Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Assist Reprod Genet ; 41(3): 751-756, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38277113

RESUMEN

PURPOSE: To investigate the genetic etiology of patients with female infertility. METHODS: Whole Exome Sequencing was performed on genomic DNA extracted from the patient's blood. Exome data were filtered for damaging rare biallelic variants in genes with possible roles in reproduction. Sanger sequencing was used to validate the selected variants and segregate them in family members. RESULTS: A novel homozygous likely pathogenic variant, c.626G>A, p.Trp209*, was identified in the TERB1 gene of the patient. Additionally, we report a second homozygous pathogenic TERB1 variant, c.1703C>G, p.Ser568*, in an infertile woman whose azoospermic brother was previously described to be homozygous for her variant. CONCLUSIONS: Here, we report for the first time two homozygous likely pathogenic and pathogenic TERB1 variants, c.626G>A, p.Trp209* and c.1703C>G, p.Ser568*, respectively, in two unrelated women with primary infertility. TERB1 is known to play an essential role in homologous chromosome movement, synapsis, and recombination during the meiotic prophase I and has an established role in male infertility in humans. Our data add TERB1 to the shortlist of Meiosis I genes associated with human infertility in both sexes.


Asunto(s)
Azoospermia , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Infertilidad Masculina , Femenino , Humanos , Azoospermia/genética , Proteínas de Ciclo Celular/genética , Homocigoto , Infertilidad Masculina/genética , Meiosis , Proteínas de Unión al ADN/genética
2.
Hum Mutat ; 43(12): 1732-1744, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35842788

RESUMEN

Hydatidiform mole (HM) is an abnormal human pregnancy characterized by excessive growth of placental trophoblasts and abnormal early embryonic development. Following a first such abnormal pregnancy, the risk for women of successive molar pregnancies significantly increases. To date variants in seven maternal-effect genes have been shown to cause recurrent HMs (RHM). NLRP7 is the major causative gene for RHM and codes for NOD-like receptor (NLR) family pyrin domain containing 7, which belongs to a family of proteins involved in inflammatory disorders. Since its identification, all NLRP7 variants have been recorded in Infevers, an online registry dedicated to autoinflammatory diseases (https://infevers.umai-montpellier.fr/web/). Here, we reviewed published and unpublished recessive NLRP7 variants associated with RHM, scored their pathogenicity according to the American College of Medical Genetics classification, and recapitulated all functional studies at the level of both the patients and the conceptions. We also provided data on further variant analyses of 32 patients and genotypes of 36 additional molar pregnancies. This comprehensive review integrates published and unpublished data on NLRP7 and aims at guiding geneticists and clinicians in variant interpretation, genetic counseling, and management of patients with this rare condition.


Asunto(s)
Mola Hidatiforme , Neoplasias Uterinas , Humanos , Femenino , Embarazo , Proteínas Adaptadoras Transductoras de Señales/genética , Placenta , Mola Hidatiforme/genética , Genotipo , Neoplasias Uterinas/genética
3.
Reprod Biomed Online ; 45(1): 125-134, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35523710

RESUMEN

RESEARCH QUESTION: What is the genetic cause of sporadic and recurrent pregnancy loss and does the frequency and nature of chromosomal abnormalities play a role? Types and frequency of all identifiable chromosomal abnormalities were determined to inform our understanding, medical management and recurrence risk for patients experiencing pregnancy loss. DESIGN: Genome-wide single-nucleotide polymorphism-based chromosomal microarray (SNP-CMA) were used to evaluate 24,900 products of conception samples from various forms of pregnancy losses. RESULTS: Sporadic miscarriage (64.7%) or recurrent pregnancy loss (RPL) (22%) were the most common referrals. Clinically significant abnormalities were observed in 55.8% (13,910) of samples, variants of uncertain significance in 1.8%, and normal results in 42.4%. In addition to autosomal trisomies (in 36% of samples), polyploidy and large segmental imbalances were identified in 7.8% and 2.8% of samples, respectively. Analysis of sequential samples from 1103 patients who had experienced RPL provided important insight into possible predispositions to RPL. CONCLUSIONS: This expansive chromosomal microarray analyses of pregnancy loss samples illuminates our understanding of the full spectrum, relative frequencies and the role of genomic abnormalities in pregnancy loss. The empiric observations described here provide useful insight for clinicians and highlight the importance of high-resolution genomic testing for comprehensive evaluation and risk assessment of individuals experiencing pregnancy loss.


Asunto(s)
Aborto Habitual , Aborto Inducido , Aborto Habitual/genética , Aberraciones Cromosómicas , Femenino , Pruebas Genéticas , Genómica , Humanos , Embarazo
4.
Am J Hum Genet ; 103(5): 740-751, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388401

RESUMEN

Androgenetic complete hydatidiform moles are human pregnancies with no embryos and affect 1 in every 1,400 pregnancies. They have mostly androgenetic monospermic genomes with all the chromosomes originating from a haploid sperm and no maternal chromosomes. Androgenetic complete hydatidiform moles were described in 1977, but how they occur has remained an open question. We identified bi-allelic deleterious mutations in MEI1, TOP6BL/C11orf80, and REC114, with roles in meiotic double-strand breaks formation in women with recurrent androgenetic complete hydatidiform moles. We investigated the occurrence of androgenesis in Mei1-deficient female mice and discovered that 8% of their oocytes lose all their chromosomes by extruding them with the spindles into the first polar body. We demonstrate that Mei1-/- oocytes are capable of fertilization and 5% produce androgenetic zygotes. Thus, we uncover a meiotic abnormality in mammals and a mechanism for the genesis of androgenetic zygotes that is the extrusion of all maternal chromosomes and their spindles into the first polar body.


Asunto(s)
Andrógenos/genética , Mola Hidatiforme/genética , Mutación/genética , Alelos , Animales , Cromosomas/genética , Femenino , Humanos , Masculino , Mamíferos/genética , Ratones , Ratones Endogámicos C57BL , Oocitos/patología , Embarazo , Cigoto/patología
5.
Clin Genet ; 99(6): 823-828, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33583041

RESUMEN

Recurrent hydatidiform moles (RHMs) are human pregnancies with abnormal embryonic development and hyperproliferating trophoblast. Biallelic mutations in NLRP7 and KHDC3L, members of the subcortical maternal complex (SCMC), explain the etiology of RHMs in only 60% of patients. Here we report the identification of seven functional variants in a recessive state in three SCMC members, five in NLRP7, one in NLRP5, and one in PADI6. In NLRP5, we report the first patient with RHMs and biallelic mutations. In PADI6, the patient had four molar pregnancies, two of which had fetuses with various abnormalities including placental mesenchymal dysplasia and intra-uterine growth restriction, which are features of Beckwith-Wiedemann syndrome and Silver Russell syndrome, respectively. Our findings corroborate recent studies and highlight the common oocyte origin of all these conditions and the continuous spectrum of abnormalities associated with deficiencies in the SCMC genes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Autoantígenos/genética , Mola Hidatiforme/genética , Proteínas Mitocondriales/genética , Mutación/genética , Recurrencia Local de Neoplasia/genética , Proteínas Nucleares/genética , Arginina Deiminasa Proteína-Tipo 6/genética , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/patología , Femenino , Humanos , Mola Hidatiforme/patología , Recurrencia Local de Neoplasia/patología , Oocitos/patología , Placenta/patología , Embarazo , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
6.
J Assist Reprod Genet ; 38(7): 1879-1886, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33751332

RESUMEN

PURPOSE: To investigate the frequency of a founder mutation in NLRP7, L750V, in independent cohorts of Mexican patients with recurrent hydatidiform moles (RHMs). METHODS: Mutation analysis was performed by Sanger sequencing on DNA from 44 unrelated Mexican patients with RHMs and seven molar tissues from seven additional unrelated patients. RESULTS: L750V was present in homozygous or heterozygous state in 37 (86%) patients and was transmitted on the same haplotype to patients from different states of Mexico. We also identified a second founder mutation, c.2810+2T>G in eight (18.1%) patients, and a novel premature stop-codon mutation W653*. CONCLUSION: Our data confirm the strong founder effect for L750V, which appears to be the most common mutation in NLRP7. We also report on six healthy live births to five patients with biallelic NLRP7 mutations, two from spontaneous conceptions and four from donated ovum and discuss our recommendations for DNA testing and genetic counseling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Efecto Fundador , Mola Hidatiforme/genética , Mutación , Femenino , Haplotipos , Heterocigoto , Humanos , Nacimiento Vivo , México , Polimorfismo de Nucleótido Simple , Embarazo
8.
Mod Pathol ; 33(5): 880-892, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31857680

RESUMEN

Hydatidiform mole (HM) is an aberrant human pregnancy characterized by excessive trophoblastic proliferation and abnormal embryonic development. HM has two morphological types, complete (CHM) and partial (PHM), and non-recurrent ones have three genotypic types, androgenetic monospermic, androgenetic dispermic, and triploid dispermic. Most available studies on risk factors predisposing to different types of HM and their malignant transformation mainly suffer from the lack of comprehensive genotypic analysis of large cohorts of molar tissues combined with accurate postmolar hCG follow-up. Moreover, 10-20% of patients with one HM have at least one non-molar miscarriage, which is higher than the frequency of two pregnancy losses in the general population (2-5%), suggesting a common genetic susceptibility to HM and miscarriages. However, the underlying causes of the miscarriages in these patients are unknown. Here, we comprehensively analyzed 204 HM, mostly from patients referred to the Quebec Registry of Trophoblastic Diseases and for which postmolar hCG monitoring is available, and 30 of their non-molar miscarriages. We revisited the risk of maternal age and neoplastic transformation across the different HM genotypic categories and investigated the presence of chromosomal abnormalities in their non-molar miscarriages. We confirm that androgenetic CHM is more prone to gestational trophoblastic neoplasia (GTN) than triploid dispermic PHM, and androgenetic dispermic CHM is more prone to high-risk GTN and choriocarcinoma (CC) than androgenetic monospermic CHM. We also confirm the association between increased maternal age and androgenetic CHM and their malignancies. Most importantly, we demonstrate for the first time that patients with an HM and miscarriages are at higher risk for aneuploid miscarriages [83.3%, 95% confidence interval (CI): 0.653-0.944] than women with sporadic (51.5%, 95% CI: 50.3-52.7%, p value = 0.0003828) or recurrent miscarriages (43.8%, 95% CI: 40.7-47.0%, p value = 0.00002). Our data suggest common genetic female germline defects predisposing to HM and aneuploid non-molar miscarriages in some patients.


Asunto(s)
Mola Hidatiforme/genética , Neoplasias Uterinas/genética , Aborto Habitual/genética , Adulto , Femenino , Genotipo , Humanos , Edad Materna , Persona de Mediana Edad , Embarazo , Factores de Riesgo
9.
Mod Pathol ; 31(7): 1116-1130, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29463882

RESUMEN

Hydatidiform mole is an aberrant human pregnancy characterized by early embryonic arrest and excessive trophoblastic proliferation. Recurrent hydatidiform moles are defined by the occurrence of at least two hydatidiform moles in the same patient. Fifty to eighty percent of patients with recurrent hydatidiform moles have biallelic pathogenic variants in NLRP7 or KHDC3L. However, in the remaining patients, the genotypic types of the moles are unknown. We characterized 80 new hydatidiform mole tissues, 57 of which were from patients with no mutations in the known genes, and we reviewed the genotypes of a total of 123 molar tissues. We also reviewed mutation analysis in 113 patients with recurrent hydatidiform moles. While all hydatidiform moles from patients with biallelic NLRP7 or KHDC3L mutations are diploid biparental, we demonstrate that those from patients without mutations are highly heterogeneous and only a small minority of them are diploid biparental (8%). The other mechanisms that were found to recur in patients without mutations are diploid androgenetic monospermic (24%) and triploid dispermic (32%); the remaining hydatidiform moles were misdiagnosed as moles due to errors in the analyses and/or their unusual mechanisms. We compared three parameters of genetic susceptibility in patients with and without mutations and show that patients without mutations are mostly from non-familial cases, have fewer reproductive losses, and more live births. Our data demonstrate that patients with recurrent hydatidiform moles and no mutations in the known genes are, in general, different from those with mutations; they have a milder genetic susceptibility and/or a multifactorial etiology underlying their recurrent hydatidiform moles. Categorizing these patients according to the genotypic types of their recurrent hydatidiform moles may facilitate the identification of novel genes for this entity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Neoplasias Primarias Secundarias/genética , Proteínas/genética , Neoplasias Uterinas/genética , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Embarazo
11.
Genes Chromosomes Cancer ; 56(12): 832-840, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28730668

RESUMEN

Miscarriages affect 15% of clinically recognized pregnancies. Recurrent miscarriage (RM) is defined by the occurrence of at least two consecutive pregnancy losses and affects 1%-5% of couples trying to conceive. In an attempt to categorize patients with RM and identify the mechanisms leading to their miscarriages, we first used flow cytometry to assess the ploidy of 93 products of conception (POCs) from 53 patients with RM (≥3 miscarriages). We identified a single patient with four triploid POCs. We then used fluorescent in situ hybridization to confirm the triploidies and fluorescent microsatellite genotyping with distal and pericentromeric markers to determine their parental origin and the mechanisms leading to their formation. We found that all four triploidies were digynic and due to a failure in meiosis II (MII), suggesting a genetic predisposition. Upon further investigation into the family, we found a remarkable history of ovarian cysts and dysfunctions on the maternal side. Notably, one maternal cousin had a mature ovarian teratoma that we analyzed and found an identical mechanism at its origin, a failure in MII. The identification of two patients in the same family with two different manifestations-digynic triploid conceptions and mature ovarian teratomas, both resulting from the failure of MII-suggests an inherited genetic susceptibility toward an error in MII segregating in the family that may manifest in the form of a triploid digynic miscarriage or a mature ovarian teratoma. Our findings may facilitate the future identification of causative mutations for MII defects.


Asunto(s)
Aborto Habitual/genética , Neoplasias Ováricas/genética , Teratoma/genética , Triploidía , Aborto Habitual/patología , Adulto , Centrómero/genética , Femenino , Humanos , Patrón de Herencia , Masculino , Meiosis/genética , Repeticiones de Microsatélite , Neoplasias Ováricas/patología , Linaje , Teratoma/patología
12.
Hum Reprod ; 30(1): 159-69, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25358348

RESUMEN

STUDY QUESTION: What is the subcellular localization in human oocytes and preimplantation embryos, of the two maternal-effect proteins, NLRP7 and KHDC3L, responsible for recurrent hydatidiform moles (RHMs)? SUMMARY ANSWER: NLRP7 and KHDC3L localize to the oocyte cytoskeleton and are polar and absent from the cell-to-cell contact region in early preimplantation embryos. WHAT IS KNOWN ALREADY: NLRP7 and KHDC3L expression has been described at the RNA level in some stages of human oocytes and preimplantation embryos and at the protein level by immunohistochemistry in human and bovine ovaries. NLRP7 and KHDC3L co-localize to the microtubule organizing center and/or the Golgi apparatus in human hematopoietic cells. STUDY DESIGN, SIZE, DURATION: A total of 164 spare human oocytes and embryos from patients undergoing in vitro fertilization were used. PARTICIPANTS/MATERIALS, SETTING, METHODS: Oocytes and early cleavage-stage embryos were fixed, immunostained with NLRP7 and/or KHDC3L antibodies, and analyzed using high-resolution confocal immunofluorescence and electron microscopies. MAIN RESULTS AND THE ROLE OF CHANCE: NLRP7 and KHDC3L localize to the cytoskeleton and are predominant at the cortical region in growing oocytes. After the first cellular division, these two maternal-effect proteins become asymmetrically confined to the outer cortical region and excluded from the cell-to-cell contact region until the blastocyst stage where NLRP7 and KHDC3L homogeneously redistribute to the cytoplasm and the nucleus, respectively. LIMITATIONS, REASONS FOR CAUTION: We could not analyze fresh human oocytes and embryos. The analyzed materials were donated by patients undergoing assisted reproductive technologies and released for research 1-3 days after their collection and the transfer of embryos to the patients. WIDER IMPLICATIONS OF THE FINDINGS: Our study is the first comprehensive and high-resolution localization of the only two known maternal-effect proteins, NLRP7 and KHDC3L, in human oocytes and preimplantation embryos. Our data contribute to a better understanding of the roles of these two proteins in the integrity of the oocytes, post-zygotic divisions, and cell-lineage differentiation. STUDY FUNDING/COMPETING INTERESTS: This work was supported by the Canadian Institute of Health Research (86546 to R.S.); E.A. was supported by fellowships from the Research Institute of the McGill University Health Centre and a CREATE award from the Réseau Québécois en Reproduction. All authors declare no conflict of interest.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/análisis , Blastocisto/metabolismo , Citoesqueleto/metabolismo , Mola Hidatiforme/genética , Oocitos/metabolismo , Proteínas/análisis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Polaridad Celular , Desarrollo Embrionario , Femenino , Humanos , Embarazo , Proteínas/metabolismo , Proteínas/fisiología
13.
Reprod Biomed Online ; 31(1): 120-4, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25982095

RESUMEN

Hydatidiform mole (HM) is an aberrant human pregnancy with abnormal embryonic development and excessive proliferation of the trophoblast. Recessive mutations in NLRP7 are responsible for recurrent HM (RHM). Women with recessive NLRP7 mutations fail to have normal pregnancies from spontaneous conceptions with the exception of three out of 131 reported patients. Because there is no treatment for RHM and maternal-effect genes are needed in the oocytes to sustain normal embryonic development until the activation of the embryonic genome, one patient with recessive NLRP7 mutations tried ovum donation and achieved a successful pregnancy. This study reports three additional live births from donated ova to two patients with recessive NLRP7 mutations. The occurrence of two live births from spontaneous conceptions to two other patients is also reported. The reproductive outcomes and mutations of all reported patients were reviewed and it was found that live births are associated with some missense mutations expected to have mild functional consequences on the protein. The data support a previous observation that ovum donation appears the best management option for these patients to achieve normal pregnancies and provide an explanation for the rare occurrence of live births from natural spontaneous conceptions in patients with two NLRP7 mutations.


Asunto(s)
Mola Hidatiforme/complicaciones , Infertilidad Femenina/genética , Nacimiento Vivo , Resultado del Embarazo , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Femenino , Fertilización In Vitro , Humanos , Mola Hidatiforme/genética , Infertilidad Femenina/terapia , Mutación , Mutación Missense , Donación de Oocito , Embarazo
14.
J Med Genet ; 51(9): 623-34, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25097207

RESUMEN

BACKGROUND: Hydatidiform mole (HM) is a human pregnancy with excessive trophoblastic proliferation and abnormal embryonic development that may be sporadic or recurrent. In the sporadic form, the HM phenotype is driven by an abnormal ratio of paternal to maternal genomes, whereas in the recurrent form, the HM phenotype is caused by maternal-recessive mutations, mostly in NLRP7, despite the diploid biparental origin of the HM tissues. In this study, we characterised the expression of the imprinted, maternally expressed gene, CDKN1C (p57(KIP2)), the genotype, and the histopathology of 36 products of conception (POC) from patients with two defective alleles in NLRP7 and looked for potential correlations between the nature of the mutations in the patients and the various HM features. METHODS/RESULTS: We found that all the 36 POCs are diploid biparental and have the same parental contribution to their genomes. However, some of them expressed variable levels of p57(KIP2) and this expression was strongly associated with the presence of embryonic tissues of inner cell mass origin and mild trophoblastic proliferation, which are features of triploid partial HMs, and were associated with missense mutations. Negative p57(KIP2) expression was associated with the absence of embryonic tissues and excessive trophoblastic proliferation, which are features of androgenetic complete HMs and were associated with protein-truncating mutations. CONCLUSIONS: Our data suggest that NLRP7, depending on the severity of its mutations, regulates the imprinted expression of p57(KIP2) and consequently the balance between tissue differentiation and proliferation during early human development. This role is novel and could not have been revealed by any other approach on somatic cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Mola Hidatiforme/genética , Trofoblastos/fisiología , Diferenciación Celular/genética , Proliferación Celular/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Análisis Mutacional de ADN , Femenino , Citometría de Flujo , Impresión Genómica/genética , Genotipo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Repeticiones de Microsatélite/genética , Mutación Missense/genética , Embarazo
15.
Nat Genet ; 38(3): 300-2, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16462743

RESUMEN

Hydatidiform mole (HM) is an abnormal human pregnancy with no embryo and cystic degeneration of placental villi. We report five mutations in the maternal gene NALP7 in individuals with familial and recurrent HMs. NALP7 is a member of the CATERPILLER protein family involved in inflammation and apoptosis. NALP7 is the first maternal effect gene identified in humans and is also responsible for recurrent spontaneous abortions, stillbirths and intrauterine growth retardation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Mola Hidatiforme/genética , Mutación , Reproducción/genética , Neoplasias Uterinas/genética , Etnicidad , Femenino , Humanos , Masculino , Linaje , Embarazo
16.
Genome Res ; 21(3): 465-76, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21324877

RESUMEN

Imprinted genes are critical for normal human growth and neurodevelopment. They are characterized by differentially methylated regions (DMRs) of DNA that confer parent of origin-specific transcription. We developed a new strategy to identify imprinted gene-associated DMRs. Using genome-wide methylation profiling of sodium bisulfite modified DNA from normal human tissues of biparental origin, candidate DMRs were identified by selecting CpGs with methylation levels consistent with putative allelic differential methylation. In parallel, the methylation profiles of tissues of uniparental origin, i.e., paternally-derived androgenetic complete hydatidiform moles (AnCHMs), and maternally-derived mature cystic ovarian teratoma (MCT), were examined and then used to identify CpGs with parent of origin-specific DNA methylation. With this approach, we found known DMRs associated with imprinted genomic regions as well as new DMRs for known imprinted genes, NAP1L5 and ZNF597, and novel candidate imprinted genes. The paternally methylated DMR for one candidate, AXL, a receptor tyrosine kinase, was also validated in experiments with mouse embryos that demonstrated Axl was expressed preferentially from the maternal allele in a DNA methylation-dependent manner.


Asunto(s)
ADN/genética , Impresión Genómica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Alelos , Animales , Secuencia de Bases , Islas de CpG/genética , ADN/química , Metilación de ADN , Embrión de Mamíferos , Femenino , Variación Genética , Genoma , Humanos , Mola Hidatiforme/genética , Mola Hidatiforme/metabolismo , Ratones , Análisis por Micromatrices/métodos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Embarazo , Complicaciones del Embarazo/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Factores Sexuales , Sulfitos/química , Teratoma/genética , Teratoma/metabolismo , Tirosina Quinasa del Receptor Axl
17.
Mol Hum Reprod ; 20(10): 990-1001, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25082979

RESUMEN

Mutations in NLRP7 (NOD-like-receptor family, pyrin domain containing 7) are responsible for a type of recurrent pregnancy loss known as recurrent hydatidiform mole (HYDM1). This condition is characterized by abnormal growth of the placenta, a lack of proper embryonic development and abnormal methylation patterns at multiple imprinted loci in diploid biparental molar tissues. The role of NLRP7 protein in the disease manifestation is currently not clear. In order to better understand how the effects of HYDM1 are associated with mutations on the structure of NLRP7, we performed an inter-domain interaction screen using a yeast two-hybrid system. Additionally, we generated in silico structural models of NLRP7 in its non-activated and activated forms. Our observations from the yeast two-hybrid screen and modeling suggest that the NACHT-associated domain (NAD) of the NLRP7 protein is central to its oligomeric assembly. Upon activation, the NAD and a small part of the leucine rich repeat (LRR) of one molecule emerged out of the protective LRR domain and interact with the NACHT domain of the second molecule to form an oligomer. Furthermore, we investigated the molecular basis for the pathophysiological effect of four missense mutations, three HYDM1-causing and one rare non-synonymous variant, on the protein using confocal microscopy of transiently transfected NLRP7 in HEK293T cells and in silico structural analysis. We found that with the two clinically severe missense mutations, L398R and R693W, the normal molecule to molecule interaction was apparently affected thus decreasing their oligomerization potential while aggresome formation was increased; these changes could disturb the normal downstream functions of NLRP7 and therefore be a possible molecular effect underlying their pathophysiological impact.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Línea Celular , Femenino , Células HEK293 , Humanos , Modelos Moleculares , Mutación Missense , NAD/genética , Placentación , Embarazo , Agregación Patológica de Proteínas/genética , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
18.
Mol Genet Genomic Med ; 12(2): e2402, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38400599

RESUMEN

BACKGROUND: Recurrent miscarriage (RM) affects 1% to 5% of couples trying to conceive. Despite extensive clinical and laboratory testing, half of the RM cases remain unexplained. We report the genetic analysis of a couple with eight miscarriages and the search for their potential genetic etiology. METHODS: Short tandem repeat (STR) markers, single nucleotide polymorphic (SNP) microarray, and human DNA methylation microarray were used to analyze the genotypes of two miscarriages. Exomes sequencing was performed on DNA from the two partners and identified variants were validated by Sanger sequencing. RESULTS: STR marker genotyping demonstrated that the two available miscarriages are triploid digynic and resulted from the failure of Meiosis II. SNP microarray analysis revealed an additional Meiosis I abnormality that is the segregation of the two maternal homologous chromosomes in one triploid miscarriage. Whole-exome sequencing on DNA from the two partners identified candidate variants only in the female partner in two genes with roles in female reproduction, a missense in EIF4ENIF1 (OMIM 607445) and a stop gain in HORMAD2 (OMIM 618842). EIF4ENIF1 is a eukaryotic translation initiation factor 4E nuclear import factor required for the oocyte germinal vesicle breakdown, and HORMAD2 is part of the synaptonemal complex that was hypothesized to act as a checkpoint mechanism to eliminate oocytes with asynapsis during meiotic prophase I in mice. CONCLUSION: While both genes may contribute to the phenotype, the Meiosis I abnormalities in the conceptions favor the causal role of HORMAD2 in the etiology of RM in this couple. This report illustrates the importance of comprehensively analyzing the products of conception to guide the search for the genetic causation of RM.


Asunto(s)
Aborto Habitual , Meiosis , Femenino , Humanos , Embarazo , Aborto Habitual/genética , Codón de Terminación , ADN , Meiosis/genética , Triploidía , Masculino
19.
Int J Gynecol Pathol ; 32(4): 399-405, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23722513

RESUMEN

Recurrent hydatidiform moles is an uncommon occurrence. Over the past decade, genetic studies of women with multiple recurrent molar pregnancies have revealed that maternal mutations in two different genes, NLRP7 and C6orf221, result in recurrent moles. We report a 23 year old woman, born of unrelated parents, who has experienced three molar pregnancies in succession. Whilst the first pregnancy was classified as a complete hydatidiform mole, the second and third moles defied classification as complete or partial mole using conventional histology, p57 nuclear staining pattern and ploidy studies. Molecular and cytogenetic studies proved that all three molar pregnancies were diploid and biparental in origin. Gene sequencing analysis showed that the patient is homozygous for a previously described mutation in NLRP7. A SNP microarray ruled out the presence of deletion of the NLRP7 locus. This case draws attention to the fact that recurrent molar pregnancies may be the result of specific, identifiable gene mutations, even in patients from non-consanguineous backgrounds. When pathologists encounter patients with molar pregnancies that are diploid and p57 negative and yet have fetal elements such as nucleated red blood cells or immature fetal tissues, it should heighten their suspicion of a possible genetic basis and appropriate molecular genetic workup performed with counseling offered.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores de Tumor/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Mola Hidatiforme/genética , Complicaciones del Embarazo , Neoplasias Uterinas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Alelos , Biomarcadores de Tumor/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Perfilación de la Expresión Génica , Técnicas de Genotipaje , Humanos , Mola Hidatiforme/clasificación , Mola Hidatiforme/patología , Hibridación Fluorescente in Situ , Mutación , Recurrencia Local de Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Ploidias , Embarazo , Análisis de Secuencia de ADN , Neoplasias Uterinas/clasificación , Neoplasias Uterinas/patología , Adulto Joven
20.
Cells ; 12(6)2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36980199

RESUMEN

BACKGROUND: Gestational choriocarcinoma (GC) is a highly malignant trophoblastic tumor that often develops from a complete hydatidiform mole (HM). NLRP7 is the major gene responsible for recurrent HM and is involved in the innate immune response, inflammation and apoptosis. NLRP7 can function in an inflammasome-dependent or -independent pathway. Recently, we have demonstrated that NLRP7 is highly expressed in GC tumor cells and contributes to their tumorigenesis. However, the underlying mechanisms are still unknown. Here, we investigated the mechanism by which NLRP7 controls these processes in malignant (JEG-3) and non-tumor (HTR8/SVneo) trophoblastic cells. Cell survival, dedifferentiation, camouflage, and aggressiveness were compared between normal JEG-3 cells or knockdown for NLRP7, JEG-3 Sh NLRP7. In addition, HTR8/SVneo cells overexpressing NLRP7 were used to determine the impact of NLRP7 overexpression on non-tumor cells. NLRP7 involvement in tumor cell growth and tolerance was further characterized in vivo using the metastatic mouse model of GC. RESULTS: We demonstrate that NLRP7 (i) functions in an inflammasome-dependent and -independent manners in HTR8/SVneo and JEG-3 cells, respectively; (ii) differentially regulates the activity of NF-κB in tumor and non-tumor cells; (iii) increases malignant cell survival, dedifferentiation, and camouflage; and (iv) facilitates tumor cells colonization of the lungs in the preclinical model of GC. CONCLUSIONS: This study demonstrates for the first time the mechanism by which NLRP7, independently of its inflammasome machinery, contributes to GC growth and tumorigenesis. The clinical relevance of NLRP7 in this rare cancer highlights its potential therapeutic promise as a molecular target to treat resistant GC patients.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Coriocarcinoma , Animales , Femenino , Humanos , Ratones , Embarazo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis , Línea Celular Tumoral , Supervivencia Celular , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Inflamasomas/metabolismo , Recurrencia Local de Neoplasia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA