Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
FASEB J ; 37(8): e23073, 2023 08.
Article in English | MEDLINE | ID: mdl-37402125

ABSTRACT

In female mammals, the oviduct and uterus are essential sites for female and male gamete transport, fertilization, implantation, and maintenance of a successful pregnancy. To delineate the reproductive function of Mothers against decapentaplegic homolog 4 (Smad4), we specifically inactivated Smad4 in ovarian granulosa cells and, oviduct and uterine mesenchymal cells using the Amhr2-cre mouse line. Deletion of exon 8 of Smad4 results in the production of an MH2-truncated SMAD4 protein. These mutant mice are infertile due to the development of oviductal diverticula and defects during the implantation process. The ovaries are fully functional as demonstrated in an ovary transfer experiment. The development of oviductal diverticula occurs shortly after puberty and is dependent on estradiol. The diverticula interfere with sperm migration and embryo transit to the uterus, reducing the number of implantation sites. Analysis of the uterus shows that, even if implantation occurs, decidualization and vascularization are defective resulting in embryo resorption as early as the seventh day of pregnancy. Thus, Smad4 plays an important function in female reproduction by controlling the structural and functional integrity of the oviduct and uterus.


Subject(s)
Estradiol , Smad4 Protein , Animals , Female , Humans , Male , Mice , Pregnancy , Embryo Implantation , Estradiol/metabolism , Mammals/metabolism , Oviducts/metabolism , Semen/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Uterus/metabolism
2.
Dev Biol ; 483: 58-65, 2022 03.
Article in English | MEDLINE | ID: mdl-34965385

ABSTRACT

The conserved 3'-5' exoribonuclease EXOSC10/Rrp6 is required for gametogenesis, brain development, erythropoiesis and blood cell enhancer function. The human ortholog is essential for mitosis in cultured cancer cells. Little is known, however, about the role of Exosc10 during embryo development and organogenesis. We generated an Exosc10 knockout model and find that Exosc10-/- mice show an embryonic lethal phenotype. We demonstrate that Exosc10 maternal wild type mRNA is present in mutant oocytes and that the gene is expressed during all stages of early embryogenesis. Furthermore, we observe that EXOSC10 early on localizes to the periphery of nucleolus precursor bodies in blastomeres, which is in keeping with the protein's role in rRNA processing and may indicate a function in the establishment of chromatin domains during initial stages of embryogenesis. Finally, we infer from genotyping data for embryonic days e7.5, e6.5 and e4.5 and embryos cultured in vitro that Exosc10-/- mutants arrest at the eight-cell embryo/morula transition. Our results demonstrate a novel essential role for Exosc10 during early embryogenesis, and they are consistent with earlier work showing that impaired ribosome biogenesis causes a developmental arrest at the morula stage.


Subject(s)
Blastocyst/metabolism , Embryonic Development/genetics , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Morula/metabolism , Signal Transduction/genetics , Animals , Blastomeres/metabolism , Cell Nucleolus/metabolism , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Oocytes/metabolism , Phenotype , RNA Processing, Post-Transcriptional/genetics , RNA, Ribosomal/metabolism , Ribosomes/metabolism
4.
Biol Reprod ; 92(3): 71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25609838

ABSTRACT

Genome-wide RNA profiling studies have identified hundreds of transcripts that are highly expressed in mammalian male germ cells, including many that are undetectable in somatic control tissues. Among them, genes important for spermatogenesis are significantly enriched. Information about mRNAs and their cognate proteins facilitates the identification of novel conserved target genes for functional studies in the mouse. By inspecting genome-wide RNA profiling data, we manually selected 81 genes for which RNA is detected almost exclusively in the human male germline and, in most cases, in rodent testicular germ cells. We observed corresponding mRNA/protein patterns in 43 cases using immunohistochemical data from the Human Protein Atlas and large-scale human protein profiling data obtained via mass spectroscopy. Protein network information enabled us to establish an interaction map of 38 proteins that points to potentially important testicular roles for some of them. We further characterized six candidate genes at the protein level in the mouse. We conclude that conserved genes induced in testis tend to show similar mRNA/protein expression patterns across species. Specifically, our results suggest roles during embryogenesis and adult spermatogenesis for Foxr1 and Sox30 and during spermiogenesis and fertility for Fam71b, 1700019N19Rik, Hmgb4, and Zfp597.


Subject(s)
Gene Expression Profiling , Gene Regulatory Networks/genetics , Genome-Wide Association Study , Protein Array Analysis , RNA, Messenger/genetics , Spermatogenesis/genetics , Amino Acid Sequence , Animals , Fertility/genetics , Humans , Male , Mice , Molecular Sequence Data , Species Specificity , Transcription Factors/genetics
5.
Development ; 138(10): 1967-75, 2011 May.
Article in English | MEDLINE | ID: mdl-21490063

ABSTRACT

During male sexual differentiation, the transforming growth factor-ß (TGF-ß) signaling molecule anti-Müllerian hormone (AMH; also known as Müllerian inhibiting substance, MIS) is secreted by the fetal testes and induces regression of the Müllerian ducts, the primordia of the female reproductive tract organs. Currently, the molecular identity of downstream events regulated by the AMH signaling pathway remains unclear. We found that male-specific Wnt4 expression in mouse Müllerian duct mesenchyme depends upon AMH signaling, implicating the WNT pathway as a downstream mediator of Müllerian duct regression. Inactivation of ß-catenin, a mediator of the canonical WNT pathway, did not affect AMH signaling activation in the Müllerian duct mesenchyme, but did block Müllerian duct regression. These data suggest that ß-catenin mediates AMH signaling for Müllerian duct regression during male sexual differentiation.


Subject(s)
Mullerian Ducts/embryology , Sex Differentiation/physiology , Testis/embryology , beta Catenin/physiology , Animals , Anti-Mullerian Hormone/physiology , Base Sequence , DNA Primers/genetics , Female , Gene Expression Regulation, Developmental , Genitalia, Female/embryology , Genitalia, Female/metabolism , Male , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Mullerian Ducts/metabolism , Pregnancy , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Receptors, Peptide/physiology , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Sex Differentiation/genetics , Signal Transduction , Testis/metabolism , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/physiology , Wnt4 Protein , beta Catenin/deficiency , beta Catenin/genetics
6.
Nat Genet ; 32(3): 408-10, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368913

ABSTRACT

Elimination of the developing female reproductive tract in male fetuses is an essential step in mammalian sexual differentiation. In males, the fetal testis produces the transforming growth factor beta (TGF-beta) family member anti-Müllerian hormone (Amh, also known as Müllerian-inhibiting substance (Mis)), which causes regression of the Müllerian ducts, the primordia of the oviducts, uterus and upper vagina. Amh induces regression by binding to a specific type II receptor (Amhr2) expressed in the mesenchyme surrounding the ductal epithelium. Mutations in AMH or AMHR2 in humans and mice disrupt signaling, producing male pseudohermaphrodites that possess oviducts and uteri. The type I receptor and Smad proteins that are required in vivo for Müllerian duct regression have not yet been identified. Here we show that targeted disruption of the widely expressed type I bone morphogenetic protein (BMP) receptor Bmpr1a (also known as Alk3) in the mesenchymal cells of the Müllerian ducts leads to retention of oviducts and uteri in males. These results identify Bmpr1a as a type I receptor for Amh-induced regression of Müllerian ducts. Because Bmpr1a is evolutionarily conserved, these findings indicate that a component of the BMP signaling pathway has been co-opted during evolution for male sexual development in amniotes.


Subject(s)
Activin Receptors, Type I/genetics , Activin Receptors, Type I/physiology , Mullerian Ducts/pathology , Mutation , Protein Serine-Threonine Kinases , Receptors, Growth Factor , Sex Differentiation , Testis/embryology , Alleles , Animals , Bone Morphogenetic Protein Receptors, Type I , Exons , In Situ Hybridization , Ligands , Male , Mice , Mice, Transgenic , Models, Genetic , Mullerian Ducts/physiology , Recombination, Genetic , Time Factors
7.
Int J Biol Sci ; 19(4): 1080-1093, 2023.
Article in English | MEDLINE | ID: mdl-36923944

ABSTRACT

EXOSC10 is a catalytic subunit of the nuclear RNA exosome, and possesses a 3'-5' exoribonuclease activity. The enzyme processes and degrades different classes of RNAs. To delineate the role of EXOSC10 during oocyte growth, specific Exosc10 inactivation was performed in oocytes from the primordial follicle stage onward using the Gdf9-iCre; Exosc10 f/- mouse model (Exosc10 cKO(Gdf9)). Exosc10 cKO(Gdf9) female mice are infertile. The onset of puberty and the estrus cycle in mutants are initially normal and ovaries contain all follicle classes. By the age of eight weeks, vaginal smears reveal irregular estrus cycles and mutant ovaries are completely depleted of follicles. Mutant oocytes retrieved from the oviduct are degenerated, and occasionally show an enlarged polar body, which may reflect a defective first meiotic division. Under fertilization conditions, the mutant oocytes do not enter into an embryonic development process. Furthermore, we conducted a comparative proteome analysis of wild type and Exosc10 knockout mouse ovaries, and identified EXOSC10-dependent proteins involved in many biological processes, such as meiotic cell cycle progression and oocyte maturation. Our results unambiguously demonstrate an essential role for EXOSC10 in oogenesis and may serve as a model for primary ovarian insufficiency in humans. Data are available via ProteomeXchange with identifier PXD039417.


Subject(s)
Biological Phenomena , Ovarian Reserve , Animals , Female , Humans , Infant , Mice , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Oocytes/metabolism , Oogenesis/genetics
8.
Front Cell Dev Biol ; 10: 1006087, 2022.
Article in English | MEDLINE | ID: mdl-36313563

ABSTRACT

A transgenic mouse approach using bacterial artificial chromosomes (BAC) was used to identify regulatory regions that direct Müllerian duct expression for Amhr2 and Osterix (Osx, also known as Sp7). Amhr2 encodes the receptor that mediates anti-Müllerian hormone (AMH) signaling for Müllerian duct regression in male embryos. Amhr2 is expressed in the Müllerian duct mesenchyme of both male and female embryos. A ∼147-kb BAC clone containing the Amhr2 locus was used to generate transgenic mice. The transgene was able to rescue the block in Müllerian duct regression of Amhr2-null males, suggesting that the BAC clone contains regulatory sequences active in male embryos. Osx is expressed in the developing skeleton of male and female embryos but is also an AMH-induced gene that is expressed in the Müllerian duct mesenchyme exclusively in male embryos. Osx-Cre transgenic mice were previously generated using a ∼204-kb BAC clone. Crosses of Osx-Cre mice to Cre-dependent lacZ reporter mice resulted in reporter expression in the developing skeleton and in the Müllerian duct mesenchyme of male but not female embryos. Osx-Cherry transgenic mice were previously generated using a 39-kb genomic region surrounding the Osx locus. Osx-Cherry embryos expressed red fluorescence in the developing skeleton and Müllerian duct mesenchyme of male but not female embryos. In addition, female Osx-Cherry embryos ectopically expressing human AMH from an Mt1-AMH transgene activated red fluorescence in the Müllerian duct mesenchyme. These results suggest that the 39-kb region used to generate Osx-Cherry contains male-specific Müllerian duct mesenchyme regulatory sequences that are responsive to AMH signaling. These BAC transgenic mouse approaches identify two distinct regions that direct Müllerian duct mesenchyme expression and contribute fundamental knowledge to define a gene regulatory network for sex differentiation.

9.
Mol Oncol ; 15(11): 3003-3023, 2021 11.
Article in English | MEDLINE | ID: mdl-33426787

ABSTRACT

Cancer/Testis (CT) genes are induced in germ cells, repressed in somatic cells, and derepressed in somatic tumors, where these genes can contribute to cancer progression. CT gene identification requires data obtained using standardized protocols and technologies. This is a challenge because data for germ cells, gonads, normal somatic tissues, and a wide range of cancer samples stem from multiple sources and were generated over substantial periods of time. We carried out a GeneChip-based RNA profiling analysis using our own data for testis and enriched germ cells, data for somatic cancers from the Expression Project for Oncology, and data for normal somatic tissues from the Gene Omnibus Repository. We identified 478 candidate loci that include known CT genes, numerous genes associated with oncogenic processes, and novel candidates that are not referenced in the Cancer/Testis Database (www.cta.lncc.br). We complemented RNA expression data at the protein level for SPESP1, GALNTL5, PDCL2, and C11orf42 using cancer tissue microarrays covering malignant tumors of breast, uterus, thyroid, and kidney, as well as published RNA profiling and immunohistochemical data provided by the Human Protein Atlas (www.proteinatlas.org). We report that combined RNA/tissue profiling identifies novel CT genes that may be of clinical interest as therapeutical targets or biomarkers. Our findings also highlight the challenges of detecting truly germ cell-specific mRNAs and the proteins they encode in highly heterogenous testicular, somatic, and tumor tissues.


Subject(s)
N-Acetylgalactosaminyltransferases , Testicular Neoplasms , Gene Expression Profiling , Humans , Male , Oligonucleotide Array Sequence Analysis , RNA
10.
Carcinogenesis ; 29(11): 2062-72, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18687666

ABSTRACT

The mechanisms of granulosa cell tumor (GCT) development may involve the dysregulation of signaling pathways downstream of follicle-stimulating hormone, including the phosphoinosite-3 kinase (PI3K)/AKT pathway. To test this hypothesis, a genetically engineered mouse model was created to derepress the PI3K/AKT pathway in granulosa cells by conditional targeting of the PI3K antagonist gene Pten (Pten(flox/flox);Amhr2(cre/+)). The majority of Pten(flox/flox);Amhr2(cre/+) mice featured no ovarian anomalies, but occasionally ( approximately 7%) developed aggressive, anaplastic GCT with pulmonary metastases. The expression of the PI3K/AKT downstream effector FOXO1 was abrogated in Pten(flox/flox);Amhr2(cre/+) GCT, indicating a mechanism by which GCT cells may increase proliferation and evade apoptosis. To relate these findings to spontaneously occurring GCT, analyses of PTEN and phospho-AKT expression were performed on human and equine tumors. Although PTEN loss was not detected, many GCT (2/5 human, 7/17 equine) featured abnormal nuclear or perinuclear localization of phospho-AKT, suggestive of altered PI3K/AKT activity. As inappropriate activation of WNT/CTNNB1 signaling causes late-onset GCT development and cross talk between the PI3K/AKT and WNT/CTNNB1 pathways has been reported, we tested whether these pathways could synergize in GCT. Activation of both the PI3K/AKT and WNT/CTNNB1 pathways in the granulosa cells of a mouse model (Pten(flox/flox);Ctnnb1(flox(ex3)/+);Amhr2(cre/+)) resulted in the development of GCT similar to those observed in Pten(flox/flox);Amhr2(cre/+) mice, but with 100% penetrance, perinatal onset, extremely rapid growth and the ability to spread by seeding into the abdominal cavity. These data indicate a synergistic effect of dysregulated PI3K/AKT and WNT/CTNNB1 signaling in the development and progression of GCT and provide the first animal models for metastatic GCT.


Subject(s)
Granulosa Cell Tumor/metabolism , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Base Sequence , DNA Primers , Disease Progression , Female , Granulosa Cell Tumor/pathology , Immunohistochemistry , Mice , Neoplasm Metastasis , Ovarian Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism
11.
J Clin Endocrinol Metab ; 93(11): 4456-61, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18697861

ABSTRACT

CONTEXT: In the polycystic ovary syndrome (PCOS), in addition to intrinsic thecal dysregulation leading to hyperandrogenism, a granulosa cell (GC) dysregulation may occur. Expression of anti-Müllerian hormone (AMH), FSH receptor (FSHR) and androgen receptor (AR) are suspected to be altered in PCOS GCs. DESIGN: The aim of this prospective study was to analyze the expression of these genes at the last stages of follicular maturation in GCs from 17 patients with PCOS and 15 controls undergoing controlled ovarian hyperstimulation during a cycle with in vitro fertilization. MATERIALS AND METHODS: On the day of oocyte retrieval, follicular fluids were collected from small follicles (SF; 8-13 mm) and large follicles (17-22 mm) in separate tubes. Total RNAs and proteins were extracted from GCs. Reverse transcription was performed and quantification of gene expression levels was achieved by real-time quantitative PCR. RESULTS: AMH and FSHR mRNA levels were significantly higher in PCOS than in controls in GCs from both SF and large follicles. Likewise, AR and AMH receptor II mRNA levels in GCs from SF were significantly higher in PCOS compared with controls. In both PCOS patients and controls, AMH and AR mRNA levels correlated strongly, positively, and independently to FSHR mRNA levels. CONCLUSION: Using quantitative RT-PCR, AMH, AMH receptor II, FSHR, and AR genes were shown to be overexpressed by GCs from stimulated follicles of women with PCOS undergoing controlled ovarian hyperstimulation. This could be the sign of a maturation defect or may reflect hyperandrogenism.


Subject(s)
Anti-Mullerian Hormone/genetics , Gene Expression Regulation , Granulosa Cells/physiology , Polycystic Ovary Syndrome/genetics , Receptors, Androgen/genetics , Receptors, FSH/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Anovulation/etiology , Female , Fertilization in Vitro/methods , Humans , Ovarian Follicle/physiology , Ovarian Follicle/physiopathology , Ovulation , Ovulation Induction/methods , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/diagnostic imaging , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Ultrasonography , Young Adult
12.
Mol Reprod Dev ; 75(7): 1154-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18213646

ABSTRACT

The Müllerian ducts give rise to the female reproductive tract, including the Fallopian tubes, uterus, cervix, and anterior vagina. In male embryos, the Müllerian ducts regress, preventing the formation of female organs. We introduced the bacterial lacZ gene, encoding beta-galactosidase (beta-gal), into the AMHR-II locus (Amhr2) by gene targeting in mouse embryonic stem (ES) cells to mark Müllerian duct differentiation and regression. We show that Amhr2-lacZ heterozygotes express beta-gal activity in an Amhr2-specific pattern. In the gonads, beta-gal activity was detected in Sertoli cells of the testes from 2 weeks after birth, and fetal ovaries and granulosa cells of the adult ovary. beta-gal activity was first detected in the rostral mesenchyme of the Müllerian ducts at 12.5 days post coitus (dpc) in both sexes but soon thereafter expression was found along the entire length of the Müllerian ducts with higher levels initially found in males. In females, beta-gal activity was restricted to one side of the ductal mesoepithelium, whereas in males beta-gal expression encircled the duct. beta-gal activity was also detected in the coelomic epithelium at 13.5 and 14.5 dpc. In male embryos, mesenchymal beta-gal activity permitted the visualization of the temporal and spatial pattern of Müllerian duct regression. This pattern was similar to that observed using a Müllerian duct mesoepithelium lacZ reporter, indicating a coordinated loss of Müllerian duct mesoepithelium and Amhr2-expressing mesenchyme.


Subject(s)
Mesoderm/cytology , Mullerian Ducts/cytology , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , beta-Galactosidase/genetics , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Genetic Vectors , Male , Mice , Mice, Transgenic , Sex Characteristics
13.
Cancer Res ; 65(20): 9206-15, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230381

ABSTRACT

Misregulation of the Wnt/beta-catenin signaling pathway is a hallmark of several forms of cancer. Components of the Wnt/beta-catenin pathway are expressed in ovarian granulosa cells; nevertheless, its potential involvement in granulosa cell tumorigenesis has not been examined. To this end, human (n = 6) and equine (n = 18) granulosa cell tumors (GCT) were analyzed for beta-catenin expression by immunohistochemistry. Unlike granulosa cells of normal ovaries, most (15 of 24) GCT samples showed nuclear localization of beta-catenin, suggesting that activation of the Wnt/beta-catenin pathway plays a role in the etiology of GCT. To confirm this hypothesis, Catnb(flox(ex3)/+); Amhr2(cre/+) mice that express a dominant stable beta-catenin mutant in their granulosa cells were generated. These mice developed follicle-like structures containing disorganized, pleiomorphic granulosa by 6 weeks of age. Even in older mice, these follicle-like lesions grew no larger than the size of antral follicles and contained very few proliferating cells. Similar to corpora lutea, the lesions were highly vascularized, although they did not express the luteinization marker Cyp11a1. Catnb(flox(ex3)/+); Amhr2(cre/+) females were also found to be severely subfertile, and fewer corpora lutea were found to form in response to exogenous gonadotropin compared with control mice. In older mice, the ovarian lesions often evolved into GCT, indicating that they represent a pretumoral intermediate stage. The GCT in Catnb(flox(ex3)/+); Amhr2(cre/+) mice featured many histopathologic similarities to the human disease, and prevalence of tumor development attained 57% at 7.5 months of age. Together, these studies show a causal link between misregulated Wnt/beta-catenin signaling and GCT development and provide a novel model system for the study of GCT biology.


Subject(s)
Granulosa Cell Tumor/metabolism , Ovarian Neoplasms/metabolism , Wnt Proteins/biosynthesis , beta Catenin/biosynthesis , Animals , Disease Models, Animal , Female , Granulosa Cell Tumor/genetics , Granulosa Cell Tumor/pathology , Horses , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta , Signal Transduction , Wnt Proteins/genetics , beta Catenin/genetics
14.
Sci Rep ; 7(1): 15065, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118343

ABSTRACT

EXOSC10 is a catalytic subunit of the exosome that processes biologically active transcripts, degrades aberrant mRNAs and targets certain long non-coding RNAs (lncRNAs). The yeast orthologue Rrp6 is required for efficient growth and gametogenesis, and becomes unstable during meiosis. However, nothing is known about the localization, stability and function of EXOSC10 in the rodent male germline. We detect the protein in nucleoli and the cytoplasm of mitotic and meiotic germ cells, and find that it transiently associates with the XY body, a structure targeted by meiotic sex chromosome inactivation (MSCI). Finally, EXOSC10 becomes unstable at later stages of gamete development. To determine Exosc10's meiotic function, we inactivated the gene specifically in male germ cells using cre recombinase controlled by Stra8 or Ddx4/Vasa promoters. Mutant mice have small testes, show impaired germ cell differentiation and are subfertile. Our results demonstrate that EXOSC10 is post-translationally regulated in germ cells, associate the protein with epigenetic chromosome silencing, and reveal its essential role in germ cell growth and development.


Subject(s)
Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Spermatogenesis/genetics , Animals , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Female , Fertility/genetics , Male , Meiosis/genetics , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Testis/metabolism
15.
Oncogene ; 24(31): 4984-92, 2005 Jul 21.
Article in English | MEDLINE | ID: mdl-15897891

ABSTRACT

Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-beta family responsible for regression of Müllerian ducts during male sexual differentiation and for regulation of gonadal steroidogenesis. AMH is also a gonadal tumor suppressor which mediates its effects through a specific type II receptor and the bone morphogenetic protein (BMP)-specific Smad proteins, suggesting that AMH and BMPs could also share type I receptors, namely activin-like kinases (ALKs)2, 3 or 6. However, attempts to identify a unique AMH type I receptor among them were unsuccessful. Here, using kinase-deficient type I receptors and small interfering RNA technology, we demonstrate that, in an AMH Sertoli target cell line, ALK3 mediates AMH effects on both Smad1 activation and P450 side-chain cleavage enzyme. In addition, transfecting a combination of normal and kinase-deficient receptors, we show that ALK2 can compensate for the absence of ALK3 and probably acts in synergy with ALK3 at high concentrations of AMH to activate Smad1, whereas ALK6 has a competitive inhibitory effect. These results are a first step in understanding how AMH transduces its effects in immature Sertoli cells.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Peptide/physiology , Sertoli Cells/physiology , Animals , Base Sequence , Bone Morphogenetic Protein Receptors, Type I , CHO Cells , Cell Line , Cricetinae , DNA Primers , Gene Expression Regulation , Humans , Male , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Receptors, Growth Factor/metabolism , Receptors, Transforming Growth Factor beta , Recombinant Proteins/metabolism , Transcription Factors/metabolism
16.
Int J Biol Sci ; 12(6): 667-76, 2016.
Article in English | MEDLINE | ID: mdl-27194944

ABSTRACT

Müllerian duct regression is a complex process which involves the AMH signalling pathway. We have previously demonstrated that besides AMH and its specific type II receptor (AMHRII), BMPR-IA and Smad5 are two essential factors implicated in this mechanism. Mothers against decapentaplegic homolog 4 (Smad4) is a transcription factor and the common Smad (co-Smad) involved in transforming growth factor beta (TGF-ß) signalling pathway superfamily. Since Smad4 null mutants die early during gastrulation, we have inactivated Smad4 in the Müllerian duct mesenchyme. Specific inactivation of Smad4 in the urogenital ridge leads to the partial persistence of the Müllerian duct in adult male mice. Careful examination of the urogenital tract reveals that the Müllerian duct retention is randomly distributed either on one side or both sides. Histological analysis shows a uterus-like structure, which is confirmed by the expression of estrogen receptor α. As previously described in a ß-catenin conditional mutant mouse model, ß-catenin contributes to Müllerian duct regression. In our mutant male embryos, it appears that ß-catenin expression is locally reduced along the urogenital ridge as compared to control mice. Moreover, the expression pattern is similar to those observed in control female mice. This study shows that reduced Smad4 expression disrupts the Wnt/ß-catenin signalling leading to the partial persistence of Müllerian duct.


Subject(s)
Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Smad4 Protein/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Exons/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Signal Transduction/genetics , Signal Transduction/physiology , Smad4 Protein/genetics , Smad5 Protein/genetics , Smad5 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
17.
Nat Commun ; 7: 10055, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26753790

ABSTRACT

Anti-Müllerian hormone (AMH) plays crucial roles in sexual differentiation and gonadal functions. However, the possible extragonadal effects of AMH on the hypothalamic-pituitary-gonadal axis remain unexplored. Here we demonstrate that a significant subset of GnRH neurons both in mice and humans express the AMH receptor, and that AMH potently activates the GnRH neuron firing in mice. Combining in vivo and in vitro experiments, we show that AMH increases GnRH-dependent LH pulsatility and secretion, supporting a central action of AMH on GnRH neurons. Increased LH pulsatility is an important pathophysiological feature in many cases of polycystic ovary syndrome (PCOS), the most common cause of female infertility, in which circulating AMH levels are also often elevated. However, the origin of this dysregulation remains unknown. Our findings raise the intriguing hypothesis that AMH-dependent regulation of GnRH release could be involved in the pathophysiology of fertility and could hold therapeutic potential for treating PCOS.


Subject(s)
Anti-Mullerian Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Follicle Stimulating Hormone/metabolism , Gene Knock-In Techniques , Humans , Hypothalamus/cytology , Immunohistochemistry , In Vitro Techniques , Luteinizing Hormone/metabolism , Mice , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Endocrinol ; 18(4): 953-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14701941

ABSTRACT

Follistatin plays an important role in female physiology by regulating FSH levels through blocking activin actions. Failure to regulate FSH has been implicated as a potential cause of premature ovarian failure. Premature ovarian failure is characterized by amenorrhea, infertility, and elevated gonadotropin levels in women under the age of 40. Because follistatin is essential for postnatal viability, we designed a cre/loxP conditional knockout system to render the follistatin gene null specifically in the granulosa cells of the postnatal ovary using Amhr2cre transgenic mice. The follistatin conditional knockout females develop fertility defects, including reduced litter number and litter sizes and, in the most severe case, infertility. Reduced numbers of ovarian follicles, ovulation and fertilization defects, elevated levels of serum FSH and LH, and reduced levels of testosterone were observed in these mice. These findings demonstrate that compromising granulosa cell follistatin function leads to findings similar to those characterized in premature ovarian failure. Follistatin conditional knockouts may therefore be a useful model with which to further study this human syndrome. These studies are the first report of a granulosa cell-specific deletion of a gene in the postnatal ovary and have important implications for future endeavors to generate ovary-specific knockout mouse models.


Subject(s)
Follistatin/metabolism , Granulosa Cells/metabolism , Infertility, Female/metabolism , Animals , Female , Infertility, Female/etiology , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Ovary/metabolism
19.
Mol Endocrinol ; 18(7): 1610-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15118069

ABSTRACT

Knockout (KO) mice lacking the orphan nuclear receptor steroidogenic factor 1 (SF-1, officially designated Nr5a1) have a compound endocrine phenotype that includes adrenal and gonadal agenesis, impaired expression of pituitary gonadotropins, and structural abnormalities of the ventromedial hypothalamic nucleus. To inactivate a conditional SF-1 allele in the gonads, we targeted the expression of Cre recombinase with a knock-in allele of the anti-Müllerian hormone type 2 receptor locus. In testes, Cre was expressed in Leydig cells. The testes of adult gonad-specific SF-1 KO mice remained at the level of the bladder and were markedly hypoplastic, due at least partly to impaired spermatogenesis. Histological abnormalities of the testes were seen from early developmental stages and were associated with markedly decreased Leydig cell expression of two essential components of testosterone biosynthesis, Cyp11a and the steroidogenic acute regulatory protein. In females, the anti-Müllerian hormone type 2 receptor-Cre allele directed Cre expression to granulosa cells. Although wild-type and SF-1 KO ovaries were indistinguishable during embryogenesis and at birth, adult females were sterile and their ovaries lacked corpora lutea and contained hemorrhagic cysts resembling those in estrogen receptor alpha and aromatase KO mice. Collectively, these studies establish definitively that SF-1 expression in the gonads is essential for normal reproductive development and function.


Subject(s)
DNA-Binding Proteins/physiology , Leydig Cells/pathology , Ovary/physiology , Testis/physiology , Transcription Factors/physiology , Animals , Cell Proliferation , Cholesterol Side-Chain Cleavage Enzyme/genetics , Corpus Luteum/abnormalities , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Granulosa Cells/physiology , Homeodomain Proteins , Infertility/genetics , Integrases/genetics , Leydig Cells/physiology , Male , Mice , Mice, Knockout , Ovary/pathology , Phosphoproteins/genetics , Receptors, Cytoplasmic and Nuclear , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta , Sex Differentiation/genetics , Steroidogenic Factor 1 , Testis/abnormalities , Testis/cytology , Transcription Factors/genetics
20.
Theriogenology ; 84(8): 1397-404, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26298408

ABSTRACT

Plasma anti-Müllerian hormone (AMH) concentrations have been recently found to be predictive of the number of embryos recovered after FSH superovulatory treatment in the cow. However, the sensitivity of the Active Müllerian-inhibiting substance/AMH ELISA (ref. 10-14400; DSL-Beckman-Coulter) used to make these measurements in bovine plasma samples is low because it was developed to measure human AMH levels. To overcome this limitation, we developed an immunoassay specific for the bovine (B), ovine (O), and caprine (C) species, the bovine-ovine-caprine (BOC) ELISA. For this purpose, we produced recombinant bovine AMH for standardization, and we used monoclonal antibodies raised against bovine AMH, previously prepared by our laboratory. We evaluated the precision, accuracy, specificity, limit of detection, and functional sensitivity of the assay. The intra-assay coefficient of variation ranged between 3.4% and 11.3% for AMH concentrations between 23.68 and 1.74 ng/mL, and the interassay coefficient of variation ranged between 4.8% and 20.5% for concentrations between 25.53 and 1.42 ng/mL, respectively. The assay displayed a good linearity, had a detection limit of 0.4 ng/mL and a functional sensitivity of 1.4 ng/mL. It also cross-reacted with ovine and caprine AMHs. Both the mean and median AMH levels measured in 40 cow plasma samples using the BOC ELISA were approximately 44 fold higher than the mean and median AMH levels measured with the Active Müllerian-inhibiting substance/AMH ELISA. Moreover, a higher correlation was observed between the average number of embryos recovered from each cow after superovulatory treatment and AMH concentrations measured with the BOC ELISA. This BOC ELISA provides a very efficient tool for evaluating the ovarian follicular reserve of cows and predicting their embryo production capacity.


Subject(s)
Anti-Mullerian Hormone/blood , Embryonic Development , Enzyme-Linked Immunosorbent Assay/veterinary , Animals , Cattle , Female , Ovarian Function Tests/methods , Ovarian Function Tests/veterinary , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL