Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
J Ovarian Res ; 17(1): 67, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528613

RESUMO

BACKGROUND: Premature ovarian insufficiency (POI) is a severe disorder leading to female infertility. Genetic mutations are important factors causing POI. TP63-truncating mutation has been reported to cause POI by increasing germ cell apoptosis, however what factors mediate this apoptosis remains unclear. METHODS: Ninety-three patients with POI were recruited from Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Whole-exome sequencing (WES) was performed for each patient. Sanger sequencing was used to confirm potential causative genetic variants. A minigene assay was performed to determine splicing effects of TP63 variants. A TP63-truncating plasmid was constructed. Real-time quantitative PCR, western blot analyses, dual luciferase reporter assays, immunofluorescence staining, and cell apoptosis assays were used to study the underlying mechanism of a TP63-truncating mutation causing POI. RESULTS: By WES of 93 sporadic patients with POI, we found a 14-bp deletion covering the splice site in the TP63 gene. A minigene assay demonstrated that the 14-bp deletion variant led to exon 13 skipping during TP63 mRNA splicing, resulting in the generation of a truncated TP63 protein (TP63-mut). Overexpression of TP63-mut accelerated cell apoptosis. Mechanistically, the TP63-mut protein could bind to the promoter region of CLCA2 and activate the transcription of CLCA2 several times compared to that of the TP63 wild-type protein. Silencing CLCA2 using a specific small interfering RNA (siRNA) or inhibiting the Ataxia Telangiectasia Mutated (ATM) pathway using the KU55933 inhibitor attenuated cell apoptosis caused by TP63-mut protein expression. CONCLUSION: Our findings revealed a crucial role for CLCA2 in mediating apoptosis in POI pathogenesis, and suggested that CLCA2 is a potential therapeutic target for POI.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Fatores de Transcrição , Proteínas Supressoras de Tumor , Feminino , Humanos , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Éxons , Menopausa Precoce/genética , Mutação , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas Supressoras de Tumor/genética
2.
J Ovarian Res ; 17(1): 32, 2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38310280

RESUMO

BACKGROUND: The etiology of premature ovarian insufficiency, that is, the loss of ovarian activity before 40 years of age, is complex. Studies suggest that genetic factors are involved in 20-25% of cases. The aim of this study was to explore the oligogenic basis of premature ovarian insufficiency. RESULTS: Whole-exome sequencing of 93 patients with POI and whole-genome sequencing of 465 controls were performed. In the gene-burden analysis, multiple genetic variants, including those associated with DNA damage repair and meiosis, were more common in participants with premature ovarian insufficiency than in controls. The ORVAL-platform analysis confirmed the pathogenicity of the RAD52 and MSH6 combination. CONCLUSIONS: The results of this study indicate that oligogenic inheritance is an important cause of premature ovarian insufficiency and provide insights into the biological mechanisms underlying premature ovarian insufficiency.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Feminino , Humanos , Insuficiência Ovariana Primária/genética , Menopausa Precoce/genética
3.
PeerJ ; 11: e16659, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107572

RESUMO

Objective: Premature ovarian failure (POF) is a disease with high clinical heterogeneity. Subsequently, its diagnosis is challenging. CXCL10 which is a small signaling protein involved in immune response and inflammation may have diagnostic potential in detection of premature ovarian insufficiency. Therefore, this study aimed to investigate CXCL10 based diagnostic biomarkers for POF. Methods: Transcriptome data for POF was obtained from the Gene Expression Omnibus (GEO) database (GSE39501). Principal component analysis (PCA) assessed CXCL10 expression in patients with POF. The receiver operating characteristic (ROC) curve, analyzed using PlotROC, demonstrated the diagnostic potential of CXCL10 and CXCL10-based models for POF. Differentially expressed genes (DEGs) in the control group of POF were identified using DEbylimma. PlotVenn was used to determine the overlap between the POF-control group and the high-/low-expression CXCL10 groups. QuadrantPlot was employed to detect CXCL10-dysregulated genes in POF. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were conducted on DEGs using RunMulti Group cluster Profiler. A POF model was induced with cisplatin (DDP) using KGN cells. RT-qPCR and Western blot were used to measure the expression of CXCL10, apoptosis-related proteins, and peroxisome proliferator-activated receptor (PPAR) signaling pathway-related proteins in this model, following siRNA-mediated silencing of CXCL10. Flow cytometry was employed to assess the apoptosis of KGN cells after CXCL10 downregulation. Results: The expression of CXCL10 is dysregulated in POF, and it shows promising diagnostic potential for POF, as evidenced by an area under the curve value of 1. In POF, we found 3,362 up-regulated and 3,969 down-regulated DEGs compared to healthy controls, while the high- and low-expression groups of POF (comprising samples above and below the median CXCL10 expression) exhibited 1,304 up-regulated and 1,315 down-regulated DEGs. Among these, 786 DEGs consistently displayed dysregulation in POF due to CXCL10 influence. Enrichment analysis indicated that the PPAR signaling pathway was activated by CXCL10 in POF. The CXCL10-based model (including CXCL10, Itga2, and Raf1) holds potential as a diagnostic biomarker for POF. Additionally, in the DDP-induced KGN cell model, interfering with CXCL10 expression promoted the secretion of estradiol, and reduced apoptosis. Furthermore, CXCL10 silencing led to decreased expression levels of PPARß and long-chain acyl-CoA synthetase 1 compared to the Si-NC group. These results suggest that CXCL10 influences the progression of POF through the PPAR signaling pathway. Conclusion: The CXCL10-based model, demonstrating perfect diagnostic accuracy for POF and comprising CXCL10, Itga2, and Raf1, holds potential as a valuable diagnostic biomarker. Thus, the expression levels of these genes may collectively provide valuable diagnostic information for POF.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Feminino , Humanos , Insuficiência Ovariana Primária/genética , Perfilação da Expressão Gênica/métodos , Receptores Ativados por Proliferador de Peroxissomo/genética , Menopausa Precoce/genética , Biomarcadores , Família Multigênica , Quimiocina CXCL10/genética
4.
J Ovarian Res ; 16(1): 203, 2023 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-37848988

RESUMO

BACKGROUND: Premature ovarian insufficiency (POI) is an important cause of infertility characterized by the functional decline of the ovary. Granulosa cells (GCs) around oocytes are critical for folliculogenesis, and GC dysfunction is one of the important etiologies of POI. The aim of this study was to explore the potential biomarkers of POI by identifying hub genes and analyze the correlation of biomarkers with immune infiltration in POI using RNA profiling and bioinformatics analysis. METHODS: RNA sequencing was performed on GCs from biochemical POI (bPOI) patients and controls. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were used to explore the candidate genes. qRT‒PCR was performed to verify the expression of hub genes. Western blot, Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU) assays, TUNEL (TdT-mediated dUTP Nick-End Labeling) and flow cytometry analysis were used to validate the possible role of ubiquitin-conjugating enzyme 2C (UBE2C) in POI. CIBERSORT was adopted to explore immune cell infiltration and the correlation between UBE2C and immune cells in bPOI. RESULTS: Through analysis of differentially expressed genes (DEGs) and WGCNA, we obtained 143 candidate genes. After construction of the protein‒protein interaction (PPI) network and analysis with Cytoscape, 10 hub genes, including UBE2C, PBK, BUB1, CDC20, NUSAP1, CENPA, CCNB2, TOP2A, AURKB, and FOXM1, were identified and verified by qRT‒PCR. Subsequently, UBE2C was chosen as a possible biomarker of POI because knockdown of UBE2C could inhibit the proliferation and promote the apoptosis of GCs. Immune infiltration analysis indicated that monocytes and M1 macrophages may be associated with the pathogenesis of POI. In addition, UBE2C was negatively correlated with monocytes and M1 macrophages in POI. CONCLUSIONS: This study identified a hub gene in GCs that might be important in the pathogenesis of POI and revealed the key role of UBE2C in driving POI. Immune infiltration may be highly related with the onset and etiology of POI.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Feminino , Humanos , Transcriptoma , Enzimas de Conjugação de Ubiquitina/genética , Insuficiência Ovariana Primária/genética , Menopausa Precoce/genética , Células da Granulosa , Biomarcadores
5.
Gynecol Endocrinol ; 39(1): 2265507, 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37839437

RESUMO

OBJECTIVE: This study aimed to identify novel pathogenic genes and variants in a Chinese family with premature ovarian insufficiency (POI). METHODS: A Chinese POI family was enrolled in this study. Whole exome sequencing was performed on the proband and her mother to identify the potential causative genes and variants and Sanger sequencing was used to confirm the finally identified potential pathogenic variant in the family. RESULTS: An assessment of the family pedigree suggested that POI was inherited in an autosomal dominant manner in this family. A novel missense variant of the laminin subunit gamma-1 gene (LAMC1; NM_002293.4: c.3281A > T, p.D1094V) was finally identified in the proband and her affected mother. This variant was not found in any public databases. In silico analysis indicated the amino acid encoded at the variant site was highly conserved among mammals and associated with decreased protein stability and disrupted protein function. Its presence in the POI family was confirmed by Sanger sequencing. CONCLUSIONS: This study firstly reported a novel missense variant of LAMC1 in a Chinese POI family, which was inherited in an autosomal dominant manner. This variant may result in the development of POI. Our results provide supporting evidence for a causative role for LAMC1 variants in POI.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Humanos , Feminino , Animais , Sequenciamento do Exoma , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/patologia , Menopausa Precoce/genética , Mutação de Sentido Incorreto , Proteínas de Ligação a DNA , Linhagem , Mamíferos/genética
7.
Nat Med ; 29(7): 1692-1699, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37349538

RESUMO

Premature ovarian insufficiency (POI) affects 1% of women and is a leading cause of infertility. It is often considered to be a monogenic disorder, with pathogenic variants in ~100 genes described in the literature. We sought to systematically evaluate the penetrance of variants in these genes using exome sequence data in 104,733 women from the UK Biobank, 2,231 (1.14%) of whom reported at natural menopause under the age of 40 years. We found limited evidence to support any previously reported autosomal dominant effect. For nearly all heterozygous effects on previously reported POI genes, we ruled out even modest penetrance, with 99.9% (13,699 out of 13,708) of all protein-truncating variants found in reproductively healthy women. We found evidence of haploinsufficiency effects in several genes, including TWNK (1.54 years earlier menopause, P = 1.59 × 10-6) and SOHLH2 (3.48 years earlier menopause, P = 1.03 × 10-4). Collectively, our results suggest that, for the vast majority of women, POI is not caused by autosomal dominant variants either in genes previously reported or currently evaluated in clinical diagnostic panels. Our findings, plus previous studies, suggest that most POI cases are likely oligogenic or polygenic in nature, which has important implications for future clinical genetic studies, and genetic counseling for families affected by POI.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Feminino , Humanos , Adulto , Penetrância , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/complicações , Insuficiência Ovariana Primária/patologia , Menopausa Precoce/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética
8.
Hum Reprod ; 38(6): 1224-1230, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37018629

RESUMO

STUDY QUESTION: Are genetic disorders and congenital malformations associated with premature ovarian insufficiency (POI)? SUMMARY ANSWER: A wide range of genetic disorder and congenital malformation diagnoses are associated with POI, especially early onset POI. WHAT IS KNOWN ALREADY: POI is known to be associated with some genetic disorders, such as Turner syndrome and Fragile X premutation. Multiple genetic syndromes, such as ataxia teleangiectasia and galactosemia, have also been associated with an increased risk of POI, and many of these genetic syndromes manifest with various congenital malformations. In previous studies, a genetic aetiology has been found for 7-15% of POI cases. STUDY DESIGN, SIZE, DURATION: This population-based study included 5011 women diagnosed with POI in 1988-2017. The data were collected from various national registries and covers women with POI nationwide. PARTICIPANTS/MATERIALS, SETTING, METHODS: We identified 5011 women diagnosed with POI from 1988 to 2017 from the drug reimbursement registry of the Social Insurance Institution of Finland. Women with surgical POI (bilateral oophorectomy for benign indications) were not included. We selected four population controls per woman with POI matched by month and year of birth and municipality of residence. Diagnostic codes for genetic disorders and congenital malformations (GD/CM) for the cases and controls were searched from the Hospital Discharge Register. Binary logistic regression was used to compare the odds for GD/CM among cases and controls. To minimize bias, for the statistical analyses, we excluded diagnoses which were reported <2 years prior to the index date. MAIN RESULTS AND THE ROLE OF CHANCE: Of the women with POI, 15.9% (n = 797) had at least one diagnostic code for GD or CM. The odds ratio (OR) for Turner syndrome was 275 (95% CI 68.1-1110), and for other sex chromosome abnormalities, it was 12.7 (95% CI 4.1-39.1). For autosomal single gene disorders, the OR was 16.5 (95% CI 6.2-43.7). Women with POI had a higher odds of having a GD/CM diagnosis in all categories. The OR for GD/CM diagnoses was highest among the youngest POI patients (10-14 years old, OR 24.1, 95% CI 15.1-38.2). The odds of having POI were higher the more GD or CM diagnoses a woman had. LIMITATIONS, REASONS FOR CAUTION: Some women with POI might not have sought help for their symptoms and therefore remain undiagnosed. Due to the register-based nature of our study, we did not have access to more specific genetic diagnoses than international classification of diseases offers. WIDER IMPLICATIONS OF THE FINDINGS: GD/CM diagnoses were strongly associated with POI, especially when POI was diagnosed at a young age. The risk of POI was highest in women with multiple GD/CM diagnoses. Early onset POI can be a sign of underlying genetic disorder or congenital anomaly, and this should serve as a reminder for clinicians to consider further examinations. To avoid unnecessary delay in the diagnosis of POI and starting relevant hormone replacement therapy treatment, clinicians should be aware of these associations. STUDY FUNDING/COMPETING INTEREST(S): Oulu University Hospital financially supported this work. H.S. has received personal grants from the Finnish Menopause Society, Oulu Medical Research Foundation, and Finnish Research Foundation of Gynaecology and Obstetrics. S.S. has received grants from the Finnish Menopause Society, the Finnish Medical Foundation, and the Juho Vainio Foundation. None of the authors have any competing interests to declare. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Síndrome de Turner , Gravidez , Feminino , Humanos , Criança , Adolescente , Síndrome de Turner/complicações , Síndrome de Turner/epidemiologia , Síndrome de Turner/genética , Insuficiência Ovariana Primária/epidemiologia , Insuficiência Ovariana Primária/genética , Menopausa Precoce/genética , Menopausa , Terapia de Reposição Hormonal
9.
Clin Genet ; 103(3): 352-357, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36373164

RESUMO

Ovarian dysfunction, including premature ovarian insufficiency and decreased ovarian reserve, affects the ovarian reserve and is one of the leading causes of female infertility. More and more cases of ovarian dysfunction are associated with genetic factors. Here, we identified eight potential variants in five genes (MSH4, HFM1, SYCE1, FSHR, and C14orf39) from six independent families by exome sequencing. The splice-site variants in SYCE1 and MSH4 affected canonical splicing isoforms, leading to missing protein domains or premature termination. Our findings expand the mutational spectrum of ovarian dysfunction and provide potential biomarkers for future genetic counseling and for more personalized treatments. Exome sequencing was shown to be a useful tool to better dissect the genetic basis for ovarian dysfunction and yielded a genetic diagnosis in about 5.0% (6/124) of cases in a cohort of 124 patients with ovarian dysfunction.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Humanos , Feminino , Insuficiência Ovariana Primária/diagnóstico , Insuficiência Ovariana Primária/genética , Menopausa Precoce/genética , Mutação , Testes Genéticos
10.
FASEB J ; 36(11): e22612, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36250920

RESUMO

Fragile X-associated premature ovarian insufficiency (FXPOI) is among a family of disorders caused by expansion of a CGG trinucleotide repeat sequence located in the 5' untranslated region (UTR) of the fragile X messenger ribonucleoprotein 1 (FMR1) gene on the X chromosome. Women with FXPOI have a depleted ovarian follicle population, resulting in amenorrhea, hypoestrogenism, and loss of fertility before the age of 40. FXPOI is caused by expansions of the CGG sequence to lengths between 55 and 200 repeats, known as a FMRI premutation, however the mechanism by which the premutation drives disease pathogenesis remains unclear. Two main hypotheses exist, which describe an mRNA toxic gain-of-function mechanism or a protein-based mechanism, where repeat-associated non-AUG (RAN) translation results in the production of an abnormal protein, called FMRpolyG. Here, we have developed an in vitro granulosa cell model of the FMR1 premutation by ectopically expressing CGG-repeat RNA and FMRpolyG protein. We show that expanded CGG-repeat RNA accumulated in intranuclear RNA structures, and these aggregates were able to cause significant granulosa cell death independent of FMRpolyG expression. Using an innovative RNA pulldown, mass spectrometry-based approach we have identified proteins that are specifically sequestered by CGG RNA aggregates in granulosa cells in vitro, and thus may be deregulated as consequence of this interaction. Furthermore, we have demonstrated reduced expression of three proteins identified via our RNA pulldown (FUS, PA2G4 and TRA2ß) in ovarian follicles in a FMR1 premutation mouse model. Collectively, these data provide evidence for the contribution of an mRNA gain-of-function mechanism to FXPOI disease biology.


Assuntos
Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Menopausa Precoce , Insuficiência Ovariana Primária , Animais , Feminino , Humanos , Camundongos , Regiões 5' não Traduzidas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/complicações , Síndrome do Cromossomo X Frágil/genética , Mutação com Ganho de Função , Menopausa Precoce/genética , Insuficiência Ovariana Primária/etiologia , Insuficiência Ovariana Primária/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Expansão das Repetições de Trinucleotídeos/genética
11.
Reprod Biomed Online ; 45(3): 421-424, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35523711

RESUMO

Recent genome-wide association studies have shown that the majority of genes involved in menopause are also instrumental in double-strand break repair and mismatch and base excision repair of DNA. Cumulative DNA damage causes cellular senescence resulting in exhaustion of somatic cell renewal capacity and cellular dysfunction, and eventually to accelerated cell death, generally called ageing. A similar erosion of the genome occurs within the germ cell line and thus in the ovaries. Subsequently, the systemic 'survival' response intentionally suppresses the sex-steroid hormone output, which in turn may contribute to the onset of menopause. The latter occurs in particular when age-dependent DNA damage accumulates. Both effects are expected to synergize to promote ovarian silencing resulting in menopause. Consequently, ageing of the soma seems to be a primary driver for the loss of ovarian function in women. Therefore menopause is the result rather than the cause of ageing.


Assuntos
Estudo de Associação Genômica Ampla , Menopausa Precoce , Envelhecimento/genética , Senescência Celular , Reparo do DNA , Feminino , Humanos , Menopausa/genética , Menopausa Precoce/genética
12.
Fertil Steril ; 117(4): 843-853, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35115167

RESUMO

OBJECTIVE: To study the diagnostic yield, including variants in genes yet to be incriminated, of whole exome sequencing (WES) in familial cases of premature ovarian insufficiency (POI). DESIGN: Cross-sectional study. SETTING: Endocrinology and reproductive medicine teaching hospital departments. PATIENTS: Familial POI cases were recruited as part of a nationwide multicentric cohort. A total of 36 index cases in 36 different families were studied. Fifty-two relatives were available, including 25 with POI and 27 affected who were nonaffected. Karyotype analysis, FMR1 screening, single nucleotide polymorphism array analysis, and WES were performed in all subjects. INTERVENTIONS: None. MAIN OUTCOME MEASURES: The primary outcome was a molecular etiology, as diagnosed by karyotype, FMR1 screening, single nucleotide polymorphism array, and WES. RESULTS: A likely molecular etiology (pathogenic or likely pathogenic variant) was identified in 18 of 36 index cases (50% diagnostic yield). In 12 families, we found a pathogenic or likely pathogenic variant in a gene previously incriminated in POI, and in 6 families, we found a pathogenic or likely pathogenic variant in new candidate genes. Most of the variants identified were located in genes involved in cell division and meiosis (n = 11) or DNA repair (n = 4). CONCLUSIONS: The genetic etiologic diagnosis in POI allows for genetic familial counseling, anticipated pregnancy planning, and ovarian tissue preservation or oocyte preservation. Identifying new genes may lead to future development of therapeutics in reproduction based on disrupted molecular pathways. CLINICAL TRIAL REGISTRATION NUMBER: NCT 01177891.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Estudos de Coortes , Estudos Transversais , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Humanos , Menopausa Precoce/genética , Insuficiência Ovariana Primária/diagnóstico , Insuficiência Ovariana Primária/genética , Sequenciamento do Exoma
13.
Hum Mol Genet ; 31(13): 2223-2235, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134173

RESUMO

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


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Animais , Elementos Facilitadores Genéticos/genética , Feminino , Variação Genética , Humanos , Menopausa Precoce/genética , Camundongos , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , RNA/genética , Fatores de Tempo
14.
Minerva Obstet Gynecol ; 74(2): 165-170, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34137566

RESUMO

INTRODUCTION: Many studies have shown that BRCA mutation is not only related to cancer but also to ovarian aging. Studies in both human and mice oocytes have shown that Double-strand breaks (DSBs) accumulate with age. EVIDENCE ACQUISITION: A review of the literature was completed through the PubMed database aiming to find articles regarding BRCA 1,2 mutation and if they are related to early menopause in order to use them as predictive biomarkers in the near future. The research used keywords in numerous combinations, such as "BRCA 1,2 mutation," "menopause," "ovarian reserves," "AMH," "genome-wide association studies," and "biomarkers." The literature was limited in this specific topic. The initial research found 16 screened articles, 7 of which were not included because there were not relevant, as far as publications in non-English language. EVIDENCE SYNTHESIS: Genome-wide association studies (GWAS) have found 44 genetic loci that are related to variations when a female is about to have menopause. BRCA1 is involved in these 44 loci that are associated with the age of menopause. This review has gathered all results of literature search about the association between BRCA genes and early menopause. Most of the articles found that women with BRCA mutation have earlier menopause compared to non-carriers. CONCLUSIONS: In conclusion, in the near future BRCA1,2 genes could be used as predictive biomarkers of menopause.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Menopausa Precoce , Reserva Ovariana , Animais , Feminino , Genes Supressores de Tumor , Estudo de Associação Genômica Ampla , Humanos , Menopausa/genética , Menopausa Precoce/genética , Camundongos , Mutação/genética , Reserva Ovariana/genética
15.
Nature ; 596(7872): 393-397, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34349265

RESUMO

Reproductive longevity is essential for fertility and influences healthy ageing in women1,2, but insights into its underlying biological mechanisms and treatments to preserve it are limited. Here we identify 290 genetic determinants of ovarian ageing, assessed using normal variation in age at natural menopause (ANM) in about 200,000 women of European ancestry. These common alleles were associated with clinical extremes of ANM; women in the top 1% of genetic susceptibility have an equivalent risk of premature ovarian insufficiency to those carrying monogenic FMR1 premutations3. The identified loci implicate a broad range of DNA damage response (DDR) processes and include loss-of-function variants in key DDR-associated genes. Integration with experimental models demonstrates that these DDR processes act across the life-course to shape the ovarian reserve and its rate of depletion. Furthermore, we demonstrate that experimental manipulation of DDR pathways highlighted by human genetics increases fertility and extends reproductive life in mice. Causal inference analyses using the identified genetic variants indicate that extending reproductive life in women improves bone health and reduces risk of type 2 diabetes, but increases the risk of hormone-sensitive cancers. These findings provide insight into the mechanisms that govern ovarian ageing, when they act, and how they might be targeted by therapeutic approaches to extend fertility and prevent disease.


Assuntos
Envelhecimento/genética , Ovário/metabolismo , Adulto , Alelos , Animais , Osso e Ossos/metabolismo , Quinase 1 do Ponto de Checagem/genética , Quinase do Ponto de Checagem 2/genética , Diabetes Mellitus Tipo 2 , Dieta , Europa (Continente)/etnologia , Ásia Oriental/etnologia , Feminino , Fertilidade/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Envelhecimento Saudável/genética , Humanos , Longevidade/genética , Menopausa/genética , Menopausa Precoce/genética , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Insuficiência Ovariana Primária/genética , Útero
16.
FASEB J ; 35(8): e21753, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34233068

RESUMO

Ovarian infertility and subfertility presenting with premature ovarian insufficiency (POI) and diminished ovarian reserve are major issues facing the developed world due to the trend of delaying childbirth. Ovarian senescence and POI represent a continuum of physiological/pathophysiological changes in ovarian follicle functions. Based on advances in whole exome sequencing, evaluation of gene copy variants, together with family-based and genome-wide association studies, we discussed genes responsible for POI and ovarian senescence. We used a gene-centric approach to sort out literature deposited in the Ovarian Kaleidoscope database (http://okdb.appliedbioinfo.net) by sub-categorizing candidate genes as ligand-receptor signaling, meiosis and DNA repair, transcriptional factors, RNA metabolism, enzymes, and others. We discussed individual gene mutations found in POI patients and verification of gene functions in gene-deleted model organisms. Decreased expression of some of the POI genes could be responsible for ovarian senescence, especially those essential for DNA repair, meiosis and mitochondrial functions. We propose to set up a candidate gene panel for targeted sequencing in POI patients together with studies on mitochondria-associated genes in middle-aged subfertile patients.


Assuntos
Ovário/metabolismo , Insuficiência Ovariana Primária/genética , Animais , Reparo do DNA/genética , Bases de Dados Genéticas , Feminino , Estudo de Associação Genômica Ampla , Humanos , Meiose/genética , Menopausa Precoce/genética , Menopausa Precoce/metabolismo , Modelos Genéticos , Reserva Ovariana/genética , Insuficiência Ovariana Primária/metabolismo , Fatores de Transcrição/genética , Sequenciamento do Exoma
17.
J Clin Endocrinol Metab ; 106(7): e2656-e2663, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33686417

RESUMO

CONTEXT: Primary ovarian insufficiency (POI) is defined by menopause before 40 years of age. POI prevalence is higher among women with autoimmune Addison's disease (AAD) than in the general population, but their clinical characteristics are insufficiently studied. OBJECTIVE: To assess the prevalence of POI in a large cohort of women with AAD and describe clinical, immunological, and genetic characteristics. METHODS: An observational population-based cohort study of the Norwegian National Addison Registry. The Norwegian Prescription Database was used to assess prescription of menopausal hormone replacement therapy (HRT). A total of 461 women with AAD were studied. The primary outcome measure was prevalence of POI. Secondary outcomes were clinical characteristics, autoantibodies, and genome-wide single nucleotide polymorphism variation. RESULTS: The prevalence of POI was 10.2% (47/461) and one-third developed POI before 30 years of age. POI preceded or coincided with AAD diagnosis in more than half of the women. The prevalence of concomitant autoimmune diseases was 72%, and AAD women with POI had more autoantibodies than AAD women without (≥2 autoantibodies in 78% vs 25%). Autoantibodies against side-chain cleavage enzyme (SCC) had the highest accuracy with a negative predictive value for POI of 96%. HRT use was high compared to the age adjusted normal population (11.3 % vs 0.7%). CONCLUSION: One in 10 women with AAD have POI. Autoantibodies against SCC are the most specific marker for autoimmune POI. We recommend testing women with AAD <40 years with menstrual disturbances or fertility concerns for autoantibodies against SCC.


Assuntos
Doença de Addison/genética , Doença de Addison/imunologia , Menopausa Precoce/genética , Menopausa Precoce/imunologia , Insuficiência Ovariana Primária/epidemiologia , Doença de Addison/complicações , Adulto , Autoanticorpos/sangue , Autoanticorpos/imunologia , Enzima de Clivagem da Cadeia Lateral do Colesterol/imunologia , Feminino , Terapia de Reposição Hormonal/estatística & dados numéricos , Humanos , Menopausa Precoce/sangue , Noruega/epidemiologia , Polimorfismo de Nucleotídeo Único , Valor Preditivo dos Testes , Prevalência , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/imunologia , Sistema de Registros
18.
Climacteric ; 24(5): 438-443, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33522318

RESUMO

Prediction of premature ovarian insufficiency (POI) would be of substantial individual benefit, but being a heterogeneous and fluctuating condition, with an extensive range of complex etiologies and arbitrary diagnostic criteria, might make this seem foolhardy. However, contemporary and complementary genetic strategies assessing consanguineous and large POI pedigrees and cohorts with age at natural menopause have shown strong enrichment in genes regulating DNA damage repair, homologous recombination, and meiosis, processes that are critical to oogenesis and folliculogenesis. Recognition of the molecular architecture of POI and its contribution to baseline genotypic risk may enable these estimates to be refined further by estimation of the residual ovarian reserve. Increasing data derived from spontaneous and gonadotoxic-induced POI cohorts demonstrate the utility of anti-Müllerian hormone (AMH) to predict POI. This review presents current understanding of how genetics in combination with AMH may facilitate the prediction of POI.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Hormônio Antimülleriano , Feminino , Genótipo , Humanos , Menopausa Precoce/genética , Oogênese , Insuficiência Ovariana Primária/genética
19.
J Med Genet ; 58(3): 168-172, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32303603

RESUMO

BACKGROUND: The genetic causes of the majority of cases of female infertility caused by premature ovarian insufficiency (POI) are unknown. OBJECTIVE: To identify the genetic causes of POI in 110 patients. METHODS: Whole-exome sequencing was performed on 110 patients with POI, and putative disease-causative variants were validated by Sanger sequencing. Bioinformatic and in vitro functional analyses were performed for functional characterisation of the identified candidate disease-causative variants. RESULTS: We identified two homozygous variants (NM_001040274: c.150_151del (p.Ser52Profs*7), c.999A>G (p.Ile333Met)) in SYCP2L in two patients, which had co-segregated with POI in these families. Bioinformatic analysis predicted that the two variants are deleterious, and in vitro functional analysis showed that mutant SYCP2L proteins exhibited mislocalisation and loss of function. CONCLUSIONS: SYCP2L is a novel gene found to be responsible for human POI. Our findings provide a potential molecular marker for POI and improve the understanding of the genetic basis of female infertility.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Infertilidade Feminina/genética , Menopausa Precoce/genética , Insuficiência Ovariana Primária/genética , Adulto , Biologia Computacional , Feminino , Homozigoto , Humanos , Infertilidade Feminina/patologia , Mutação/genética , Linhagem , Insuficiência Ovariana Primária/epidemiologia , Insuficiência Ovariana Primária/patologia , Sequenciamento do Exoma
20.
Gynecol Endocrinol ; 37(3): 246-250, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33274669

RESUMO

OBJECTIVE: Forkhead box P3 (FOXP3), a transcription factor, is regarding critical regulator of the function of regulatory T (Treg) cells and plays a crucial role in the development of autoimmune diseases. Premature ovarian insufficiency (POI) is an autoimmune disease; however, little is known about the association between FOXP3 variants and the susceptibility to POI. METHODS: Long-range polymerase chain reaction was used to analyze complete FOXP3 gene sequences from 153 patients with POI. The frequencies of genotypes and alleles of the FOXP3 gene were compared between patients with POI and 269 East Asian women from the Genome Aggregation (gnomAD) database. RESULTS: Forty-three single-nucleotide polymorphisms (SNPs) were detected, including 25 known SNPs and 18 novel SNPs. The genotype distributions and allele frequencies of two known SNPs (rs17847094 and rs76798919) and three novel SNPs (NC_000023.11:g.49112832G > A, NC_000023.11:g.49112833G > A, and NC_000023.11:g.49120479CT > C) were significantly different between the two groups. Linkage disequilibrium and haplotype analyses of the rs57734889, rs2232365, rs3761548, and rs34629506 SNPs in FOXP3 were performed and compared, and the high D' (standardized disequilibrium coefficients) value indicated that these polymorphisms may contribute to the risk of POI. CONCLUSIONS: This study is the first to show that genetic variants in the regulatory regions of FOXP3 play a vital role in idiopathic POI in the Chinese population.


Assuntos
Fatores de Transcrição Forkhead/genética , Insuficiência Ovariana Primária/genética , Adulto , Povo Asiático/genética , Estudos de Casos e Controles , China/epidemiologia , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Menopausa Precoce/etnologia , Menopausa Precoce/genética , Polimorfismo de Nucleotídeo Único , Insuficiência Ovariana Primária/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...