Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 298(1): 101429, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34801555

RESUMO

Noncovalent complexes of transforming growth factor-ß family growth/differentiation factors with their prodomains are classified as latent or active, depending on whether the complexes can bind their respective receptors. For the anti-Müllerian hormone (AMH), the hormone-prodomain complex is active, and the prodomain is displaced upon binding to its type II receptor, AMH receptor type-2 (AMHR2), on the cell surface. However, the mechanism by which this displacement occurs is unclear. Here, we used ELISA assays to measure the dependence of prodomain displacement on AMH concentration and analyzed results with respect to the behavior expected for reversible binding in combination with ligand-induced receptor dimerization. We found that, in solution, the prodomain has a high affinity for the growth factor (GF) (Kd = 0.4 pM). Binding of the AMH complex to a single AMHR2 molecule does not affect this Kd and does not induce prodomain displacement, indicating that the receptor binding site in the AMH complex is fully accessible to AMHR2. However, recruitment of a second AMHR2 molecule to bind the ligand bivalently leads to a 1000-fold increase in the Kd for the AMH complex, resulting in rapid release of the prodomain. Displacement occurs only if the AMHR2 is presented on a surface, indicating that prodomain displacement is caused by a conformational change in the GF induced by bivalent binding to AMHR2. In addition, we demonstrate that the bone morphogenetic protein 7 prodomain is displaced from the complex with its GF by a similar process, suggesting that this may represent a general mechanism for receptor-mediated prodomain displacement in this ligand family.


Assuntos
Hormônio Antimülleriano , Hormônios Peptídicos , Hormônio Antimülleriano/metabolismo , Ligantes , Hormônios Peptídicos/metabolismo , Domínios Proteicos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Hum Mol Genet ; 28(19): 3211-3218, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31238341

RESUMO

The persistent Müllerian duct syndrome (PMDS) is a 46,XY disorder of sexual development characterized by the persistence of Müllerian duct derivatives, uterus and tubes, in otherwise normally masculinized males. The condition, transmitted as a recessive autosomal trait, is usually due to mutations in either the anti-Müllerian hormone (AMH) gene or its main receptor. Many variants of these genes have been described, all targeting the coding sequences. We report the first case of PMDS due to a regulatory mutation. The AMH promoter contains two binding sites for steroidogenic factor 1 (SF1), one at -102 and the other at -228. Our patient carries a single base deletion at -225, significantly decreasing its capacity for binding SF1, as measured by the electrophoresis mobility shift assay. Furthermore, by linking the AMH promoter to the luciferase gene, we show that the transactivation capacity of the promoter is significantly decreased by the mutation, in contrast to the disruption of the -102 binding site. To explain the difference in impact we hypothesize that SF1 could partially overcome the lack of binding to the -102 binding site by interacting with a GATA4 molecule linked to a nearby response element. We show that disruption of both the -102 SF1 and the -84 GATA response elements significantly decreases the transactivation capacity of the promoter. In conclusion, we suggest that the distance between mutated SF1 sites and potentially rescuing GATA binding motifs might play a role in the development of PMDS.


Assuntos
Hormônio Antimülleriano/química , Hormônio Antimülleriano/metabolismo , Transtorno 46,XY do Desenvolvimento Sexual/genética , Mutação , Fatores de Processamento de RNA/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Hormônio Antimülleriano/genética , Sítios de Ligação/genética , Linhagem Celular , Criança , Pré-Escolar , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Regiões Promotoras Genéticas , Ligação Proteica
3.
Cell Mol Life Sci ; 77(6): 1177-1196, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31327046

RESUMO

In mammalian ovaries, the theca layers of growing follicles are critical for maintaining their structural integrity and supporting androgen synthesis. Through combining the postnatal monitoring of ovaries by abdominal magnetic resonance imaging, endocrine profiling, hormonal analysis of the follicular fluid of growing follicles, and transcriptomic analysis of follicular theca cells, we provide evidence that the exposure of ovine fetuses to testosterone excess activates postnatal follicular growth and strongly affects the functions of follicular theca in adulthood. Prenatal exposure to testosterone impaired androgen synthesis in the small antral follicles of adults and affected the expression in their theca cells of a wide array of genes encoding extracellular matrix components, their membrane receptors, and signaling pathways. Most expression changes were uncorrelated with the concentrations of gonadotropins, steroids, and anti-Müllerian hormone in the recent hormonal environment of theca cells, suggesting that these changes rather result from the long-term developmental effects of testosterone on theca cell precursors in fetal ovaries. Disruptions of the extracellular matrix structure and signaling in the follicular theca and ovarian cortex can explain the acceleration of follicle growth through altering the stiffness of ovarian tissue. We propose that these mechanisms participate in the etiology of the polycystic ovarian syndrome, a major reproductive pathology in woman.


Assuntos
Síndrome do Ovário Policístico/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Testosterona/metabolismo , Células Tecais/metabolismo , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Folículo Ovariano/citologia , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Síndrome do Ovário Policístico/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Ovinos , Células Tecais/citologia , Células Tecais/ultraestrutura
4.
Biol Reprod ; 99(6): 1303-1312, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29985989

RESUMO

Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.


Assuntos
Androgênios/farmacologia , Hormônio Antimülleriano/metabolismo , Células de Sertoli/fisiologia , Fator Esteroidogênico 1/metabolismo , Animais , Hormônio Antimülleriano/genética , Linhagem Celular , Imunoprecipitação da Cromatina , Regulação para Baixo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Regiões Promotoras Genéticas , Receptores Androgênicos/metabolismo , Fator Esteroidogênico 1/genética , Transcriptoma
5.
J Cell Sci ; 128(7): 1352-64, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25663701

RESUMO

The levels and intracellular localization of wild-type transforming growth factor ß superfamily (TGFß-SF) receptors are tightly regulated by endocytic trafficking, shedding and degradation. In contrast, a main regulatory mechanism of mutation-bearing receptors involves their intracellular retention. Anti-Müllerian hormone receptor II (AMHRII, also known as AMHR2) is the type-II receptor for anti-Müllerian hormone (AMH), a TGFß-SF ligand that mediates Müllerian duct regression in males. Here, we studied AMHRII processing and identified novel mechanisms of its constitutive negative regulation. Immunoblot analysis revealed that a significant portion of AMHRII was missing most of its extracellular domain (ECD) and, although glycosylated, was unfolded and retained in the endoplasmic reticulum. Exogenous expression of AMHRII, but not of type-II TGF-ß receptor (TßRII, also known as TGFR2), resulted in its disulfide-bond-mediated homo-oligomerization and intracellular retention, and in a decrease in its AMH-binding capacity. At the plasma membrane, AMHRII differed from TßRII, forming high levels of non-covalent homomeric complexes, which exhibited a clustered distribution and restricted lateral mobility. This study identifies novel mechanisms of negative regulation of a type-II TGFß-SF receptor through cleavage, intracellular retention and/or promiscuous disulfide-bond mediated homo-oligomerization.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Hormônio Antimülleriano/metabolismo , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Ligação Proteica , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Peptídeos/química , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/metabolismo
6.
Development ; 139(13): 2283-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22627284

RESUMO

The function of AMH (Anti-Müllerian hormone), a phylogenetically ancient member of the TGFß family of proteins, in lower vertebrates is largely unknown. Previously, we have shown that the gene encoding the type II anti-Müllerian hormone receptor, amhrII, is responsible for excessive germ cell proliferation and male-to-female sex reversal in the medaka hotei mutant. In this study, functional analyses in cultured cells and of other amhrII mutant alleles indicate that lack of AMH signaling causes the hotei phenotype. BrdU incorporation experiments identified the existence of both quiescent and mitotically active germ cells among the self-renewing, type I population of germ cells in the developing gonad. AMH signaling acts in supporting cells to promote the proliferation of mitotically active germ cells but does not trigger quiescent germ cells to proliferate in the developing gonad. Furthermore, we show that the male-to-female sex reversal phenotype in hotei mutants is not a direct consequence of AMH signaling in supporting cells, but is instead mediated by germ cells. Our data demonstrate that interfollicular AMH signaling regulates proliferation at a specific stage of germ cell development, and that this regulation is crucial for the proper manifestation of gonadal sex directed by sex determination genes.


Assuntos
Hormônio Antimülleriano/fisiologia , Proliferação de Células , Células Germinativas/citologia , Oryzias/crescimento & desenvolvimento , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Diferenciação Sexual , Animais , Células Cultivadas , Feminino , Células Germinativas/fisiologia , Masculino , Mitose , Mutação , Oryzias/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais
7.
Hum Reprod ; 28(3): 762-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23321213

RESUMO

STUDY QUESTION: Are anti-Müllerian hormone (AMH) and AMH type II receptor (AMHR-II) mRNAs similarly regulated by gonadotrophins in lutein granulosa cells (GCs) from control, normo-ovulatory and oligo/anovulatory women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER: AMH mRNA expression was induced by LH only in lutein GC of oligo/anovulatory PCOS women; down-regulation of AMHR-II, induced by LH in control and normo-ovulatory PCOS women, was absent in oligo/anovulatory women. WHAT IS KNOWN ALREADY: It was suggested that AMH could be responsible for the blockade of follicles at the small antral stage in PCOS women. In keeping with this hypothesis, both AMH and AMHR-II are overexpressed in lutein GCs from oligo/anovulatory PCOS women. STUDY DESIGN, SIZE, DURATION: Women undergoing IVF were included in this prospective study, either in the control group (30 women) or in the PCOS group (21 normo-ovulatory and 19 oligo/anovulatory patients) between January 2010 and July 2012. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human lutein GCs were isolated from follicular fluid during IVF protocols. Twenty-four hours after seeding, lutein GCs from each woman were serum starved and cultured for 48 h ± FSH, LH or cAMP. Then AMH and AMHR-II mRNAs were quantified by quantitative RT-PCR and AMH protein concentration was measured in the culture medium by ELISA. Experimental results were analyzed, within each group of women, by the non-parametric Wilcoxon test for paired comparisons between cells cultured in control medium and FSH, LH or cAMP treated cells. Clinical comparisons between the three groups of women were performed on log values using the ANOVA test with Bonferroni correction. MAIN RESULTS AND THE ROLE OF CHANCE: FSH up-regulated both AMH expression and secretion by lutein GCs from the three groups of women (P < 0.05). LH had no effect on AMH mRNAs levels in lutein GCs from controls and normo-ovulatory PCOS women, but increased AMH expression in oligo/anovulatory PCOS women (P < 0.05). Interestingly, LH and cAMP treatments reduced AMHR-II expression by lutein GCs from controls and normo-ovulatory PCOS women (P < 0.05), but had no effect on AMHR-II mRNA levels in oligo/anovulatory PCOS women. LIMITATIONS, REASONS FOR CAUTION: The lutein GCs are not the best model to study AMH and AMHR-II regulation by gonadotrophins. Indeed, AMH and AMHR-II are down-regulated in luteinized cells. Furthermore, these cells have been exposed to non-physiological levels of gonadotrophins and hCG. However, AMH and AMHR-II mRNAs are quantifiable by real-time RT-PCR, and the cells are still responsive to FSH and LH. The age of patients is significantly different between control and oligo/anovulatory PCOS women: this may be a bias in the interpretation of results but older women in the control group had a good ovarian reserve. WIDER IMPLICATIONS OF THE FINDINGS: The overexpression of AMH and AMHR-II in oligo/anovulatory PCOS women could be due to increased LH levels and/or inhibition of its repressive action. The fact that this dysregulation is observed in oligo/anovulatory, but not in normo-ovulatory, PCOS women emphasizes the role of LH in the follicular arrest of PCOS women and suggests that this involves the AMH/AMHR-II system. STUDY FUNDING/COMPETING INTEREST(S): The Assistance-Publique Hôpitaux de Paris provided a Contrat d'Interface and the Agence de Biomédecine provided a grant to Nathalie di Clemente. Schering-Plough provided an FARO grant to Alice Pierre. The authors have nothing to disclose.


Assuntos
Anovulação/etiologia , Células da Granulosa/metabolismo , Fase Luteal/metabolismo , Hormônio Luteinizante/metabolismo , Síndrome do Ovário Policístico/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Regulação para Cima , Adulto , Hormônio Antimülleriano/biossíntese , Hormônio Antimülleriano/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Regulação para Baixo , Feminino , Hormônio Foliculoestimulante/metabolismo , Líquido Folicular , Células da Granulosa/patologia , Humanos , Síndrome do Ovário Policístico/fisiopatologia , Estudos Prospectivos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/genética , Índice de Gravidade de Doença
8.
Biomedicines ; 10(10)2022 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-36289767

RESUMO

This article reviews the main findings on anti-Müllerian hormone (AMH) and its involvement in the pathogenesis of polycystic ovary syndrome (PCOS) and its male equivalent. In women, AMH is produced by granulosa cells from the mid-fetal life to menopause and is a reliable indirect marker of ovarian reserve. AMH protects follicles from atresia, inhibits their differentiation in the ovary, and stimulates gonadotrophin-releasing hormone neurons pulsatility. AMH overexpression in women with PCOS likely contributes to the increase of the follicle cohort and of androgen levels, leading to follicular arrest and anovulation. In the male, AMH is synthesized at high levels by Sertoli cells from fetal life to puberty when serum AMH falls to levels similar to those observed in women. AMH is involved in the differentiation of the genital tract during fetal life and plays a role in Sertoli and Leydig cells differentiation and function. Serum AMH is used to assess Sertoli cell function in children with disorders of sex development and various conditions affecting the hypothalamic-pituitary-testicular axis. Although the reproductive function of male relative of women with PCOS has been poorly investigated, adolescents have elevated levels of AMH which could play a detrimental role on their fertility.

9.
Am J Physiol Endocrinol Metab ; 301(3): E539-47, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21693691

RESUMO

In Sertoli cells, anti-Müllerian hormone (AMH) expression is upregulated by FSH via cyclic AMP (cAMP), although no classical cAMP response elements exist in the AMH promoter. The response to cAMP involves NF-κB and AP2; however, targeted mutagenesis of their binding sites in the AMH promoter do not completely abolish the response. In this work we assessed whether SOX9, SF1, GATA4, and AP1 might represent alternative pathways involved in cAMP-mediated AMH upregulation, using real-time RT-PCR (qPCR), targeted mutagenesis, luciferase assays, and immunocytochemistry in the Sertoli cell line SMAT1. We also explored the signaling cascades potentially involved. In qPCR experiments, Amh, Sox9, Sf1, and Gata4 mRNA levels increased after SMAT1 cells were incubated with cAMP. Blocking PKA abolished the effect of cAMP on Sox9, Sf1, and Gata4 expression, inhibiting PI3K/PKB impaired the effect on Sf1 and Gata4, and reducing MEK1/2 and p38 MAPK activities curtailed Gata4 increase. SOX9 and SF1 translocated to the nucleus after incubation with cAMP. Mutations of the SOX9 or SF1 sites, but not of GAT4 or AP1 sites, precluded the response of a 3,063-bp AMH promoter to cAMP. In conclusion, in the Sertoli cell line SMAT1 cAMP upregulates SOX9, SF1, and GATA4 expression and induces SOX9 and SF1 nuclear translocation mainly through PKA, although other kinases may also participate. SOX9 and SF1 binding to the AMH promoter is essential to increase the activity of the AMH promoter in response to cAMP.


Assuntos
Hormônio Antimülleriano/metabolismo , AMP Cíclico/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células de Sertoli/metabolismo , Fator Esteroidogênico 1/metabolismo , Hormônio Antimülleriano/genética , Linhagem Celular , AMP Cíclico/genética , Proteínas de Ligação a DNA , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Expressão Gênica , Humanos , Masculino , Regiões Promotoras Genéticas , Fatores de Processamento de RNA , Fatores de Transcrição SOX9/genética , Transdução de Sinais/fisiologia , Fator Esteroidogênico 1/genética , Fatores de Transcrição , Regulação para Cima
10.
Hum Mol Genet ; 18(16): 3002-13, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19457927

RESUMO

The anti-Müllerian hormone type II (AMHRII) receptor is the primary receptor for anti-Müllerian hormone (AMH), a protein produced by Sertoli cells and responsible for the regression of the Müllerian duct in males. AMHRII is a membrane protein containing an N-terminal extracellular domain (ECD) that binds AMH, a transmembrane domain, and an intracellular domain with serine/threonine kinase activity. Mutations in the AMHRII gene lead to persistent Müllerian duct syndrome in human males. In this paper, we have investigated the effects of 10 AMHRII mutations, namely 4 mutations in the ECD and 6 in the intracellular domain. Molecular models of the extra- and intracellular domains are presented and provide insight into how the structure and function of eight of the mutant receptors, which are still expressed at the cell surface, are affected by their mutations. Interestingly, two soluble receptors truncated upstream of the transmembrane domain are not secreted, unless the transforming growth factor beta type II receptor signal sequence is substituted for the endogenous one. This shows that the AMHRII signal sequence is defective and suggests that AMHRII uses its transmembrane domain instead of its signal sequence to translocate to the endoplasmic reticulum, a characteristic of type III membrane proteins.


Assuntos
Hormônio Antimülleriano/metabolismo , Transtornos do Desenvolvimento Sexual/genética , Mutação , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Transtornos do Desenvolvimento Sexual/metabolismo , Humanos , Ligantes , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores de Peptídeos/química , Receptores de Fatores de Crescimento Transformadores beta/química , Alinhamento de Sequência
11.
J Pathol ; 221(4): 443-51, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20593483

RESUMO

Transgenic mice bearing a construct in which the expression of the SV40 oncogene is directed by the AMH promoter (AT mice) develop testicular tumours in adult life. We aimed to study early steps of tumour development and characterize tumours at different ages by histological, morphometric, and immunohistochemical techniques. One- to 3-month-old AT mice depicted multifocal Leydig cell hyperplasia. The testicular volume occupied by interstitial tissue was significantly higher in 3-month-old AT mice in comparison with littermate controls. Between 5 1/2 and 7 months, microscopic interstitial tumours developed that progressively evolved to form large confluent areas of high mitotic index in 7- to 14-month-old AT mice. Tumour cells had the characteristics and histoarchitecture of Leydig cells, or formed solid cord-like structures reminiscent of those seen in Sertoli cell tumours. Hyperplastic areas and tumours diffusely expressed 3beta-hydroxysteroid dehydrogenase (3beta-HSD) in Leydig cell areas. AMH expression was negative in Leydig cell conglomerates and tumours and variable in cord-like tumours. The SV40 T antigen and markers of cell proliferation (PCNA) were intensely positive in hyperplastic cells and tumours. Control mice of similar ages showed neither hyperplasia nor tumours, and SV40 T expression was always negative. In conclusion, transgenic mice develop large testicular tumours that are preceded by interstitial hyperplasia and microtumours. The histological and immunohistochemical phenotype of tumours (Leydig and Sertoli cell differentiation, positive 3beta-HSD, and variable AMH) suggests a mixed differentiation of somatic cells of the specialized gonadal stroma. The finding that an oncogene directed by a promoter specifically active in fetal Sertoli cells has given rise to testicular tumours of mixed differentiation is compatible with a common origin of Leydig and Sertoli cells from the specific stroma of the gonadal ridge, as supported by double labelling experiments in fetal mice showing co-localization of the transgene with Sertoli and Leydig cell markers.


Assuntos
Tumor de Células de Leydig/patologia , Neoplasias Testiculares/patologia , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Diferenciação Celular , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Hiperplasia/patologia , Tumor de Células de Leydig/metabolismo , Células Intersticiais do Testículo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Células de Sertoli/patologia , Neoplasias Testiculares/metabolismo
12.
Endocr Rev ; 42(6): 753-782, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33851994

RESUMO

Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.


Assuntos
Hormônios Peptídicos , Síndrome do Ovário Policístico , Hormônio Antimülleriano/metabolismo , Feminino , Células da Granulosa , Humanos , Hormônios Peptídicos/metabolismo , Síndrome do Ovário Policístico/metabolismo , Reprodução
13.
Nat Commun ; 12(1): 1064, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33594056

RESUMO

Polycystic ovary syndrome (PCOS) is characterized by an oligo-anovulation, hyperandrogenism and polycystic ovarian morphology combined with major metabolic disturbances. However, despite the high prevalence and the human and economic consequences of this syndrome, its etiology remains unknown. In this study, we show that female Goto-Kakizaki (GK) rats, a type 2 diabetes mellitus model, encapsulate naturally all the reproductive and metabolic hallmarks of lean women with PCOS at puberty and in adulthood. The analysis of their gestation and of their fetuses demonstrates that this PCOS-like phenotype is developmentally programmed. GK rats also develop features of ovarian hyperstimulation syndrome. Lastly, a comparison between GK rats and a cohort of women with PCOS reveals a similar reproductive signature. Thus, this spontaneous rodent model of PCOS represents an original tool for the identification of the mechanisms involved in its pathogenesis and for the development of novel strategies for its treatment.


Assuntos
Síndrome do Ovário Policístico/patologia , Adiposidade , Animais , Animais Recém-Nascidos , Peso Corporal , Análise Discriminante , Modelos Animais de Doenças , Dislipidemias/patologia , Sistema Endócrino/patologia , Ciclo Estral , Feminino , Teste de Tolerância a Glucose , Gonadotropinas/farmacologia , Hormônios/sangue , Humanos , Secreção de Insulina , Análise dos Mínimos Quadrados , Lipídeos/química , Masculino , Troca Materno-Fetal , Análise Multivariada , Ovário/patologia , Ovário/fisiopatologia , Fenótipo , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/fisiopatologia , Gravidez , Ratos Wistar , Reprodução , Maturidade Sexual
14.
Genes (Basel) ; 12(11)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34828406

RESUMO

Down syndrome (DS) is the most common chromosomal disorder. It is responsible for intellectual disability (ID) and several medical conditions. Although men with DS are thought to be infertile, some spontaneous paternities have been reported. The few studies of the mechanism of infertility in men with DS are now dated. Recent research in zebrafish has indicated that overexpression of DYRK1A (the protein primarily responsible for ID in DS) impairs gonadogenesis at the embryonic stage. To better ascertain DYRK1A's role in infertility in DS, we investigated the effect of DYRK1A overexpression in a transgenic mouse model. We found that overexpression of DYRK1A impairs fertility in transgenic male mice. Interestingly, the mechanism in mice differs slightly from that observed in zebrafish but, with disruption of the early stages of spermatogenesis, is similar to that seen in humans. Unexpectedly, we observed hypogonadotropic hypogonadism in the transgenic mice.


Assuntos
Hipogonadismo/genética , Infertilidade Masculina/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Espermatogênese , Animais , Hipogonadismo/patologia , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Testículo/embriologia , Testículo/patologia , Regulação para Cima , Quinases Dyrk
15.
J Clin Endocrinol Metab ; 106(3): e1271-e1289, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33247926

RESUMO

PURPOSE: A protective effect of anti-Müllerian hormone (AMH) on follicle atresia was recently demonstrated using long-term treatments, but this effect has never been supported by mechanistic studies. This work aimed to gain an insight into the mechanism of action of AMH on follicle atresia and on how this could account for the increased follicle pool observed in women with polycystic ovary syndrome (PCOS). METHODS: In vivo and in vitro experiments were performed to study the effects of AMH on follicle atresia and on the proliferation and apoptosis of granulosa cells (GCs). RNA-sequencing was carried out to identify new AMH target genes in GCs. The expression of some of these genes in GCs from control and PCOS women was compared using microfluidic real time quantitative RT-PCR. RESULTS: A short-term AMH treatment prevented follicle atresia in prepubertal mice. Consistent with this result, AMH inhibited apoptosis and promoted proliferation of different models of GCs. Moreover, integrative biology analyses of 965 AMH target genes identified in 1 of these GC models, confirmed that AMH had initiated a gene expression program favoring cell survival and proliferation. Finally, on 43 genes selected among the most up- and down-regulated AMH targets, 8 were up-regulated in GCs isolated from PCOS women, of which 5 are involved in cell survival. MAIN CONCLUSIONS: Our results provide for the first time cellular and molecular evidence that AMH protects follicles from atresia by controlling GC survival and suggest that AMH could participate in the increased follicle pool of PCOS patients.


Assuntos
Hormônio Antimülleriano/farmacologia , Apoptose , Células da Granulosa/efeitos dos fármacos , Síndrome do Ovário Policístico/patologia , Adulto , Animais , Hormônio Antimülleriano/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/patologia , Células da Granulosa/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo
16.
J Dev Orig Health Dis ; 11(2): 154-158, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31309911

RESUMO

Epidemiological studies have demonstrated an increased risk of developing non-transmittable diseases in adults subjected to adverse early developmental conditions. Metabolic and cardiovascular diseases have been the focus of most studies. Nevertheless, data from animal models also suggest early programming of fertility. In humans, it is difficult to assess the impact of the in utero environment retrospectively. Birthweight is commonly used as an indirect indicator of intrauterine development. This research is part of the ALIFERT study. We investigated a potential link between ponderal index at birth and female fertility in adulthood. Data from 51 infertile and 74 fertile women were analysed. BW was on average higher in infertile women, whereas birth length did not differ between the two groups; thus, resulting in a significantly higher ponderal index at birth in infertile women. Ponderal index at birth has been identified as a risk factor for infertility. These results suggest the importance of the intra-uterine environment, not only for long-term metabolic health but also for fertility.


Assuntos
Peso ao Nascer/fisiologia , Estatura/fisiologia , Transtornos da Nutrição Fetal/epidemiologia , Infertilidade Feminina/epidemiologia , Adolescente , Adulto , Estudos de Casos e Controles , Feminino , Fertilidade/fisiologia , Transtornos da Nutrição Fetal/diagnóstico , Transtornos da Nutrição Fetal/fisiopatologia , Humanos , Infertilidade Feminina/fisiopatologia , Gravidez , Estudos Prospectivos , Estudos Retrospectivos , Fatores de Risco , Circunferência da Cintura/fisiologia , Adulto Jovem
17.
Sci Rep ; 10(1): 15062, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934281

RESUMO

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Assuntos
Síndrome de Resistência a Andrógenos/metabolismo , Hormônio Antimülleriano/metabolismo , Receptor alfa de Estrogênio/biossíntese , Proteínas de Neoplasias/biossíntese , Síndrome de Peutz-Jeghers/metabolismo , Elementos de Resposta , Células de Sertoli/metabolismo , Síndrome de Resistência a Andrógenos/patologia , Animais , Linhagem Celular , Criança , Pré-Escolar , Estradiol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Síndrome de Peutz-Jeghers/patologia , Células de Sertoli/patologia
18.
Oncogene ; 39(9): 1875-1890, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31745296

RESUMO

Ovarian granulosa cell tumors (GCTs) are indolent tumors of the ovary affecting women at all ages and potentially displaying late recurrence. Even if there is still little information regarding the mechanisms involved in GCT development and progression, FOXL2 would be a major tumor suppressor gene in granulosa cells. We analyzed the mechanisms underlying GCT initiation and progression by using mice with targeted expression of SV40 large T-antigen in granulosa cells (AT mouse), which develop GCTs. Consistent with patients, AT mice with developing GCTs displayed increased levels in circulating anti-Müllerian hormone (AMH), estradiol and androgens, as well as decreased FOXL2 protein abundance. Very few mice developed metastases (1 out of 30). In situ analyses revealed that GCT initiation resulted from both increased granulosa cell survival and proliferation in large antral follicles. Tumorigenesis was associated with the combined inactivation of p53 and Rb pathways, as shown by the impaired expression of respective downstream targets regulating cell apoptosis and proliferation, i.e., Bax, Bak, Gadd45a, Ccna2, Ccne1, E2f1, and Orc1. Importantly, the expression of FOXL2 was still present in newly developed GCTs and its downregulation only started during GCT growth. Collectively, our experiments provide evidence that disrupted p53/Rb signaling can drive tumor initiation and growth. This model challenges the current paradigm that impaired FOXL2 signaling is a major switch of granulosa cell tumorigenesis, albeit possibly contributing to tumor growth.


Assuntos
Carcinogênese/patologia , Proteína Forkhead Box L2/metabolismo , Tumor de Células da Granulosa/patologia , Células da Granulosa/patologia , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Células Cultivadas , Regulação para Baixo , Feminino , Proteína Forkhead Box L2/genética , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , Células da Granulosa/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética
19.
Endocr Relat Cancer ; 15(1): 217-27, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18310289

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) results from the mutation of the predisposing gene, MEN1. Heterozygous Men1 mutant mice previously generated by several laboratories, including ours, mimic largely MEN1 pathology. Interestingly, our heterozygous Men1 mutant mice exhibit not only the endocrine tumours commonly seen in MEN1 patients, but also Leydig cell tumours (LCT) with high frequency, accompanied systematically by loss of the wild-type Men1 allele. As there exists a similarity of tumour phenotype between these mice and those mutated for the components of anti-Mullerian hormone (AMH)/bone morphogenic protein (BMP) pathway belonging to transforming growth factor-beta (TGF-beta) family, we investigated the expression and the activity of this pathway, known to have an important biological role in Leydig cells. Here, we report that the expression of AMH receptor type 2 is reduced in Men1 LCTs. Both immunostaining and western blot analyses also demonstrate a markedly decreased nuclear expression of Smad1, 3, 4 and 5 in the tumours. More interestingly, we show that the reconstituted menin expression in Men1-deficient Leydig cells derived from LCTs can significantly increase the transcriptional activity of a BMP pathway target promoter, XVent2. Furthermore, we found that the expression of p18, p27 and cyclin dependant kinase 4 (Cdk4), targets of TGF-beta pathways, is altered in the Leydig cell lesions. Our data provide the evidence of the deregulation of AMH/BMP and TGF-beta pathways in mouse Men1 LCTs, highlighting their involvement in tumorigenesis of Leydig cells due to Men1 inactivation.


Assuntos
Hormônio Antimülleriano/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Tumor de Células de Leydig/metabolismo , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Northern Blotting , Western Blotting , Inibidor de Quinase Dependente de Ciclina p18/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Heterozigoto , Técnicas Imunoenzimáticas , Imunoprecipitação , Tumor de Células de Leydig/patologia , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neoplasia Endócrina Múltipla Tipo 1/patologia , Plasmídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Smad/metabolismo
20.
J Clin Endocrinol Metab ; 93(11): 4456-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18697861

RESUMO

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.


Assuntos
Hormônio Antimülleriano/genética , Regulação da Expressão Gênica , Células da Granulosa/fisiologia , Síndrome do Ovário Policístico/genética , Receptores Androgênicos/genética , Receptores do FSH/genética , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Adulto , Anovulação/etiologia , Feminino , Fertilização in vitro/métodos , Humanos , Folículo Ovariano/fisiologia , Folículo Ovariano/fisiopatologia , Ovulação , Indução da Ovulação/métodos , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/diagnóstico por imagem , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ultrassonografia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa