Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 34(1): 571-587, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914586

RESUMO

Beyond the study of its transcriptional target genes, the identification of the various interactors of a transcription factor (TF) is crucial to understand its diverse cellular roles. We focused on FOXL2, a winged-helix forkhead TF important for ovarian development and maintenance. FOXL2 has been implicated in diverse cellular processes, including apoptosis, the control of cell cycle or the regulation of steroid hormone synthesis. To reliably identify partners of endogenous FOXL2, we performed a proteome-wide analysis using co-immunoprecipitation in the murine granulosa cell-derived AT29c and the pituitary-derived alpha-T3 cell lines, using three antibodies targeting different parts of the protein. Following a stringent selection of mass spectrometry data on the basis of identification reliability and protein enrichment, we identified a core set of 255 partners common to both cell lines. Their analysis showed that we could co-precipitate several complexes involved in mRNA processing, chromatin remodeling and DNA replication and repair. We further validated (direct and/or indirect) interactions with selected partners, suggesting an unexpected role for FOXL2 in those processes. Overall, this comprehensive analysis of the endogenous FOXL2 interactome sheds light on its numerous and diverse interactors and unconventional cellular roles.


Assuntos
Proteína Forkhead Box L2/metabolismo , Células da Granulosa/metabolismo , Hipófise/metabolismo , Mapas de Interação de Proteínas , Proteoma/metabolismo , Animais , Células Cultivadas , Feminino , Células da Granulosa/citologia , Camundongos , Hipófise/citologia , Proteoma/análise
2.
J Med Genet ; 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32482800

RESUMO

BACKGROUND: Primary ovarian insufficiency (POI) affects 1% of women under 40 years and is a public health problem. The genetic causes of POI are highly heterogeneous with isolated or syndromic forms. Recently, variants in genes involved in DNA repair have been shown to cause POI. Notably, syndromic POI with Fanconi anaemia (FA) traits related to biallelic BRCA2 truncated variants has been reported. Here, we report a novel phenotype of isolated POI with a BRCA2 variant in a consanguineous Turkish family. METHODS: Exome sequencing (ES) was performed in the patient. We also performed functional studies, including a homologous recombination (HR) test, cell proliferation, radiation-induced RAD51 foci formation assays and chromosome breakage studies in primary and lymphoblastoid immortalised cells. The expression of BRCA2 in human foetal ovaries was studied. RESULTS: ES identified a homozygous missense c.8524C>T/p.R2842C-BRCA2 variant. BRCA2 defects induce cancer predisposition and FA. Remarkably, neither the patient nor her family exhibited somatic pathologies. The patient's cells showed intermediate levels of chromosomal breaks, cell proliferation and radiation-induced RAD51 foci formation compared with controls and FA cells. R2842C-BRCA2 only partially complemented HR efficiency compared with wild type-BRCA2. BRCA2 is expressed in human foetal ovaries in pachytene stage oocytes, when meiotic HR occurs. CONCLUSION: We describe the functional assessment of a homozygous hypomorphic BRCA2 variant in a patient with POI without cancer or FA trait. Our findings extend the phenotype of BRCA2 biallelic alterations to fully isolated POI. This study has a major impact on the management and genetic counselling of patients with POI.

3.
Clin Genet ; 98(3): 293-298, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32613604

RESUMO

Primary ovarian insufficiency (POI) implies the cessation of menstruation for several months in women before the age of 40 years and is a major cause of infertility. The study of the contribution of genetic factors to POI has been fueled by the use of whole exome sequencing (WES). Here, to uncover novel causative pathogenic variants and risk alleles, WES has been performed in 12 patients with familial POI (eight unrelated index cases and two pairs of sisters) and six women with early menopause and family history of POI (four index cases and one pair of sisters). Likely causative variants in NR5A1 and MCM9 genes were identified as well as a variant in INHA that requires further investigation. Moreover, we have identified more than one candidate variant in 3 out of 15 familial cases. Taken together, our results highlight the genetic heterogeneity of POI and early menopause and support the hypothesis of an oligogenic inheritance of such conditions, in addition to monogenic inheritance.


Assuntos
Inibinas/genética , Proteínas de Manutenção de Minicromossomo/genética , Insuficiência Ovariana Primária/genética , Fator Esteroidogênico 1/genética , Adolescente , Adulto , Alelos , Exoma/genética , Feminino , Predisposição Genética para Doença , Humanos , Insuficiência Ovariana Primária/patologia , Fatores de Risco , Sequenciamento do Exoma , Adulto Jovem
4.
Hum Mol Genet ; 24(23): 6687-98, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26362254

RESUMO

Juvenile granulosa cell tumors (JGCTs) of the ovary are pediatric neoplasms representing 5% of all granulosa cell tumors (GCTs). Most GCTs are of adult type (AGCTs) and bear a mutation in the FOXL2 gene. The molecular basis of JGCTs is poorly understood, although mutations in the GNAS gene have been reported. We have detected in-frame duplications within the oncogene AKT1 in >60% of the JGCTs studied. Here, to evaluate the functional impact of these duplications and the existence of potential co-driver alterations, we have sequenced the transcriptome of four JGCTs and compared them with control transcriptomes. A search for gene variants detected only private alterations probably unrelated with tumorigenesis, suggesting that tandem duplications are the best candidates to underlie tumor formation in the absence of GNAS alterations. We previously showed that the duplications were specific to JGCTs. However, the screening of eight AGCTs samples without FOXL2 mutation showed the existence of an AKT1 duplication in one case, also having a stromal luteoma. The analysis of RNA-Seq data pinpointed a series of differentially expressed genes, involved in cytokine and hormone signaling and cell division-related processes. Further analyses pointed to the existence of a possible dedifferentiation process and suggested that most of the transcriptomic dysregulation might be mediated by a limited set of transcription factors perturbed by AKT1 activation. Finally, we show that commercially available AKT inhibitors can modulate the in vitro activity of various mutated forms. These results shed light on the pathogenesis of JGCTs and provide therapeutic leads for a targeted treatment.


Assuntos
Tumor de Células da Granulosa/genética , Mutação , Neoplasias Ovarianas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Adolescente , Divisão Celular/genética , Criança , Pré-Escolar , Citocinas , Análise Mutacional de DNA , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Tumor de Células da Granulosa/metabolismo , Hormônios , Humanos , Lactente , Recém-Nascido , Neoplasias Ovarianas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/genética
5.
N Engl J Med ; 370(10): 943-949, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24597867

RESUMO

Premature ovarian failure is a major cause of female infertility. The genetic causes of this disorder remain unknown in most patients. Using whole-exome sequence analysis of a large consanguineous family with inherited premature ovarian failure, we identified a homozygous 1-bp deletion inducing a frameshift mutation in STAG3 on chromosome 7. STAG3 encodes a meiosis-specific subunit of the cohesin ring, which ensures correct sister chromatid cohesion. Female mice devoid of Stag3 are sterile, and their fetal oocytes are arrested at early prophase I, leading to oocyte depletion at 1 week of age.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Mutação , Proteínas Nucleares/genética , Insuficiência Ovariana Primária/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Infertilidade Feminina/genética , Camundongos , Linhagem , Coesinas
6.
Clin Endocrinol (Oxf) ; 87(5): 539-544, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28708305

RESUMO

OBJECTIVE: Disorders of sex development (DSD) are a heterogeneous group of conditions affecting the differentiation and development of the internal and external genitalia. Here, we aimed at identifying the genetic cause of DSD in two 46,XY sisters from a consanguineous family. DESIGN: We performed a whole-exome sequencing of two 46,XY female individuals. Sanger sequencing was used to validate the most likely candidate variant, affecting the desert hedgehog (DHH) gene. Molecular dynamics simulations were performed to get insights into the impact of the variant on protein structure and on its interaction with the protein partner BOC (brother of CDO/cell adhesion molecule, downregulated by oncogenes). PATIENTS: The index patient presented with a female phenotype, primary amenorrhoea (low oestradiol and testosterone and high FSH and LH). She also had an apparent absence of intra-abdominal gonads and uterus, facial dysmorphy, psychomotor retardation and neuropathy. Her sister displayed a similar gonadal and endocrinological picture, without dysmorphy or psychomotor retardation. RESULTS: Whole-exome sequencing revealed a homozygous variant in DHH leading to the p.Trp173Cys substitution. The relevant Trp residue is conserved, and its alteration was predicted to be deleterious. Molecular dynamics simulations showed that the mutation increases the conformational flexibility of the protein and potentially alters its interaction with BOC, a positive regulator of Hedgehog signalling. We do not exclude an interference of the mutation with DHH-intein-mediated auto-processing. CONCLUSIONS: This report increases the number of described homozygous DHH variants and highlights the importance of advanced bioinformatic tools to better understand the pathogenicity of human variants.


Assuntos
Transtorno 46,XY do Desenvolvimento Sexual/genética , Proteínas Hedgehog/genética , Adulto , Substituição de Aminoácidos , Saúde da Família , Feminino , Variação Genética , Homozigoto , Humanos , Simulação de Dinâmica Molecular , Linhagem , Conformação Proteica , Irmãos , Sequenciamento do Exoma
7.
Hum Mol Genet ; 23(13): 3421-31, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24608227

RESUMO

Oligo- and azoospermia are severe forms of male infertility. However, known genetic factors account only for a small fraction of the cases. Recently, whole-exome sequencing in a large consanguineous family with inherited premature ovarian failure (POF) identified a homozygous frameshift mutation in the STAG3 gene leading to a premature stop codon. STAG3 encodes a meiosis-specific subunit of the cohesin complex, a large proteinaceous ring with DNA-entrapping ability that ensures sister chromatid cohesion and enables correct synapsis and segregation of homologous chromosomes during meiosis. The pathogenicity of the STAG3 mutations was functionally validated with a loss-of-function mouse model for STAG3 in oogenesis. However, and since none of the male members of this family was homozygous for the mutant allele, we only could hypothesized its putative involvement in male infertility. In this report, we show that male mice devoid of Stag3 display a severe meiotic phenotype that includes a meiotic arrest at zygonema-like shortening of their chromosome axial elements/lateral elements, partial loss of centromeric cohesion at early prophase and maintenance of the ability to initiate but not complete RAD51- and DMC1-mediated double-strand break repair, demonstrating that STAG3 is a crucial cohesin subunit in mammalian gametogenesis and supporting our proposal that STAG3 is a strong candidate gene for human male infertility.


Assuntos
Infertilidade Masculina/genética , Proteínas Nucleares/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Feminino , Masculino , Meiose/genética , Meiose/fisiologia , Camundongos , Proteínas Nucleares/genética , Proteínas de Ligação a Fosfato , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Complexo Sinaptonêmico/metabolismo , Coesinas
8.
BMC Cancer ; 15: 251, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25884336

RESUMO

BACKGROUND: Ovarian granulosa cell tumors (GCTs) are the most frequent sex cord-stromal tumors. Several studies have shown that a somatic mutation leading to a C134W substitution in the transcription factor FOXL2 appears in more than 95% of adult-type GCTs. Its pervasive presence suggests that FOXL2 is the main cancer driver gene. However, other mutations and genomic changes might also contribute to tumor formation and/or progression. METHODS: We have performed a combined comparative genomic hybridization and transcriptomic analyses of 10 adult-type GCTs to obtain a picture of the genomic landscape of this cancer type and to identify new candidate co-driver genes. RESULTS: Our results, along with a review of previous molecular studies, show the existence of highly recurrent chromosomal imbalances (especially, trisomy 14 and monosomy 22) and preferential co-occurrences (i.e. trisomy 14/monosomy 22 and trisomy 7/monosomy 16q). In-depth analyses showed the presence of recurrently broken, amplified/duplicated or deleted genes. Many of these genes, such as AKT1, RUNX1 and LIMA1, are known to be involved in cancer and related processes. Further genomic explorations suggest that they are functionally related. CONCLUSIONS: Our combined analysis identifies potential candidate genes, whose alterations might contribute to adult-type GCT formation/progression together with the recurrent FOXL2 somatic mutation.


Assuntos
Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Tumor de Células da Granulosa/genética , Proteínas de Neoplasias/biossíntese , Hibridização Genômica Comparativa , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Variações do Número de Cópias de DNA/genética , Feminino , Proteína Forkhead Box L2 , Regulação Neoplásica da Expressão Gênica , Estudos de Associação Genética , Genômica , Tumor de Células da Granulosa/patologia , Humanos , Proteínas de Neoplasias/genética , Mutação Puntual , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética
9.
Trends Genet ; 27(6): 224-32, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21507500

RESUMO

Forkhead box (FOX) proteins constitute an evolutionarily conserved family of transcription factors with a central role not only during development, but also in the adult organism. Thus, the misregulation and/or mutation of FOX genes often induce human genetic diseases, promote cancer or deregulate ageing. Indeed, germinal FOX gene mutations cause diseases ranging from infertility to language and/or speech disorders and immunological defects. Moreover, because of their central role in signalling pathways and in the regulation of homeostasis, somatic misregulation and/or mutation of FOX genes are associated with cancer. FOX proteins have undergone diversification in terms of their sequence, regulation and function. In addition to dedicated roles, evidence suggests that Forkhead factors have retained some functional redundancy. Thus, combinations of slightly defective alleles might induce disease phenotypes in humans, acting as quantitative trait loci. Uncovering such variants would be a big step towards understanding the functional interdependencies of different FOX members and their implications in complex pathologies.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Envelhecimento , Animais , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Humanos , Mutação , Processamento de Proteína Pós-Traducional
10.
Hum Mol Genet ; 20(13): 2642-50, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21505076

RESUMO

Primary ovarian insufficiency (POI) is characterized by the loss of ovarian function before the age of 40 in humans. Although most cases of POI are idiopathic, many are familial, underlying a genetic origin of the disease. Mutations in genes involved in the control of steroidogenesis, such as NR5A1 (SF-1, Steroidogenic Factor 1), CYP17, CYP19A1 (aromatase), StAR (Steroidogenic Acute Regulatory), and the forkhead transcription factor FOXL2 have been associated with different forms of POI. In males, the homeobox transcription factors Dlx5 and Dlx6 are involved in the control of steroidogenesis through the activation of GATA4-induced-StAR transcription. Here, we analyze the potential involvement of Dlx5 and Dlx6 in female reproduction. To this end, we make use of an existing mouse model in which Dlx5 and Dlx6 are simultaneously disrupted. We show that: (i) allelic reduction of Dlx5 and Dlx6 in the mouse results in a POI-like phenotype, characterized by reduced fertility and early follicular exhaustion; (ii) in granulosa cell lines, a reciprocal regulation exists between Dlx5 and Foxl2; (iii) in the mouse ovary, allelic reduction of Dlx5/6 results in the upregulation of Foxl2. We propose that the mutual regulation between Dlx5/6 and Foxl2 and their opposite effects on StAR expression might contribute to determine the homeostatic control of steroidogenesis within the ovary. Dysregulation of this homeostatic control would result in abnormal follicular maturation and reduced fertility. Our results bring new elements to our conceptual model of follicle maturation and maintenance and provide new potential genetic targets for cases of familial POI.


Assuntos
Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/patologia , Alelos , Animais , Linhagem Celular , Feminino , Fertilidade/genética , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Ordem dos Genes , Células da Granulosa/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Insuficiência Ovariana Primária/metabolismo
11.
Hum Mol Genet ; 20(9): 1673-86, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21289058

RESUMO

FOXL2 is a transcription factor that is essential for ovarian function and maintenance, the germline mutations of which are responsible for the Blepharophimosis Ptosis Epicanthus-inversus Syndrome (BPES), often associated with premature ovarian failure. Recent evidence has linked FOXL2 downregulation or somatic mutation (p.Cys134Trp) to cancer, although underlying molecular mechanisms remain unclear. Using a functional genomic approach, we find that FOXL2 modulates cell-cycle regulators in a way which tends to induce G1 arrest. Indeed, FOXL2 upregulation promotes cell accumulation in G1 phase and protects cells from oxidative damage, notably by promoting oxidized DNA repair and by increasing the amounts of anti-oxidant agent glutathione. In agreement with clinical observations, we find that FOXL2-mutated versions leading to BPES along with ovarian dysfunction mostly fail to transactivate cell-cycle and DNA repair targets, whereas mutations leading to isolated craniofacial defects (and normal ovarian function) activate them correctly. Interestingly, these assays revealed a mild promoter-specific hypomorphy of the tumor-associated mutation (p.Cys134Trp). Finally, the SIRT1 deacetylase suppresses FOXL2 activity on targets linked to cell-cycle and DNA repair in a dose-dependent manner. Accordingly, we find that SIRT1 inhibition by nicotinamide limits proliferation, notably by increasing endogenous FOXL2 amount/activity. The body of evidence presented here supports the idea that FOXL2 plays a key role in granulosa cell homeostasis, the failure of which is central to ovarian ageing and tumorigenesis. As granulosa cell tumors respond poorly to conventional chemotherapy, our findings on the deacetylase inhibitor nicotinamide provide an interesting option for targeted therapy.


Assuntos
Ciclo Celular , Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Células da Granulosa/citologia , Estresse Oxidativo , Sirtuína 1/metabolismo , Linhagem Celular , Reparo do DNA , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Células da Granulosa/enzimologia , Células da Granulosa/metabolismo , Humanos , Sirtuína 1/genética
12.
FASEB J ; 24(2): 346-56, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19762556

RESUMO

A transcription reaction relies on the specific recognition of cis-regulatory regions containing short DNA motifs. Such sequences are bound by transcription factors (TFs) involved in the recruitment, direct or not, of the transcriptional machinery. A eukaryotic genome can contain tens of genes encoding TFs that recognize very similar consensus DNA target sequences. In this review, we explore in a simple way how TFs coexpressed in the same cells and recognizing generic consensus sites with generic DNA-binding domains can achieve a specific modulation of target gene expression. We dissect the strategy followed by eukaryotes, which involves the formation of complex nucleoprotein structures involving many TFs and their cognate binding sites. This multiplicity of actors increases the effective length of the target DNA recognized by the TFs and might help paralogous TFs establish specific interactions. From this perspective, eukaryotic gene regulation implies the cooperation of several TFs, which is also the basis of information integration. Such cooperative TFs are likely to form a combinatorial partner code whose ultimate molecular hallmark is the assembly of enhanceosome-like structures ensuring the formation of an activation surface that is complementary to other coactivators and to the transcriptional machinery itself.


Assuntos
Sítios de Ligação/genética , DNA/metabolismo , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição/fisiologia , Sequência de Bases , DNA/genética , Eucariotos , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Homeodomínio/fisiologia , Multimerização Proteica , Fatores de Transcrição/genética
13.
Hum Mol Genet ; 17(20): 3118-27, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18635577

RESUMO

The Forkhead transcription factor FOXL2 plays a crucial role in ovarian development and maintenance. In humans, its mutations lead to craniofacial abnormalities, isolated or associated with ovarian dysfunction. Using a combinatorial approach, we identified and characterized a FoxL2 response element (FLRE) and showed that it is highly specific and that it diverges from that of other Forkhead transcription factors. This specificity should prevent aberrant regulation of FOXL2 targets by other members of the family and should prevent ectopic activation of the ovarian differentiation program in testes. We provide evidence that the FLRE is used in naturally occurring promoters. We show that polyAlanine expansions of FOXL2, which are the most frequent pathogenic mutations, induce a length-dependent loss of response on different artificial promoter reporters depending on the number and sequence of the FLREs that they contain. Thus, we provide clear mechanistic evidence explaining how the architecture of promoters influences their sensitivity to decreased transcription factor availability. Furthermore, we speculate that the generally absent ovarian phenotype of patients carrying the most frequent polyAlanine expansion should come from its ability to properly regulate high-affinity ovarian targets. The existence of critical high-affinity ovarian targets would be compatible with the role of FOXL2 in reproduction and ensure developmental and functional robustness. Taken together, our results give mechanistic insights on the molecular pathogenesis of FOXL2 polyAlanine expansions.


Assuntos
Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Mutação , Elementos de Resposta , Alelos , Sequência de Bases , Linhagem Celular , DNA/genética , DNA/metabolismo , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/química , Células da Granulosa/metabolismo , Humanos , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Peptídeos/química , Peptídeos/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Expansão das Repetições de Trinucleotídeos
15.
Elife ; 92020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32845237

RESUMO

Primary Ovarian Insufficiency (POI) is a major cause of infertility, but its etiology remains poorly understood. Using whole-exome sequencing in a family with three cases of POI, we identified the candidate missense variant S167L in HSF2BP, an essential meiotic gene. Functional analysis of the HSF2BP-S167L variant in mouse showed that it behaves as a hypomorphic allele compared to a new loss-of-function (knock-out) mouse model. Hsf2bpS167L/S167L females show reduced fertility with smaller litter sizes. To obtain mechanistic insights, we identified C19ORF57/BRME1 as a strong interactor and stabilizer of HSF2BP and showed that the BRME1/HSF2BP protein complex co-immunoprecipitates with BRCA2, RAD51, RPA and PALB2. Meiocytes bearing the HSF2BP-S167L variant showed a strongly decreased staining of both HSF2BP and BRME1 at the recombination nodules and a reduced number of the foci formed by the recombinases RAD51/DMC1, thus leading to a lower frequency of crossovers. Our results provide insights into the molecular mechanism of HSF2BP-S167L in human ovarian insufficiency and sub(in)fertility.


Assuntos
Proteínas de Transporte , Proteínas de Choque Térmico , Meiose/genética , Mutação de Sentido Incorreto/genética , Insuficiência Ovariana Primária/genética , Recombinação Genética/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Camundongos , Camundongos Knockout , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Sequenciamento do Exoma
16.
EBioMedicine ; 42: 524-531, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31000419

RESUMO

BACKGROUND: Primary Ovarian Insufficiency (POI), a major cause of infertility, affects about 1-3% of women under forty years of age. Although there is a growing list of causal genetic alterations, POI remains mostly idiopathic. METHODS: We performed exome sequencing (WES) of two sisters affected with POI, one unaffected sister and their mother from a consanguineous family. We assessed the impact of the identified MEIOB variant with a minigene assay and by sequencing illegitimate transcripts from the proband's leukocytes. We studied its functional impact on the interaction between MEIOB with its partner SPATA22 and their localization to DNA double-strand breaks (DSB). FINDINGS: We identified a homozygous variant in the last base of exon 12 of MEIOB, which encodes a factor essential for meiotic recombination. This variant was predicted to strongly affect MEIOB pre-mRNA splicing. Consistently, a minigene assay showed that the variant induced exon 12 skipping, which was confirmed in vivo in the proband's leukocytes. Aberrant splicing leads to the production of a C-terminally truncated protein that cannot interact with SPATA22, abolishing their recruitment to DSBs. INTERPRETATION: This truncating MEIOB variant is expected to provoke meiotic defects and a depleted follicular stock, as in Meiob-/- mice. This is the first molecular defect reported in a meiosis-specific single-stranded DNA-binding protein (SSB) responsible for POI. We hypothesise that alterations in other SSB proteins could explain cases of syndromic or isolated ovarian insufficiency. FUND: Université Paris Diderot, Fondation pour la Recherche Médicale, Fondation ARC contre le cancer, Commissariat à l'Energie Atomique and Institut Universitaire de France.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Mutação , Insuficiência Ovariana Primária/etiologia , Insuficiência Ovariana Primária/metabolismo , Adolescente , Adulto , Animais , Biomarcadores , Linhagem Celular , Consanguinidade , Feminino , Expressão Gênica , Humanos , Informática/métodos , Camundongos , Linhagem , Insuficiência Ovariana Primária/diagnóstico , Ligação Proteica , Sequenciamento do Exoma , Adulto Jovem
17.
Proteomics ; 8(15): 3118-23, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18604817

RESUMO

The transcription factor Forkhead box L subfamily member 2 (FOXL2) is involved in craniofacial development and ovarian function. Using 2-DE and immunoblotting, we show that it is highly modified post-translationally. The most outstanding feature of its migration profile is the presence of two distinct modification "trains" and the absence of intermediates. A theoretical analysis of the modification profile of FOXL2 suggests that it undergoes parallel processive/concerted modifications. The absence of intermediates is compatible with the recruitment of poorly modified FOXL2 into a post-translational "modification factory."


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais/fisiologia , Western Blotting , Linhagem Celular Tumoral , Eletroforese em Gel Bidimensional , Proteína Forkhead Box L2 , Humanos
18.
Oncotarget ; 9(5): 6144-6155, 2018 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-29464061

RESUMO

Lactotroph adenoma, also called prolactinoma, is the most common pituitary tumor but little is known about its pathogenesis. Mouse models of prolactinoma can be useful to better understand molecular mechanisms involved in abnormal lactotroph cell proliferation and secretion. We have previously developed a prolactin receptor deficient (Prlr-/- ) mouse, which develops prolactinoma. The present study aims to explore the natural history of prolactinoma formation in Prlr-/- mice, using hormonal, radiological, histological and molecular analyses to uncover mechanisms involved in lactotroph adenoma development. Prlr-/- females develop large secreting prolactinomas from 12 months of age, with a penetrance of 100%, mimicking human aggressive densely granulated macroprolactinoma, which is a highly secreting subtype. Mean blood PRL measurements reach 14 902 ng/mL at 24 months in Prlr-/- females while PRL levels were below 15 ng/mL in control mice (p < 0.01). By comparing pituitary microarray data of Prlr-/- mice and an estrogen-induced prolactinoma model in ACI rats, we pinpointed 218 concordantly differentially expressed (DE) genes involved in cell cycle, mitosis, cell adhesion molecules, dopaminergic synapse and estrogen signaling. Pathway/gene-set enrichment analyses suggest that the transcriptomic dysregulation in both models of prolactinoma might be mediated by a limited set of transcription factors (i.e., STAT5, STAT3, AhR, ESR1, BRD4, CEBPD, YAP, FOXO1) and kinases (i.e., JAK2, AKT1, BRAF, BMPR1A, CDK8, HUNK, ALK, FGFR1, ILK). Our experimental results and their bioinformatic analysis provide insights into early genomic changes in murine models of the most frequent human pituitary tumor.

19.
Eur J Endocrinol ; 176(5): K9-K14, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28348023

RESUMO

CONTEXT: PCOS is a heterogeneous condition characterized by hyperandrogenism and chronic anovulation and affects about 10% of women. Its etiology is poorly known, but a dysregulation of gonadotropin secretion is one of its hallmarks. OBJECTIVE: As the etiology of PCOS is unclear, we have performed a genome-wide analysis of a consanguineous family with three sisters diagnosed with PCOS. METHODS: Whole-exome sequencing and Sanger sequencing confirmation. RESULTS: Whole-exome sequencing allowed the detection of the missense variant rs104893836 located in the first coding exon of the GNRHR gene and leading to the p.Gln106Arg (p.Q106R) substitution. Sanger sequencing of all available individuals of the family confirmed that the variant was homozygous in the three affected sisters and heterozygous in both parents. CONCLUSIONS: This is the first description of a GNRHR gene mutation in patients diagnosed with PCOS. Although we do not exclude a possible interaction of the identified variant with the genetic background and/or the environment, our result suggests that genetic alterations in the hypothalamo-pituitary axis may play role in the pathogenesis of PCOS.


Assuntos
Consanguinidade , Homozigoto , Mutação , Síndrome do Ovário Policístico/genética , Receptores LHRH/genética , Feminino , Hormônio Foliculoestimulante/sangue , Humanos , Israel , Hormônio Luteinizante/sangue , Linhagem , Arábia Saudita/etnologia , Análise de Sequência de DNA
20.
Elife ; 62017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29231814

RESUMO

Primary Ovarian Insufficiency (POI) affects ~1% of women under forty. Exome sequencing of two Finnish sisters with non-syndromic POI revealed a homozygous mutation in FANCM, leading to a truncated protein (p.Gln1701*). FANCM is a DNA-damage response gene whose heterozygous mutations predispose to breast cancer. Compared to the mother's cells, the patients' lymphocytes displayed higher levels of basal and mitomycin C (MMC)-induced chromosomal abnormalities. Their lymphoblasts were hypersensitive to MMC and MMC-induced monoubiquitination of FANCD2 was impaired. Genetic complementation of patient's cells with wild-type FANCM improved their resistance to MMC re-establishing FANCD2 monoubiquitination. FANCM was more strongly expressed in human fetal germ cells than in somatic cells. FANCM protein was preferentially expressed along the chromosomes in pachytene cells, which undergo meiotic recombination. This mutation may provoke meiotic defects leading to a depleted follicular stock, as in Fancm-/- mice. Our findings document the first Mendelian phenotype due to a biallelic FANCM mutation.


Assuntos
DNA Helicases/genética , Homozigoto , Mutação , Ovário/fisiopatologia , Insuficiência Ovariana Primária/genética , Adulto , Aberrações Cromossômicas , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Feminino , Predisposição Genética para Doença , Genótipo , Recombinação Homóloga , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Insuficiência Ovariana Primária/patologia , Ubiquitinação , Sequenciamento do Exoma , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA