature. Key ovarian markers, including Wnt4 and Foxl2, were repressed. Sertoli cells expressing the hallmark testis-determining gene Sox9 were formed, although they did not assemble into normal testis cords. Other bipotential lineages differentiated into testicular cell types, including steroidogenic fetal Leydig cells and non-meiotic germ cells. As a consequence, male internal and external reproductive organs developed postnatally, with an absence of female reproductive tissues. These results reveal that Dmrt1 has retained its ability to act as the primary testis-determining trigger in mammals, even though this function is no longer normally required. Thus, Dmrt1 provides a common thread in the evolution of sex determination mechanisms in metazoans.


Subject(s)
Biological Evolution , Gene Expression Regulation, Developmental/physiology , Mice, Transgenic/metabolism , Sex Determination Processes/physiology , Sex Differentiation/physiology , Transcription Factors/metabolism , Animals , Chromosomes, Artificial, Bacterial , Cloning, Molecular , Female , Gene Expression Regulation, Developmental/genetics , Genetic Vectors/genetics , Germ Cells/cytology , Leydig Cells/cytology , Male , Mice , Mice, Transgenic/genetics , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Sex Determination Processes/genetics , Sex Differentiation/genetics , Testis/growth & development
18.
Hum Mol Genet ; 24(23): 6699-710, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26362256

ABSTRACT

Patients with 46,XY gonadal dysgenesis (GD) exhibit genital anomalies, which range from hypospadias to complete male-to-female sex reversal. However, a molecular diagnosis is made in only 30% of cases. Heterozygous mutations in the human FGFR2 gene cause various craniosynostosis syndromes including Crouzon and Pfeiffer, but testicular defects were not reported. Here, we describe a patient whose features we would suggest represent a new FGFR2-related syndrome, craniosynostosis with XY male-to-female sex reversal or CSR. The craniosynostosis patient was chromosomally XY, but presented as a phenotypic female due to complete GD. DNA sequencing identified the FGFR2c heterozygous missense mutation, c.1025G>C (p.Cys342Ser). Substitution of Cys342 by Ser or other amino acids (Arg/Phe/Try/Tyr) has been previously reported in Crouzon and Pfeiffer syndrome. We show that the 'knock-in' Crouzon mouse model Fgfr2c(C342Y/C342Y) carrying a Cys342Tyr substitution displays XY gonadal sex reversal with variable expressivity. We also show that despite FGFR2c-Cys342Tyr being widely considered a gain-of-function mutation, Cys342Tyr substitution in the gonad leads to loss of function, as demonstrated by sex reversal in Fgfr2c(C342Y/-) mice carrying the knock-in allele on a null background. The rarity of our patient suggests the influence of modifier genes which exacerbated the testicular phenotype. Indeed, patient whole exome analysis revealed several potential modifiers expressed in Sertoli cells at the time of testis determination in mice. In summary, this study identifies the first FGFR2 mutation in a 46,XY GD patient. We conclude that, in certain rare genetic contexts, maintaining normal levels of FGFR2 signaling is important for human testis determination.


Subject(s)
Craniosynostoses/genetics , Gonadal Dysgenesis, 46,XY/genetics , Mutation, Missense , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adolescent , Animals , Craniosynostoses/metabolism , DNA Mutational Analysis , Disease Models, Animal , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Mutant Strains , Syndrome
19.
Development ; 141(11): 2195-205, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24866114

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.


Subject(s)
Gene Expression Regulation, Developmental , Ovary/embryology , Sex-Determining Region Y Protein/physiology , Testis/embryology , Animals , Cell Lineage , Epigenesis, Genetic , Female , GATA4 Transcription Factor/metabolism , Humans , Male , Mice , Promoter Regions, Genetic , Steroidogenic Factor 1/metabolism , Transcription, Genetic , WT1 Proteins/metabolism , Y Chromosome
20.
Proc Natl Acad Sci U S A ; 111(32): 11768-73, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25074915

ABSTRACT

The mammalian sex-determining factor SRY comprises a conserved high-mobility group (HMG) box DNA-binding domain and poorly conserved regions outside the HMG box. Mouse Sry is unusual in that it includes a C-terminal polyglutamine (polyQ) tract that is absent in nonrodent SRY proteins, and yet, paradoxically, is essential for male sex determination. To dissect the molecular functions of this domain, we generated a series of Sry mutants, and studied their biochemical properties in cell lines and transgenic mouse embryos. Sry protein lacking the polyQ domain was unstable, due to proteasomal degradation. Replacing this domain with irrelevant sequences stabilized the protein but failed to restore Sry's ability to up-regulate its key target gene SRY-box 9 (Sox9) and its sex-determining function in vivo. These functions were restored only when a VP16 transactivation domain was substituted. We conclude that the polyQ domain has important roles in protein stabilization and transcriptional activation, both of which are essential for male sex determination in mice. Our data disprove the hypothesis that the conserved HMG box domain is the only functional domain of Sry, and highlight an evolutionary paradox whereby mouse Sry has evolved a novel bifunctional module to activate Sox9 directly, whereas SRY proteins in other taxa, including humans, seem to lack this ability, presumably making them dependent on partner proteins(s) to provide this function.


Subject(s)
Genes, sry , Sex Determination Processes , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Animals , Evolution, Molecular , Female , Genes, Reporter , Male , Mice , Mice, Transgenic , Mutagenesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptides/chemistry , Pregnancy , Proteasome Endopeptidase Complex/metabolism , Protein Denaturation , Protein Stability , Protein Structure, Tertiary , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sequence Deletion , Sex-Determining Region Y Protein/chemistry , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL