Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 825
Filtrar
1.
Reproduction ; 168(4)2024 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-39042717

RESUMEN

In brief: Repro57 mice, bearing an Rnf212 gene mutation, exhibit infertility in both homozygous mutant males and females, revealing arrested spermatogenesis in males and investigating unclear mechanisms in females. The study highlights aneuploidy and altered kinetochore patterns in repro57 homozygous mutant oocytes, which impact later stages of embryo development. Abstract: Repro57 mice, induced with N-ethyl-N-nitrosourea and harboring a mutation in the Rnf212 gene, exhibit infertility in both homozygous mutant males and females. Rnf212 plays a crucial role in recombination and crossover designation. In male repro57 homozygous mutants, spermatocytes often degenerate during late prophase, and mature spermatozoa are absent in the seminiferous epithelium, indicating arrested spermatogenesis as the cause of infertility. Despite reports of infertility in Rnf212-knockout female mice, the specific mechanisms underlying infertility in female repro57 homozygous mutants remain elusive. This study investigates the chromosomal and kinetochore patterns of mature oocytes and their developmental potential following in vitro fertilization in female repro57 homozygous mutant mice. While all wild-type oocytes progress to metaphase II and exhibit euploidy, all repro57 homozygous mutant mouse oocytes display aneuploidy. Additionally, kinetochore distances in repro57 homozygous mutant oocytes exceed those observed in wild-type counterparts. Although no significant differences are noted in fertilization and early embryo development rates between wild-type and repro57 homozygous mutant mice, embryos derived from repro57 homozygous mutants exhibit significantly lower morula and blastocyst rates, accompanied by frequent cytokinesis failure and vacuole formation. These findings suggest that the premature segregation of sister chromatids in repro57 homozygous mutant mice adversely impacts the later stages of embryo development.


Asunto(s)
Desarrollo Embrionario , Homocigoto , Mutación , Oocitos , Animales , Femenino , Desarrollo Embrionario/genética , Ratones , Masculino , Oocitos/patología , Segregación Cromosómica , Ubiquitina-Proteína Ligasas/genética , Aneuploidia , Cinetocoros/metabolismo , Espermatogénesis/genética , Ratones Endogámicos C57BL
2.
J Assist Reprod Genet ; 41(9): 2279-2288, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38995507

RESUMEN

PURPOSE: To analyze the copy number variation (CNV) in the X-linked genes BCORL1, POF1B, and USP9X in idiopathic diminished ovarian reserve (DOR). METHODS: This case-control study included 47 women, 26 with DOR and 21 in the control group. Age, weight, height, BMI, and FSH level were evaluated, as well as antral follicle count (AFC), oocyte retrieval after controlled ovarian stimulation, and metaphase II (MII) oocytes. The CNVs of BCORL1, USP9X, and POF1B genes were measured by quantitative real time PCR (qPCR) using two reference genes, the HPRT1 (X-linked) and MFN2 (autosomal). Protein-protein interaction network and functional enrichment analysis were performed using the STRING database. RESULTS: The mean age was 36.52 ± 4.75 in DOR women and 35.38 ± 4.14 in control. Anthropometric measures did not differ between the DOR and control groups. DOR women presented higher FSH (p = 0.0025) and lower AFC (p < .0001), oocyte retrieval after COS (p = 0.0004), and MII oocytes (p < .0001) when compared to the control group. BCORL1 and POF1B did not differ in copy number between DOR and control. However, DOR women had more copies of USP9X than the control group (p = 0.028). CONCLUSION: The increase in the number of copies of the USP9X gene may lead to overexpression in idiopathic DOR and contribute to altered folliculogenesis and oocyte retrieval.


Asunto(s)
Variaciones en el Número de Copia de ADN , Reserva Ovárica , Ubiquitina Tiolesterasa , Humanos , Femenino , Reserva Ovárica/genética , Adulto , Variaciones en el Número de Copia de ADN/genética , Ubiquitina Tiolesterasa/genética , Estudios de Casos y Controles , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/patología , Recuperación del Oocito , Proteínas Represoras/genética , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Oocitos/patología
3.
J Assist Reprod Genet ; 41(8): 2011-2020, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38951359

RESUMEN

PURPOSE: Oocytes from women presenting primary ovarian insufficiency (POI) generate viable embryos at a lower rate than non-POI women, but the mechanisms responsible for the lower oocyte quality remain elusive. Due to the scarcity of human oocytes for research, animal models provide a promising way forward. We aimed at investigating the molecular events characterizing final maturation in POI oocytes in a well-defined POI-like bovine model. METHODS: Single-cell RNA-sequencing of bovine control and POI-like, GV, and MII oocytes (n = 5 per group) was performed. DEseq2 was used to identify differentially expressed genes. Further, a Gene set enrichment analysis and a transcriptomic meta-analysis between bovine and human oocytes were performed. RESULTS: In control cows, we found 2223 differentially expressed genes between the GV and MII stages. Specifically, the affected genes were related to RNA processing and transport, protein synthesis, organelle remodeling and reorganization, and metabolism. The meta-analysis with a set of young human oocytes at different maturation stages revealed 315 conserved genes through the GV-MII transition in cows and humans, mostly related to meiotic progression and cell cycle. Gene expression analysis between GV and MII of POI-like oocytes showed no differences in terms of differentially expressed genes, pointing towards a substantial failure to properly remodel the transcriptome in the POI model, and with the clustering analysis indicating that the cow's genetic background had a higher impact than the oocyte's maturation stage. CONCLUSION: Overall, we have identified and characterized a valuable animal model of POI, paving the way to identifying new molecular mechanisms involved in POI.


Asunto(s)
Meiosis , Oocitos , Insuficiencia Ovárica Primaria , Bovinos , Femenino , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/patología , Animales , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Oocitos/patología , Meiosis/genética , Humanos , Transcriptoma/genética , Modelos Animales de Enfermedad , Oogénesis/genética
4.
J Assist Reprod Genet ; 41(8): 1965-1976, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38954294

RESUMEN

PURPOSE: Oocyte maturation defect (OOMD) is a rare cause of in vitro fertilization failure characterized by the production of immature oocytes. Compound heterozygous or homozygous PATL2 mutations have been associated with oocyte arrest at the germinal vesicle (GV), metaphase I (MI), and metaphase II (MII) stages, as well as morphological changes. METHODS: In this study, we recruited three OOMD cases and conducted a comprehensive multiplatform laboratory investigation. RESULTS: Whole exome sequence (WES) revealed four diagnostic variants in PATL2, nonsense mutation c.709C > T (p.R237*) and frameshift mutation c.1486_1487delinsT (p.A496Sfs*4) were novel mutations that have not been reported previously. Furthermore, the pathogenicity of these variants was predicted using in silico analysis, which indicated detrimental effects. Molecular dynamic analysis suggested that the A496S variant disrupted the hydrophobic segment, leading to structural changes that affected the overall protein folding and stability. Additionally, biochemical and molecular experiments were conducted on cells transfected with wild-type (WT) or mutant PATL2 (p.R237* and p.A496Sfs*4) plasmid vectors. CONCLUSIONS: The results demonstrated that PATL2A496Sfs*4 and PATL2R237* had impacts on protein size and expression level. Interestingly, expression levels of specific genes involved in oocyte maturation and early embryonic development were found to be simultaneously deregulated. The findings in our study expand the variation spectrum of the PATL2 gene, provide solid evidence for counseling on future pregnancies in affected families, strongly support the application of in the diagnosis of OOMD, and contribute to the understanding of PATL2 function.


Asunto(s)
Secuenciación del Exoma , Infertilidad Femenina , Proteínas Nucleares , Oocitos , Oogénesis , Proteínas de Unión al ARN , Adulto , Femenino , Humanos , Codón sin Sentido/genética , Fertilización In Vitro , Mutación del Sistema de Lectura/genética , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Mutación/genética , Oocitos/crecimiento & desarrollo , Oocitos/patología , Oocitos/metabolismo , Oogénesis/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética
5.
J Assist Reprod Genet ; 41(8): 1955-1963, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38829516

RESUMEN

PURPOSE: To explore the pathogenesis of oocyte maturation defects. METHODS: Whole exome sequencing was conducted to identify potential variants, which were then confirmed within the pedigree through Sanger sequencing. The functional characterization of the identified variants responsible for the disease, including their subcellular localization, protein levels, and interactions with other proteins, was verified through transient transfection in HeLa cells in vitro. Additionally, we employed real-time RT-PCR and single-cell RNA sequencing to examine the impact of ZFP36L2 pathogenic variants on mRNA metabolism in both HeLa cells and mouse or human oocytes. RESULTS: A novel compound heterozygous variant in ZFP36L2 (c.186T > G, p.His62Gln and c.869 C > T, p.Pro290Leu) was discovered in a patient with oocyte maturation defects. Our findings indicate that these variants lead to compromised binding capacity of the ZFP36L2-CONT6L complex and impaired mRNA degradation in HeLa cells and mouse oocytes. Furthermore, we characterized the changes in the human oocyte transcriptome associated with ZFP36L2 variants, with a particular emphasis on cell division, mitochondrial function, and ribosome metabolism. CONCLUSIONS: This study broadens the mutation spectrum of ZFP36L2 and constitutes the first report of human oocyte transcriptome alterations linked to ZFP36L2 variants. In conjunction with existing knowledge of ZFP36L2, our research lays the groundwork for genetic counseling aimed at addressing female infertility.


Asunto(s)
Infertilidad Femenina , Oocitos , Humanos , Femenino , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Oocitos/patología , Ratones , Células HeLa , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Animales , Secuenciación del Exoma , Linaje , Heterocigoto , Oogénesis/genética , Tristetraprolina/genética , Tristetraprolina/metabolismo , Mutación/genética , Adulto
6.
J Assist Reprod Genet ; 41(5): 1233-1243, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38536595

RESUMEN

AIM: Abnormalities in oocyte maturation, fertilization, and early embryonic development are major causes of primary infertility in women who are undergoing IVF/ICSI attempts. Although many genetic factors responsible for these abnormal phenotypes have been identified, there are more additional pathogenic genes and variants yet to be discovered. Previous studies confirmed that bi-allelic PATL2 deficiency is an important factor for female infertility. In this study, 935 infertile patients with IVF/ICSI failure were selected for whole-exome sequencing, and 18 probands carrying PATL2 variants with a recessive inheritance pattern were identified. METHODS: We estimated that the prevalence contributed by PATL2 was 1.93% (18/935) in our study cohort. RESULTS: 15 novel variants were found in those families, including c.1093C > T, c.1609dupA, c.1204C > T, c.643dupG, c.877-2A > G, c.1228C > G, c.925G > A, c.958G > A, c.4A > G, c.1258T > C, c.1337G > A, c.1264dupA, c.88G > T, c.1065-2A > G, and c.1271T > C. The amino acids altered by the corresponding variants were highly conserved in mammals, and in silico analysis and 3D molecular modeling suggested that the PATL2 mutants impaired the physiologic function of the resulting proteins. Diverse clinical phenotypes, including oocyte maturation defect, fertilization failure, and early embryonic arrest might result from different variants of PATL2. CONCLUSIONS: These results expand the spectrum of PATL2 variants and provide an important reference for genetic counseling for female infertility, and they increase our understanding of the mechanisms of oocyte maturation arrest caused by PATL2 deficiency.


Asunto(s)
Secuenciación del Exoma , Fertilización In Vitro , Infertilidad Femenina , Mutación , Proteínas Nucleares , Fenotipo , Proteínas de Unión al ARN , Inyecciones de Esperma Intracitoplasmáticas , Adulto , Femenino , Humanos , Embarazo , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Mutación/genética , Oocitos/crecimiento & desarrollo , Oocitos/patología , Linaje , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética
7.
Arch Gynecol Obstet ; 309(5): 2143-2152, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38494510

RESUMEN

PURPOSE: What are the reproductive outcomes of women who had fertility preservation (FP) using either oocyte or embryo vitrification after fertility-sparing surgery (FSS) for a borderline ovarian tumor (BOT)? METHODS: A retrospective, single-center cohort study was conducted between January 2013 and December 2021. Patients with BOT who resorted to FP by vitrifying oocytes or embryos were included. Both clinical and reproductive parameters were reviewed. The primary outcome was live birth. RESULTS: In total, thirteen patients who performed 31 FP cycles were included. Of those, six patients achieved eight live births after a mean follow-up period of 79 months. Three further pregnancies are still ongoing. All pregnancies/live births were obtained without using their cryopreserved oocytes or embryos. CONCLUSION: Women who had FSS for BOT have favorable prospects of live offspring, even without the need to use their cryopreserved material. Fertility preservation in patients with BOT has to be considered as a tool to mitigate the risk of infertility that may arise in case of BOT recurrence requiring castrating surgery.


Asunto(s)
Preservación de la Fertilidad , Neoplasias Ováricas , Embarazo , Humanos , Femenino , Estudios Retrospectivos , Estudios de Cohortes , Criopreservación , Oocitos/patología , Neoplasias Ováricas/cirugía , Neoplasias Ováricas/patología
8.
Aging Dis ; 15(2): 804-823, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37611899

RESUMEN

Premature ovarian insufficiency (POI), which is defined as loss of ovarian function that occurs before the age of 40, causes menstrual disturbances, infertility, and diverse health problems in females. Despite the limited understanding of the molecular basis underlying POI pathology, we had previously demonstrated that the cooperation of miR-106a and FBXO31 plays a pivotal role in diminished ovarian reserve (DOR), with FBXO31 serving as a putative target of miR-106a. In this study, we found that FBXO31 is aberrantly expressed in granulosa cells of POI patients, leading to accumulated reactive oxygen species (ROS) and cell apoptosis via the p53/ROS pathway. Furthermore, our results demonstrated that high levels of FBXO31 in mouse ovaries impair oocyte quality. Our study revealed that FBXO31 may serve as a novel indicator and play a significant role in the etiology of POI.


Asunto(s)
Proteínas F-Box , Menopausia Prematura , MicroARNs , Insuficiencia Ovárica Primaria , Ratones , Femenino , Animales , Humanos , Especies Reactivas de Oxígeno , Insuficiencia Ovárica Primaria/etiología , Oocitos/patología , Proteínas Supresoras de Tumor , Proteínas F-Box/genética
9.
BMC Med Genomics ; 16(1): 271, 2023 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-37904145

RESUMEN

Tubulin beta-8 (TUBB8) is expressed exclusively in the oocyte and early embryo, encoding a beta-tubulin polypeptide that participates in the assembly of microtubules. TUBB8 was first attributed to being responsible for oocyte MI arrest. Further studies have demonstrated that patients with different pathogenic variants have variable phenotypes. We report a TUBB8 variant (c.10 A > C) in two siblings who presented different clinical features of primary infertility. The younger sister showed severe oocyte maturation arrest with abnormal morphology, whereas a few mature oocytes and zygotes could be retrieved from the older sister, but no embryo was available for transfer. This variant was previously reported without in vitro functional assays. In the present study, RT‒qPCR and western blot analyses revealed that c.10 A > C reduces TUBB8 mRNA and protein levels; however, immunofluorescence demonstrated that this variant does not change the localization of the protein. These findings confirm the pathogenicity of the c.10 A > C variant and support the relationship between the variant and phenotype in the patients.


Asunto(s)
Infertilidad Femenina , Tubulina (Proteína) , Femenino , Humanos , Variación Biológica Poblacional , Infertilidad Femenina/genética , Oocitos/metabolismo , Oocitos/patología , Hermanos , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
10.
Eur J Obstet Gynecol Reprod Biol ; 290: 93-100, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37757729

RESUMEN

OBJECTIVE: To the best of our knowledge, the available evidence on the effect and efficacy of controlled ovarian stimulation (COS) in this group of patients remains poorly reported. Concerns related to the impact of stimulation to cancer progression and recurrence, as well as the risk of disease dissemination during egg collection, might explain the aforementioned trend. METHODS: Overall, our FP Service received 192 gynaecological referrals, between 2005 and 2021, regarding gynaecologic conditions mainly cancer related. A total of 68 (35.4%) patients underwent COS. These patients were diagnosed with the following gynaecologic pathologies: 33 cases (48,5%) of cervical cancer were noted (stage 1b1-2b), 25 ovarian pathology (36.7%), 9 cases (13.2%) of endometrial cancer, and a single case of vaginal cancer (1.5%). RESULTS: The mean age of patients attending the fertility preservation service was 31.5 (std 5.8). The patients presenting to their initial appointment with a mean BMI 24.5 (IQR 6.9) and a median AFC of 12 (IQR 13). The mean duration of COS was 11 days (IQR 3), and the median dose of gonadotrophins was calculated at 300 IU (IQR 75 IU). In 95.4% of the cases, GnRH agonist was used as a trigger for final maturation. The median number of follicles measuring more than 14 mm at the time of trigger was 11 (IQR 8), whereas the median number of oocytes collected was 11 (IQR 9). The complication rate was reported at less than 2%. So far, one in four women of this FP group (17/68, 25% of the overall group) returned to our service to claim their cryopreserved eggs/embryos and successful livebirths were reported in 58.8% of this sample (10/17 cases). The mean time to return to use their oocytes/embryos was 36 months (min value 16 months - max value 85 months). There was no significant difference in mortality rate between patients who received FP vs those who did not (hazard ratio of mortality was estimated at 0.91 (p-value 0.88)). CONCLUSION: Based on our findings, ovarian stimulation for patients presenting with gynaecologic malignancy is a safe and efficient method of fertility preservation. Undoubtedly, the sample size is limited, however our results are reassuring and highlight the efficacy of COS for the purpose of FP based on data coming from the largest Assisted Conception Unit of the South-East of the UK.


Asunto(s)
Preservación de la Fertilidad , Neoplasias de los Genitales Femeninos , Humanos , Femenino , Preservación de la Fertilidad/métodos , Criopreservación/métodos , Oocitos/patología , Inducción de la Ovulación/efectos adversos , Inducción de la Ovulación/métodos , Estudios Retrospectivos , Recuperación del Oocito
11.
Zygote ; 31(4): 316-341, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37212058

RESUMEN

Fertilization failure (FF) and zygotic arrest after ICSI have a huge effect on both patients and clinicians, but both problems are usually unexpected and cannot be properly diagnosed. Fortunately, in recent years, gene sequencing has allowed the identification of multiple genetic variants underlying failed ICSI outcomes, but the use of this approach is still far from routine in the fertility clinic. In this systematic review, the genetic variants associated with FF, abnormal fertilization and/or zygotic arrest after ICSI are compiled and analyzed. Forty-seven studies were included. Data from 141 patients carrying 121 genetic variants affecting 16 genes were recorded and analyzed. In total, 27 variants in PLCZ1 (in 50 men) and 26 variants in WEE2 (in 24 women) are two of the factors related to oocyte activation failure that could explain a high percentage of male-related and female-related FF. Additional variants identified were reported in WBP2NL, ACTL9, ACTLA7, and DNAH17 (in men), and TUBB8, PATL2, TLE6, PADI6, TRIP13, BGT4, NLRP5, NLRP7, CDC20 and ZAR1 (in women). Most of these variants are pathogenic or potentially pathogenic (89/121, 72.9%), as demonstrated by experimental and/or in silico approaches. Most individuals carried bi-allelic variants (89/141, 63.1%), but pathogenic variants in heterozygosity have been identified for PLCZ1 and TUBB8. Clinical treatment options for affected individuals, such as chemical-assisted oocyte activation (AOA) or PLCZ1 cRNA injection in the oocyte, are still experimental. In conclusion, a genetic study of known pathogenic variants may help in diagnosing recurrent FF and zygotic arrest and guide patient counselling and future research perspectives.


Asunto(s)
Inyecciones de Esperma Intracitoplasmáticas , Cigoto , Masculino , Femenino , Animales , Oocitos/patología , Fertilización/genética
12.
J Assist Reprod Genet ; 40(3): 473-480, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36752941

RESUMEN

PURPOSE: The objective of the present study was to evaluate whether oocyte vitrification following controlled ovarian stimulation (COS) for fertility preservation (FP) delays the initiation of neoadjuvant chemotherapy (NAC) for breast cancer (BC) as compared to in vitro maturation (IVM). METHODS: We performed a retrospective cohort study including all BC patients eligible for oocyte vitrification following COS or in vitro maturation (IVM) before initiation of NAC between January 2016 and December 2020. The inclusion criteria were female patients aged between 18 and 40, with confirmed non metastatic BC, with indication of NAC, who have had oocyte retrieval for FP after COS, or IVM + / - cryopreservation of ovarian tissue (OTC). Various time points related to cancer diagnosis, FP, or chemotherapy were obtained from a medical record review. RESULTS: A total of 197 patients with confirmed BC who had oocyte retrieval following COS (n = 57) or IVM + / - OTC (n = 140) for FP prior to NAC were included. Overall, the average time from cancer diagnosis to chemotherapy start was similar between patients having undergone COS or IVM before oocyte vitrification (37.3 ± 13.8 vs. 36. 8 ± 13.5 days; p = 0.89). CONCLUSIONS: The indication of NAC for BC should not be considered as an impediment to urgent COS for oocyte vitrification for FP.


Asunto(s)
Preservación de la Fertilidad , Neoplasias , Femenino , Masculino , Humanos , Vitrificación , Estudios Retrospectivos , Terapia Neoadyuvante , Oocitos/patología , Criopreservación , Recuperación del Oocito , Neoplasias/patología , Técnicas de Maduración In Vitro de los Oocitos
13.
Genes (Basel) ; 13(11)2022 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-36360157

RESUMEN

Various pathogenic factors can lead to oogenesis failure and seriously affect both female reproductive health and fertility. Genetic factors play an important role in folliculogenesis and oocyte maturation but still need to be clarified. Oocyte maturation is a well-organized complex process, regulated by a large number of genes. Pathogenic variants in these genes as well as aneuploidy, defects in mitochondrial genome, and other genetic and epigenetic factors can result in unexplained infertility, early pregnancy loss, and recurrent failures of IVF/ICSI programs due to poor ovarian response to stimulation, oocyte maturation arrest, poor gamete quality, fertilization failure, or early embryonic developmental arrest. In this paper, we review the main genes, as well as provide a description of the defects in the mitochondrial genome, associated with female infertility.


Asunto(s)
Fertilización In Vitro , Infertilidad Femenina , Embarazo , Humanos , Femenino , Oogénesis/genética , Oocitos/patología , Infertilidad Femenina/genética , Desarrollo Embrionario/genética
14.
Eur J Obstet Gynecol Reprod Biol ; 279: 132-139, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36335766

RESUMEN

PURPOSE: Variations in many genes may lead to the occurrence of oocyte maturation defectsand female infertility. The objective was to describe newly discovered mutations in TUBB8 and ZP3, and to characterise the accompanying spectrum of phenotypes and modes of inheritance. METHODS: TUBB8 and ZP3 were sequenced from genomic DNA samples extracted from peripheral blood of patients and their family members by the whole-exome sequencing. The TUBB8 and ZP3 sequences are then aligned with cryptographic software to identify rare variations. Sanger sequencing and mass spectrometry were used to validate mutations. ExAC database was used to retrieve the frequency of corresponding mutations. PolyPhen-2 and PROVEAN were analyzed for mutations using silicon. RESULTS: We identified Three novel mutations and two known variant in TUBB8 and ZP3 associated with maturation in five families, and fertilization and developmental arrest are in these patients. These mutations include four heterozygous mutations in TUBB8 (c.730G > A, p.Gly244Ser, c.124C > G, p.Leu42Val, c.1172G > T, p.Arg391Leu and c.178G > A, p.Val60Met), and a heterozygous mutation in ZP3 (c.400G > A, p.Ala134Thr). Among them, these variants of TUBB8 were highly conserved among primates. CONCLUSION: As far as we know, the TUBB8 mutations detected in our study at four sites have not been reported before, and the variant of ZP3 has been published as pathogenic. Our findings extend the known mutant spectrum of TUBB8 and ZP3, and provide insights into the etiology of infertility in human women. The exact molecular mechanism has not been analyzed and should be further investigated in the future.


Asunto(s)
Infertilidad Femenina , Animales , Humanos , Femenino , Infertilidad Femenina/genética , Tubulina (Proteína)/genética , Oogénesis/genética , Oocitos/patología , Mutación , Glicoproteínas de la Zona Pelúcida/genética
15.
Reprod Biomed Online ; 45(4): 763-778, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35945106

RESUMEN

Ovarian tissue cryopreservation and subsequent autotransplantation is a successful technique for fertility preservation in oncological patients. However, there are concerns regarding safety, as the graft may contain malignant cells that could lead to the reintroduction of cancer. To circumvent this problem several experimental strategies are being pursued. This systematic review was conducted to provide an overview of the strategies aiming to safely use cryopreserved human ovarian tissue to restore fertility after cancer. Thirty-one studies were included, covering five different experimental strategies: (i) in-vitro maturation of oocytes, (ii) constructing an artificial ovary as a scaffold for reseeding pre-antral follicles, (iii) purging strategies aimed at the eradication of contaminating malignant cells, (iv) maturation of oocytes by xenotransplantation, and (v) stem cell-based oogenesis. These strategies to circumvent the reintroduction of cancer cells through ovarian tissue autotransplantation are being developed, but so far have not reached the stage of clinical trials. Further research is required to establish their risks and effectiveness while the ethical aspects associated with these strategies also need to be discussed. Despite the fact that these experimental procedures are still under development, they might provide safe fertility restoration options for oncological patients in the future.


Asunto(s)
Preservación de la Fertilidad , Neoplasias , Criopreservación/métodos , Femenino , Preservación de la Fertilidad/métodos , Humanos , Oocitos/patología , Oogénesis , Ovario/trasplante
16.
Nat Cancer ; 3(8): 1-13, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36008687

RESUMEN

Loss of fertility is a major concern for female reproductive-age cancer survivors, since a common side-effect of conventional cytotoxic cancer therapies is permanent damage to the ovary. While immunotherapies are increasingly becoming a standard of care for many cancers-including in the curative setting-their impacts on ovarian function and fertility are unknown. We evaluated the effect of immune checkpoint inhibitors blocking programmed cell death protein ligand 1 and cytotoxic T lymphocyte-associated antigen 4 on the ovary using tumor-bearing and tumor-free mouse models. We find that immune checkpoint inhibition increases immune cell infiltration and tumor necrosis factor-α expression within the ovary, diminishes the ovarian follicular reserve and impairs the ability of oocytes to mature and ovulate. These data demonstrate that immune checkpoint inhibitors have the potential to impair both immediate and future fertility, and studies in women should be prioritized. Additionally, fertility preservation should be strongly considered for women receiving these immunotherapies, and preventative strategies should be investigated in future studies.


Asunto(s)
Preservación de la Fertilidad , Neoplasias , Animales , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia/efectos adversos , Ratones , Oocitos/patología
17.
Reprod Biomed Online ; 45(3): 508-518, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35798635

RESUMEN

RESEARCH QUESTION: Can a methodology be developed for case selection and whole-exome sequencing (WES) analysis of women who are infertile owing to recurrent oocyte maturation defects (OOMD) and/or preimplantation embryo lethality (PREMBL)? DESIGN: Data were collected from IVF patients attending the Istanbul Memorial Hospital (2015-2021). A statistical methodology to identify infertile endophenotypes (recurrent low oocyte maturation rate, low fertilization rate and preimplantation developmental arrest) was developed using a large IVF dataset (11,221 couples). Twenty-eight infertile women with OOMD/PREMBL were subsequently enrolled for WES on their genomic DNA. Pathogenic variants were prioritized using a custom-made bioinformatic pipeline set to minimize false-positive discoveries through resampling in control cohorts (the Human Genome Diversity Project and 1343 whole-exome sequences from oocyte donors). Individual single-cell RNA sequencing data from 18 human metaphase II (MII) oocytes and antral granulosa cells was used for genome-wide validation. WES and bioinformatics were performed at Igenomix and the National Research Council, Italy. RESULTS: Variant prioritization analysis identified 265 unique variants in 248 genes (average 22.4 per sample). Of the genes harbouring high-impact variants 78% were expressed by MII oocytes and/or antral granulosa cells, significantly higher than for random sample of controls (odds ratio = 5, Fisher's exact P = 0.0004). Seven of the 28 women (25%) were homozygous carriers of missense pathogenic variants in known candidate genes for OOMD/PREMBL, including PATL2, NLRP5 (n = 2),TLE6, PADI6, TUBB8 and TRIP13. Furthermore, novel gene-disease associations were identified. In fact, one woman with a low oocyte maturation rate was a homozygous carrier of high-impact variants in ENSA, an essential gene for prophase I meiotic transition in mice. CONCLUSIONS: This analytical framework could reveal known and new genes associated with isolated recurrent OOMD/PREMBL, providing essential indications for scaling this strategy to larger studies.


Asunto(s)
Infertilidad Femenina , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Proteínas de Ciclo Celular/genética , Exoma , Femenino , Humanos , Infertilidad Femenina/genética , Ratones , Oocitos/patología , Oogénesis , Tubulina (Proteína)/genética , Secuenciación del Exoma
18.
J Assist Reprod Genet ; 39(8): 1901-1908, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35834089

RESUMEN

PURPOSE: Oocyte death is a severe clinical phenotype that causes female infertility and recurrent in vitro fertilization and intracytoplasmic sperm injection failure. We aimed to identify pathogenic variants in a female infertility patient with oocyte death phenotype. METHODS: Sanger sequencing was performed to screen PANX1 variants in the affected patient. Western blot analysis was used to check the effect of the variant on PANX1 glycosylation pattern in vitro. RESULTS: We identified a novel PANX1 variant (NM_015368.4 c.86G > A, (p. Arg29Gln)) associated with the phenotype of oocyte death in a non-consanguineous family. This variant displayed an autosomal dominant inheritance pattern with reduced penetrance. Western blot analysis confirmed that the missense mutation of PANX1 (c.86G > A) altered the glycosylation pattern in HeLa cells. Moreover, the mutation effects on the function of PANX1 were weaker than recently reported variants. CONCLUSION: Our findings expand the inheritance pattern of PANX1 variants to an autosomal dominant mode with reduced penetrance and enrich the variational spectrum of PANX1. These results help us to better understand the genetic basis of female infertility with oocyte death.


Asunto(s)
Infertilidad Femenina , Conexinas/genética , Femenino , Células HeLa , Heterocigoto , Humanos , Infertilidad Femenina/patología , Masculino , Proteínas del Tejido Nervioso/genética , Oocitos/patología , Semen
19.
Rev Prat ; 72(6): 621-626, 2022 Jun.
Artículo en Francés | MEDLINE | ID: mdl-35899661

RESUMEN

FERTILITY PRESERVATION BEFORE OVARIAN MALIGNANCY TREATMENT While most ovarian epithelial malignancies affect postmenopausal women, 12% occur in reproductive age patients. In addition, borderline ovarian tumors and rare non epithelial ovarian tumors are diagnosed in young patients as well. The prognosis of early-stage epithelial tumors, non-epithelial and frontier tumors is good. Increased knowledge in this specific field now allows the development of fertility preservation strategies. They include conservative surgery when applicable, associated with oocyte and / or ovarian tissue cryopreservation. Indications remain limited, and any decision must be validated by a multidisciplinary expert committee. The different strategies depend on specific tumoral or genetic context.


PRÉSERVATION DE LA FERTILITÉ AVANT TRAITEMENT D'UN CANCER DE L'OVAIRE Le cancer épithélial de l'ovaire atteint en majorité les patientes ménopausées. Cependant, 12 % des patientes ont moins de 44 ans. Les tumeurs frontières de l'ovaire et les tumeurs rares non épithéliales sont diagnostiquées plus fréquemment chez les femmes en âge de procréer. Le pronostic des stades précoces et des tumeurs non épithéliales est favorable. Les avancées scientifiques permettent de développer des stratégies de préservation de la fertilité, qui reposent d'abord sur la possibilité d'une chirurgie conservatrice et sur la préservation de gamètes ou de tissus germinaux. Les indications restent néanmoins limitées, et toute décision doit être discutée en réunion de concertation pluridisciplinaire (RCP) de recours. Les différentes stratégies dépendent du contexte tumoral ou génétique.


Asunto(s)
Preservación de la Fertilidad , Neoplasias Ováricas , Criopreservación , Femenino , Humanos , Oocitos/patología , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía
20.
Cell Death Dis ; 13(7): 579, 2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35787614

RESUMEN

Endometriosis (EMs) occurs in approximately 50% of women with infertility. The main causes of EMs-related infertility are follicle dysplasia and reduced oocyte quality. Iron overload occurs in ovarian follicular fluid (FF) of patients with EMs, and this condition is associated with oocyte maturation disorder. However, the underlying molecular mechanism remains largely unknown. In the present study, we identified the mechanism underlying ferroptosis in ovarian granulosa cells and oocyte maturation failure in EMs based on a retrospective review of in vitro fertilization/intracytoplasmic sperm injection-frozen embryo transfer outcomes in infertile patients with EMs. Mouse granulosa cells were treated with EMs-related infertile patients' follicular fluid (EMFF) in vitro. Western blot analysis, quantitative polymerase chain reaction, fluorescence staining, and transmission electron microscopy were used to assess granulosa cells ferroptosis. The effects of exosomes were examined by nanoparticle tracking analysis, RNA-seq, and Western blot analysis. Finally, the therapeutic values of vitamin E and iron chelator (deferoxamine mesylate) in vivo were evaluated in an EMs-related infertility model. Patients with ovarian EMs experienced poorer oocyte fertility than patients with non-ovarian EMs. We observed that EMFF with iron overload-induced granulosa cell ferroptosis in vitro and in vivo. Mechanically, nuclear receptor coactivator four-dependent ferritinophagy was involved in this process. Notably, granulosa cells undergoing ferroptosis further suppressed oocyte maturation by releasing exosomes from granulosa cells. In therapeutic studies, vitamin E and iron chelators effectively alleviated EMs-related infertility models. Our study indicates a novel mechanism through which EMFF with iron overload induces ferroptosis of granulosa cells and oocyte dysmaturity in EMs-related infertility, providing a potential therapeutic strategy for EMs-related infertility.


Asunto(s)
Endometriosis , Ferroptosis , Sobrecarga de Hierro , Animales , Deferoxamina/farmacología , Endometriosis/complicaciones , Femenino , Líquido Folicular , Células de la Granulosa/citología , Humanos , Infertilidad Femenina/complicaciones , Hierro , Sobrecarga de Hierro/complicaciones , Ratones , Oocitos/patología , Vitamina E/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA