Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
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
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
5.
Cancers (Basel) ; 13(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203890

RESUMEN

The inflammatory gene NLRP7 is the major gene responsible for recurrent complete hydatidiform moles (CHM), an abnormal pregnancy that can develop into gestational choriocarcinoma (CC). However, the role of NLRP7 in the development and immune tolerance of CC has not been investigated. Three approaches were employed to define the role of NLRP7 in CC development: (i) a clinical study that analyzed human placenta and sera collected from women with normal pregnancies, CHM or CC; (ii) an in vitro study that investigated the impact of NLRP7 knockdown on tumor growth and organization; and (iii) an in vivo study that used two CC mouse models, including an orthotopic model. NLRP7 and circulating inflammatory cytokines were upregulated in tumor cells and in CHM and CC. In tumor cells, NLRP7 functions in an inflammasome-independent manner and promoted their proliferation and 3D organization. Gravid mice placentas injected with CC cells invalidated for NLRP7, exhibited higher maternal immune response, developed smaller tumors, and displayed less metastases. Our data characterized the critical role of NLRP7 in CC and provided evidence of its contribution to the development of an immunosuppressive maternal microenvironment that not only downregulates the maternal immune response but also fosters the growth and progression of CC.

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
7.
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
8.
Virchows Arch ; 477(2): 309-315, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32055942

RESUMEN

NLRP7 is a maternal-effect gene that has a primary role in the oocyte. Its biallelic mutations are a major cause for recurrent diploid biparental hydatidiform moles (HMs). Here, we describe the full characterization of four HMs from a patient with a novel homozygous protein-truncating mutation in NLRP7. We found that some HMs have features of both complete and partial moles. Two HMs expressed p57 in the cytotrophoblast and stromal cells and exhibited divergent and discordant immunostaining. Microsatellite DNA-genotyping demonstrated that two HMs are diploid biparental and one is triploid digynic due to the failure of meiosis II. FISH analysis demonstrated triploidy in the cytotrophoblast and stromal cells in all villi. Our data highlight the atypical features of HM from patients with recessive NLRP7 mutations and the important relationship between NLRP7 defects in the oocyte and p57 expression that appear to be the main contributor to the molar phenotype regardless of the zygote genotype.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Mola Hidatiforme/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Neoplasias Uterinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Femenino , Genotipo , Enfermedad Trofoblástica Gestacional , Humanos , Mola Hidatiforme/genética , Mutación/genética , Recurrencia Local de Neoplasia/genética , Nevo Pigmentado/genética , Fenotipo , Embarazo
10.
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
11.
J Vis Exp ; (152)2019 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-31680667

RESUMEN

Hydatidiform mole (HM) is an abnormal human pregnancy characterized by excessive trophoblastic proliferation and abnormal embryonic development. There are two types of HM based on microscopic morphological evaluation, complete HM (CHM) and partial HM (PHM). These can be further subdivided based on the parental contribution to the molar genomes. Such characterization of HM, by morphology and genotype analyses, is crucial for patient management and for the fundamental understanding of this intriguing pathology. It is well documented that morphological analysis of HM is subject to wide interobserver variability and is not sufficient on its own to accurately classify HM into CHM and PHM and distinguish them from hydropic non-molar abortions. Genotyping analysis is mostly performed on DNA and tissues from formalin-fixed paraffin-embedded (FFPE) products of conception, which have less than optimal quality and may consequently lead to wrong conclusions. In this article, detailed protocols for multiplex genotyping and flow cytometry analyses of FFPE molar tissues are provided, along with the interpretation of the results of these methods, their troubleshooting, and integration with the morphological evaluation, p57KIP2 immunohistochemistry, and fluorescence in situ hybridization (FISH) to reach a correct and robust diagnosis. Here, the authors share the methods and lessons learned in the past 10 years from the analysis of approximately 400 products of conception.


Asunto(s)
Citometría de Flujo/métodos , Técnicas de Genotipaje/métodos , Mola Hidatiforme/genética , Repeticiones de Microsatélite/genética , Ploidias , Neoplasias Uterinas/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/análisis , Femenino , Humanos , Mola Hidatiforme/patología , Adhesión en Parafina , Embarazo , Neoplasias Uterinas/patología
12.
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
13.
Placenta ; 65: 29-36, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29908639

RESUMEN

INTRODUCTION: Hydatidiform mole (HM) is an aberrant human pregnancy with abnormal trophoblastic development, migration/invasion of the extravillous trophoblast in the decidua. These abnormalities are established in a hypoxic environment during the first trimester of gestation. METHODS: Using text mining, we identified 72 unique genes that are linked to HM (HM-linked genes) that we studied by bioinformatic analysis in publicly available transcriptomes of primary chorionic villous cells (cytotrophoblast, syncytiotrophoblast, extravillous trophoblast, and arterial and venous endothelial) isolated from normal placentas or established trophoblastic cell lines cultured under different oxygen concentrations. RESULTS: We show that the majority of HM-linked genes (75%) are involved in normal trophoblastic differentiation, arranged in clusters, and some of them are implicated in chorionic villous invasion or regulated by oxygen concentrations. DISCUSSION: Our analysis integrates the various aspects of the pathophysiology of HM and highlights the importance of trophoblastic differentiation in this pathology.


Asunto(s)
Diferenciación Celular/fisiología , Mola Hidatiforme/genética , Transcriptoma , Trofoblastos/fisiología , Neoplasias Uterinas/genética , Diferenciación Celular/genética , Biología Computacional , Femenino , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Humanos , Mola Hidatiforme/patología , Análisis por Micromatrices , Placenta/metabolismo , Placenta/patología , Embarazo , Trofoblastos/patología , Neoplasias Uterinas/patología , Estudios de Validación como Asunto
14.
Eur J Hum Genet ; 26(7): 1007-1013, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29693651

RESUMEN

Recurrent hydatidiform moles (RHM) are aberrant human pregnancies characterized by absence of, or abnormal, embryonic development, hydropic degeneration of chorionic villi, and hyperproliferation of the trophoblast. Biallelic mutations in two maternal-effect genes, NLRP7 and KHDC3L, underlie the causation of RHM in 60% of patients. We performed exome sequencing on a patient with six pregnancy losses, two miscarriages and four HM, and found no variants that affect the functions of the known genes. We found biallelic missense variants that affect conserved amino acids in PADI6 and segregate with the disease phenotype in the family. PADI6 is another maternal-effect gene and a member of the subcortical maternal complex that has been shown to have recessive variants that affect the gene function in four unrelated women with infertility who also experienced early embryonic arrest during preimplantation development after IVF. We demonstrated that PADI6 co-localizes with NLRP7 in human oocytes and preimplantation embryos and reviewed the morphology and genotypes of four products of conception from our patient. Our data expand the involvement of PADI6 to other forms of reproductive loss and highlight the commonality between infertility, miscarriages, and molar pregnancies, in some cases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mola Hidatiforme/genética , Infertilidad/genética , Desiminasas de la Arginina Proteica/genética , Proteínas/genética , Aborto Espontáneo/genética , Aborto Espontáneo/fisiopatología , Adulto , Vellosidades Coriónicas/patología , Femenino , Genotipo , Humanos , Mola Hidatiforme/fisiopatología , Infertilidad/fisiopatología , Mutación , Oocitos/patología , Embarazo , Arginina Deiminasa Proteína-Tipo 6 , Secuenciación del Exoma
15.
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
16.
Clin Cancer Res ; 23(22): 7130-7140, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28899975

RESUMEN

Purpose: Choriocarcinoma (CC) is the most malignant gestational trophoblastic disease that often develops from complete hydatidiform moles (CHM). Neither the mechanism of CC development nor its progression is yet characterized. We recently identified endocrine gland-derived vascular endothelial growth factor (EG-VEGF) as a novel key placental growth factor that controls trophoblast proliferation and invasion. EG-VEGF acts via two receptors, PROKR1 and PROKR2. Here, we demonstrate that EG-VEGF receptors can be targeted for CC therapy.Experimental Design: Three approaches were used: (i) a clinical investigation comparing circulating EG-VEGF in control (n = 20) and in distinctive CHM (n = 38) and CC (n = 9) cohorts, (ii) an in vitro study investigating EG-VEGF effects on the CC cell line JEG3, and (iii) an in vivo study including the development of a novel CC mouse model, through a direct injection of JEG3-luciferase into the placenta of gravid SCID-mice.Results: Both placental and circulating EG-VEGF levels were increased in CHM and CC (×5) patients. EG-VEGF increased JEG3 proliferation, migration, and invasion in two-dimensional (2D) and three-dimensional (3D) culture systems. JEG3 injection in the placenta caused CC development with large metastases compared with their injection into the uterine horn. Treatment of the animal model with EG-VEGF receptor's antagonists significantly reduced tumor development and progression and preserved pregnancy. Antibody-array and immunohistological analyses further deciphered the mechanism of the antagonist's actions.Conclusions: Our work describes a novel preclinical animal model of CC and presents evidence that EG-VEGF receptors can be targeted for CC therapy. This may provide safe and less toxic therapeutic options compared with the currently used multi-agent chemotherapies. Clin Cancer Res; 23(22); 7130-40. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/antagonistas & inhibidores , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Coriocarcinoma/tratamiento farmacológico , Coriocarcinoma/mortalidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Genes Reporteros , Humanos , Ratones , Terapia Molecular Dirigida , Pronóstico , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Transducción de Señal , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/sangre , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
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
18.
Clin Exp Reprod Med ; 44(1): 40-46, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28428943

RESUMEN

OBJECTIVE: To describe in vitro development of human embryos derived from an individual with a homozygous pathogenic variant in NLRP7 (19q13.42) and recurrent hydatidiform mole (HM), an autosomal recessive condition thought to occur secondary to an oocyte defect. METHODS: A patient with five consecutive HM pregnancies was genomically evaluated via next generation sequencing followed by controlled ovarian hyperstimulation, in vitro fertilization (IVF) with intracytoplasmic sperm injection, embryo culture, and preimplantation genetic screening. Findings in NLRP7 were recorded and embryo culture and biopsy data were tabulated as a function of parental origin for any identified ploidy error. RESULTS: The patient was found to have a pathogenic variant in NLRP7 (c.2810+2T>G) in a homozygous state. Fifteen oocytes were retrieved and 10 embryos were available after fertilization via intracytoplasmic sperm injection. Developmental arrest was noted for all 10 embryos after 144 hours in culture, thus no transfer was possible. These non-viable embryos were evaluated by karyomapping and all were diploid biparental; two were euploid and eight had various aneuploidies all of maternal origin. CONCLUSION: This is the first report of early human embryo development from a patient with any NLRP7 mutation. The pathogenic variant identified here resulted in global developmental arrest at or before blastocyst stage. Standard IVF should therefore be discouraged for such patients, who instead need to consider oocyte (or embryo) donation with IVF as preferred clinical methods to treat infertility.

19.
Hum Genome Var ; 3: 16027, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27621838

RESUMEN

Recurrent hydatidiform mole (RHM) is defined by the occurrence of repeated molar pregnancies in affected women. Two genes, NLRP7 and KHDC3L, play a causal role in RHM and are responsible for 48-80% and 5% of cases, respectively. Here, we report the results of screening these two genes for mutations in one Iranian and one Indian patient with RHM. No mutations in NLRP7 were identified in the two patients. KHDC3L sequencing identified two novel protein-truncating mutations in a homozygous state, a 4-bp deletion, c.17_20delGGTT (p.Arg6Leufs*7), in the Iranian patient and a splice mutation, c.349+1G>A, that affects the invariant donor site at the junction of exon 2 and intron 2 in the Indian patient. To date, only four mutations in KHDC3L have been reported. The identification of two additional mutations provides further evidence for the important role of KHDC3L in the pathophysiology of RHM and increases the diversity of mutations described in Asian populations.

20.
Eur J Hum Genet ; 24(10): 1445-52, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26956250

RESUMEN

NLRP7 is a major gene responsible for recurrent hydatidiform moles. Here, we report 11 novel NLRP7 protein truncating variants, of which five deletions of more than 1-kb. We analyzed the transcriptional consequences of four variants. We demonstrate that one large homozygous deletion removes NLRP7 transcription start site and results in the complete absence of its transcripts in a patient in good health besides her reproductive problem. This observation strengthens existing data on the requirement of NLRP7 only for female reproduction. We show that two other variants affecting the splice acceptor of exon 6 lead to its in-frame skipping while another variant affecting the splice donor site of exon 9 leads to an in-frame insertion of 54 amino acids. Our characterization of the deletion breakpoints demonstrated that most of the breakpoints occurred within Alu repeats and the deletions were most likely mediated by microhomology events. Our data define a hotspot of Alu instability and deletions in intron 5 with six different breakpoints and rearrangements. Analysis of NLRP7 genomic sequences for repetitive elements demonstrated that Alu repeats represent 48% of its intronic sequences and these repeats seem to have been inserted into the common NLRP2/7 primate ancestor before its duplication into two genes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Elementos Alu/genética , Eliminación de Gen , Mola Hidatiforme/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Puntos de Rotura del Cromosoma , Evolución Molecular , Exones , Femenino , Humanos , Mola Hidatiforme/diagnóstico , Mutagénesis Insercional , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA