Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Differentiation ; 138: 100791, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38941819

RESUMEN

A Wt1 conditional deletion, nuclear red fluorescent protein (RFP) reporter allele was generated in the mouse by gene targeting in embryonic stem cells. Upon Cre-mediated recombination, a deletion allele is generated that expresses RFP in a Wt1-specific pattern. RFP expression was detected in embryonic and adult tissues known to express Wt1, including the kidney, mesonephros, and testis. In addition, RFP expression and WT1 co-localization was detected in the adult uterine stroma and myometrium, suggesting a role in uterine function. Crosses with Wnt7a-Cre transgenic mice that express Cre in the Müllerian duct epithelium activate Wt1-directed RFP expression in the epithelium of the oviduct but not the stroma and myometrium of the uterus. This new mouse strain should be a useful resource for studies of Wt1 function and marking Wt1-expressing cells.


Asunto(s)
Alelos , Proteínas Luminiscentes , Ratones Transgénicos , Proteína Fluorescente Roja , Proteínas WT1 , Animales , Ratones , Proteínas WT1/genética , Proteínas WT1/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Femenino , Genes Reporteros , Masculino , Eliminación de Gen
2.
Development ; 144(1): 44-53, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27888191

RESUMEN

Supporting cells (Sertoli and granulosa) and steroidogenic cells (Leydig and theca-interstitium) are two major somatic cell types in mammalian gonads, but the mechanisms that control their differentiation during gonad development remain elusive. In this study, we found that deletion of Wt1 in the ovary after sex determination caused ectopic development of steroidogenic cells at the embryonic stage. Furthermore, differentiation of both Sertoli and granulosa cells was blocked when Wt1 was deleted before sex determination and most genital ridge somatic cells differentiated into steroidogenic cells in both male and female gonads. Further studies revealed that WT1 repressed Sf1 expression by directly binding to the Sf1 promoter region, and the repressive function was completely abolished when WT1 binding sites were mutated. This study demonstrates that Wt1 is required for the lineage specification of both Sertoli and granulosa cells by repressing Sf1 expression. Without Wt1, the expression of Sf1 was upregulated and the somatic cells differentiated into steroidogenic cells instead of supporting cells. Our study uncovers a novel mechanism of somatic cell differentiation during gonad development.


Asunto(s)
Linaje de la Célula/genética , Células de la Granulosa/fisiología , Factores de Empalme de ARN/genética , Proteínas Represoras/fisiología , Células de Sertoli/fisiología , Diferenciación Sexual/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Regulación hacia Abajo/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Embarazo , Células de Sertoli/metabolismo , Procesos de Determinación del Sexo/genética , Proteínas WT1
3.
Proc Natl Acad Sci U S A ; 112(13): 4003-8, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775596

RESUMEN

Sertoli and Leydig cells, the two major somatic cell types in the testis, have different morphologies and functions. Both are essential for gonad development and spermatogenesis. However, whether these cells are derived from the same progenitor cells and the mechanism regulating the differentiation between these two cell types during gonad development remains unclear. A previous study showed that overactivation of Ctnnb1 (cadherin-associated protein, beta 1) in Sertoli cells resulted in Sertoli cell tumors. Surprisingly, in the present study, we found that simultaneous deletion of Wilms' Tumor Gene 1 (Wt1) and overactivation of Ctnnb1 in Sertoli cells led to Leydig cell-like tumor development. Lineage tracing experiments revealed that the Leydig-like tumor cells were derived from Sertoli cells. Further studies confirmed that Wt1 is required for the maintenance of the Sertoli cell lineage and that deletion of Wt1 resulted in the reprogramming of Sertoli cells to Leydig cells. Consistent with this interpretation, overexpression of Wt1 in Leydig cells led to the up-regulation of Sertoli cell-specific gene expression and the down-regulation of steroidogenic gene expression. These results demonstrate that the distinction between Sertoli cells and Leydig cells is regulated by Wt1, implying that these two cell types most likely originate from the same progenitor cells. This study thus provides a novel concept for somatic cell fate determination in testis development that may also represent an etiology of male infertility in human patients.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Intersticiales del Testículo/citología , Células de Sertoli/citología , Testículo/crecimiento & desarrollo , Proteínas WT1/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Eliminación de Gen , Inmunohistoquímica , Masculino , Ratones Noqueados , Testículo/embriología , beta Catenina/genética
4.
J Med Genet ; 53(6): 385-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26566882

RESUMEN

Wilms tumour (WT), a paediatric renal cancer, is the most common childhood kidney cancer. The aetiology of WT is heterogeneous with multiple genes known to result in WT tumorigenesis. However, these genes are rarely associated with familial Wilms tumour (FWT). To identify mutations predisposing to FWT, we performed whole-genome sequencing using genomic DNA from three affected/obligate carriers in a large WT family, followed by Sanger sequencing of candidate gene mutations in 47 additional WT families to determine their frequency in FWT. As a result, we identified two novel germline DICER1 mutations (G803R and R800Xfs5) co-segregating in two families, thus expanding the number of reported WT families with unique germline DICER1 mutations. The one large family was found to include individuals with multiple DICER1 syndrome phenotypes, including four WT cases. Interestingly, carriers of the DICER1 mutation displayed a greatly increased frequency of WT development compared with the penetrance observed in previously published pedigrees. Also uniquely, in one tumour this DICER1 mutant allele (G803R) was reduced to homozygosity in contrast to the somatic hotspot mutations typically observed in tumours in DICER1 families.


Asunto(s)
ARN Helicasas DEAD-box/genética , Mutación de Línea Germinal/genética , Pérdida de Heterocigocidad/genética , Ribonucleasa III/genética , Tumor de Wilms/genética , Adolescente , Adulto , Preescolar , Femenino , Predisposición Genética a la Enfermedad/genética , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Linaje
5.
Hum Mol Genet ; 23(2): 333-41, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24009315

RESUMEN

The Wt1 gene encodes a nuclear transcription factor that is specifically expressed in ovarian granulosa cells. However, the physiological significance of Wt1 in ovarian follicle development remains elusive. In this study, we found that Wt1(+/R394W) mice were grossly normal, however, the females displayed severe reproductive defects. Only ∼15% of the Wt1(+/R394W) females became pregnant after mating with wild-type males, compared with 88.2% of control females. Further study revealed that the subfertility of Wt1(+/R394W) females was caused by aberrant ovarian follicle development. Compared with control females, the ovary size and the number of developing follicles was significantly decreased in Wt1 mutant ovaries which was very similar to premature ovarian failure (POF) in human patients. The results of in vitro studies demonstrated that the expression of follicle stimulating hormone receptor (FSHR), 3ß-hydroxysteroid dehydrogenase and Aromatase was inhibited by Wt1 in granulosa cells, and mutation of Wt1 resulted in the upregulation of these genes and in the premature differentiation of granulosa cells. We also found that Wt1 was likely involved in granulosa cell development via the regulation of E-cadherin and Par6b expression. Mutation in Wt1 caused defects in polarity establishment in granulosa cells, which also likely contributed to the observed aberrant follicle development. The results of this study provide new mechanisms for understanding the regulation of ovarian follicle development and potential pathological cause of POF in human patients.


Asunto(s)
Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Folículo Ovárico/fisiología , Proteínas WT1/fisiología , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aromatasa/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Polaridad Celular , Femenino , Fertilidad , Regulación de la Expresión Génica , Células de la Granulosa/patología , Humanos , Masculino , Ratones , Folículo Ovárico/patología , Ovulación , Embarazo , Insuficiencia Ovárica Primaria/patología , Receptores de HFE/metabolismo , Proteínas WT1/genética
6.
Proc Natl Acad Sci U S A ; 110(10): 3985-90, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23426633

RESUMEN

Next-generation sequencing is revolutionizing genomic analysis, but this analysis can be compromised by high rates of missing true variants. To develop a robust statistical method capable of identifying variants that would otherwise not be called, we conducted sequence data simulations and both whole-genome and targeted sequencing data analysis of 28 families. Our method (Family-Based Sequencing Program, FamSeq) integrates Mendelian transmission information and raw sequencing reads. Sequence analysis using FamSeq reduced the number of false negative variants by 14-33% as assessed by HapMap sample genotype confirmation. In a large family affected with Wilms tumor, 84% of variants uniquely identified by FamSeq were confirmed by Sanger sequencing. In children with early-onset neurodevelopmental disorders from 26 families, de novo variant calls in disease candidate genes were corrected by FamSeq as mendelian variants, and the number of uniquely identified variants in affected individuals increased proportionally as additional family members were included in the analysis. To gain insight into maximizing variant detection, we studied factors impacting actual improvements of family-based calling, including pedigree structure, allele frequency (common vs. rare variants), prior settings of minor allele frequency, sequence signal-to-noise ratio, and coverage depth (∼20× to >200×). These data will help guide the design, analysis, and interpretation of family-based sequencing studies to improve the ability to identify new disease-associated genes.


Asunto(s)
Variación Genética , Análisis de Secuencia de ADN/métodos , Teorema de Bayes , Familia , Femenino , Genoma Humano , Humanos , Neoplasias Renales/genética , Funciones de Verosimilitud , Masculino , Enfermedades Mitocondriales/genética , Modelos Genéticos , Enfermedades Neurodegenerativas/genética , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/estadística & datos numéricos , Programas Informáticos , Tumor de Wilms/genética
7.
PLoS Genet ; 9(8): e1003645, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935527

RESUMEN

Azoospermia is one of the major reproductive disorders which cause male infertility in humans; however, the etiology of this disease is largely unknown. In the present study, six missense mutations of WT1 gene were detected in 529 human patients with non-obstructive azoospermia (NOA), indicating a strong association between WT1 mutation and NOA. The Wilms tumor gene, Wt1, is specifically expressed in Sertoli cells (SCs) which support spermatogenesis. To examine the functions of this gene in spermatogenesis, Wt1 was deleted in adult testis using Wt1(flox) and Cre-ER(TM) mice strains. We found that inactivation of Wt1 resulted in massive germ cell death and only SCs were present in most of the seminiferous tubules which was very similar to NOA in humans. In investigating the potential mechanism for this, histological studies revealed that the blood-testis barrier (BTB) was disrupted in Wt1 deficient testes. In vitro studies demonstrated that Wt1 was essential for cell polarity maintenance in SCs. Further studies found that the expression of cell polarity associated genes (Par6b and E-cadherin) and Wnt signaling genes (Wnt4, Wnt11) were downregulated in Wt1 deficient SCs, and that the expression of Par6b and E-cadherin was regulated by Wnt4. Our findings suggest that Wt1 is important in spermatogenesis by regulating the polarity of SCs via Wnt signaling pathway and that WT1 mutation is one of the genetic causes of NOA in humans.


Asunto(s)
Azoospermia/genética , Infertilidad Masculina/patología , Espermatogénesis/genética , Proteínas WT1/genética , Animales , Azoospermia/patología , Polaridad Celular , Humanos , Infertilidad Masculina/genética , Masculino , Ratones , Células de Sertoli/metabolismo , Células de Sertoli/patología , Proteínas WT1/metabolismo , Proteínas Wnt/genética , Proteína Wnt4/genética
8.
Cancer Cell ; 11(2): 105-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17292822

RESUMEN

The study of the genetics of Wilms tumor has led to several highly unexpected and precedent-establishing discoveries. Ironically, however, the identification of "WT genes" has been painfully slow, and gene mutations have been identified in only approximately 25% of tumors. The discovery of an X chromosome gene, WTX, that is mutated somatically in approximately 30% of Wilms tumors is notable both for helping to explain the genetic etiology of a substantial proportion of tumors and also for underscoring the role that X chromosome genes can play in cancer genetics.


Asunto(s)
Cromosomas Humanos X/genética , Genes del Tumor de Wilms , Neoplasias Renales/genética , Proteínas Supresoras de Tumor/genética , Tumor de Wilms/genética , Proteínas Adaptadoras Transductoras de Señales , Humanos
9.
J Pathol ; 230(1): 39-47, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23288785

RESUMEN

A significant number of patients with germline mutations in the Wilms' tumour 1 (WT1) gene, a transcriptional factor essential for early renal and gonadal development, display cryptorchidism or non-scrotal testis position. We show here that WT1 is expressed during development in the mouse gubernacular ligament connecting the testis to the abdominal wall. Conditional inactivation of Wt1 in the gubernaculum (GU-WT1KO animals) resulted in abnormal differentiation of the gubernacula during development and, in about 40% of adult males, unilateral, always left-sided, cryptorchidism. At birth the right testis was positioned above the processus vaginalis and eventually moved into the developing scrotal pouch. In affected mutants the left testis was displaced from the normal position and the left processus vaginalis failed to form. The analysis of testicular descent at different stages of postnatal development suggests that unilateral cryptorchidism might be caused by asymmetry in the positions of the abdominal organs providing a higher degree of mobility for the left testis. Spermatogenesis in GU-WT1KO animals was blocked in cryptorchid testes located in a high pararenal position, but was maintained in testes located in a low abdominal position. Conditional inactivation of both Wt1 and androgen receptor (Ar) genes in the gubernaculum led to a bilateral asymmetrical cryptorchidism in all mutant males, with the left testis again located higher than the right one. The malformations induced by WT1 and AR deficiency in the gubernaculum and processus vaginalis, in combination with mechanical constraints on testis descent, determine the final position of the testes. In summary, our data indicate that WT1 is directly involved in gubernaculum differentiation. Taken together, the results of the study underline the complex nature of testicular descent, with an involvement in this process of several genetic factors and developmental events.


Asunto(s)
Criptorquidismo/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Testículo/anomalías , Testículo/fisiología , Proteínas WT1/genética , Animales , Animales Recién Nacidos , Femenino , Citometría de Flujo , Eliminación de Gen , Conducto Inguinal/crecimiento & desarrollo , Conducto Inguinal/fisiología , Riñón/crecimiento & desarrollo , Riñón/fisiología , Operón Lac , Masculino , Ratones , Ratones Noqueados , Receptores Androgénicos/genética , Testículo/crecimiento & desarrollo
10.
Exp Cell Res ; 319(5): 612-22, 2013 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-23291318

RESUMEN

Wilms tumor gene WT1 encodes a zinc finger-containing transcription factor which is required for renal development. Mutations in WT1 are observed in 20% of Wilms tumors (a pediatric kidney cancer), but the in vivo WT1 targets and associated molecular pathways involved in the etiology of Wilms tumor are still elusive. To identify WT1 targets we performed genome-wide comprehensive expression profiling using Affymetrix Gene Chip Mouse Genome 430 2.0 Arrays, comparing E13.5 mouse kidneys in which Wt1 had been somatically ablated with littermate controls. We identified Usp18 as the most differentially expressed gene in mutant kidney. Using tetracycline inducible cells we demonstrated a repressive effect of WT1 on USP18 expression. Conversely, knockdown of WT1 led to the upregulation of Usp18. Furthermore, direct binding of WT1 to the Usp18 promoter was demonstrated by ChIP assay. Overexpression of USP18 in murine and human cell lines resulted in cell proliferation. Additionally, Usp18 upregulation was observed in a mouse model of Wilms tumor. Taken together our data demonstrate that Usp18 is a transcriptional target of WT1 and suggest that increased expression of USP18 following WT1 loss contributes to Wilms tumorigenesis.


Asunto(s)
Endopeptidasas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , Proteínas WT1/fisiología , Tumor de Wilms/genética , Animales , Secuencia de Bases , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Proliferación Celular , Inmunoprecipitación de Cromatina , Endopeptidasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Hibridación in Situ , Riñón/embriología , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/patología , Luciferasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Activación Transcripcional , Transfección , Ubiquitina Tiolesterasa , Proteínas WT1/antagonistas & inhibidores , Tumor de Wilms/patología
11.
Res Sq ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38947076

RESUMEN

Background: The demand for genetic services has outpaced the availability of resources, challenging clinicians untrained in genetic integration into clinical decision-making. The UTHealth Adult Cardiovascular Genomics Certificate (CGC) program trains non-genetic healthcare professionals to recognize, assess, and refer patients with heritable cardiovascular diseases. This asynchronous online course includes 24 modules in three tiers of increasing complexity, using realistic clinical scenarios, interactive dialogues, quizzes, and tests to reinforce learning. We hypothesized that the CGC will increase genomic competencies in this underserved audience and encourage applying genomic concepts in clinical practice. Methods: Required course evaluations include pre- and post-assessments, knowledge checks in each module, and surveys for module-specific feedback. After 6 months, longitudinal feedback surveys gathered data on the long-term impact of the course on clinical practice and conducted focused interviews with learners. Results: The CGC was accredited in September 2022. Principal learners were nurses (24%), nurse practitioners (21%), physicians (16%), and physician assistants. Scores of 283 learners in paired pre- and post-assessments increased specific skills related to recognizing heritable diseases, understanding inheritance patterns, and interpreting genetic tests. Interviews highlighted the CGC's modular structure and linked resources as key strengths. Learners endorsed confidence to use genetic information in clinical practice, such as discussing genetic concepts and risks with patients and referring patients for genetic testing. Learners were highly likely to recommend the CGC to colleagues, citing its role in enhancing heritable disease awareness. Conclusions: The CGC program effectively empowers non-genetic clinicians to master genomic competencies, fostering collaboration to prevent deaths from heritable cardiovascular diseases, and potentially transforming healthcare education and clinical practice.

12.
Nat Rev Urol ; 21(3): 158-180, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37848532

RESUMEN

The modern study of Wilms tumour was prompted nearly 50 years ago, when Alfred Knudson proposed the 'two-hit' model of tumour development. Since then, the efforts of researchers worldwide have substantially expanded our knowledge of Wilms tumour biology, including major advances in genetics - from cloning the first Wilms tumour gene to high-throughput studies that have revealed the genetic landscape of this tumour. These discoveries improve understanding of the embryonal origin of Wilms tumour, familial occurrences and associated syndromic conditions. Many efforts have been made to find and clinically apply prognostic biomarkers to Wilms tumour, for which outcomes are generally favourable, but treatment of some affected individuals remains challenging. Challenges are also posed by the intratumoural heterogeneity of biomarkers. Furthermore, preclinical models of Wilms tumour, from cell lines to organoid cultures, have evolved. Despite these many achievements, much still remains to be discovered: further molecular understanding of relapse in Wilms tumour and of the multiple origins of bilateral Wilms tumour are two examples of areas under active investigation. International collaboration, especially when large tumour series are required to obtain robust data, will help to answer some of the remaining unresolved questions.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Humanos , Neoplasias Renales/terapia , Recurrencia Local de Neoplasia , Tumor de Wilms/terapia , Biomarcadores , Biología
13.
Pediatr Blood Cancer ; 60(6): 994-1000, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23255438

RESUMEN

Renal malignancies are among the most prevalent pediatric cancers. The most common is favorable histology Wilms tumor (FHWT), which has 5-year overall survival exceeding 90%. Other pediatric renal malignancies, including anaplastic Wilms tumor, clear cell sarcoma, malignant rhabdoid tumor, and renal cell carcinoma, have less favorable outcomes. Recent clinical trials have identified gain of chromosome 1q as a prognostic marker for FHWT. Upcoming studies will evaluate therapy adjustments based on this and other novel biomarkers. For high-risk renal tumors, new treatment regimens will incorporate biological therapies. A research blueprint, viewed from the perspective of the Children's Oncology Group, is presented.


Asunto(s)
Ensayos Clínicos como Asunto , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Renales/terapia , Quimioterapia Adyuvante , Niño , Humanos , Nefrectomía , Investigación
14.
J Pathol ; 228(1): 119-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22374738

RESUMEN

Infantile fibrosarcoma (IFS; also known as cellular congenital mesoblastic nephroma, CMN, when in the kidney) is a rare, undifferentiated tumour often characterized by the ETV6-NTRK3 fusion transcript. Our goal was to identify downstream pathways, diagnostic markers and potential therapeutic targets for IFS/CMN. Global gene expression, reverse-phase protein array and ETV6-NTRK3 fusion analyses were performed on 14 IFS/CMN and compared with 41 other paediatric renal tumours. These analyses confirm significant receptor tyrosine kinase (RTK) activation, with evidence of PI3-Akt, MAPK and SRC activation. In particular, GAB2 docking protein, STAT5-pTyr-694, STAT3-pSer-729 and YAP-pSer-127 were elevated, and TAZ-pSer-89 was decreased. This provides mRNA and proteomic evidence that GAB2, STAT activation and phosphorylation of the Hippo pathway transcription co-activators YAP and TAZ contribute to the RTK signal transduction in IFS/CMN. All IFS/CMN tumours displayed a distinctive gene expression pattern that may be diagnostically useful. Unexpectedly, abundant ETV6-NTRK3 transcript copies were present in only 7/14 IFS, with very low copy number in 3/14. An additional 4/14 were negative by RT-PCR and absence of ETV6-NTRK3 was confirmed by FISH for both ETV6 and NTRK3. Therefore, molecular mechanisms other than ETV6-NTRK3 fusion are responsible for the development of some IFS/CMNs and the absence of ETV6-NTRK3 fusion products should not exclude IFS/CMN as a diagnosis.


Asunto(s)
Neoplasias Renales/genética , Nefroma Mesoblástico/genética , Receptor trkC/metabolismo , Biomarcadores de Tumor/metabolismo , ADN de Neoplasias/análisis , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Hibridación Fluorescente in Situ , Neoplasias Renales/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nefroma Mesoblástico/metabolismo , Proteínas de Fusión Oncogénica/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor trkC/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína ETS de Variante de Translocación 6
15.
Res Sq ; 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36993649

RESUMEN

This study comprehensively evaluated the landscape of genetic and epigenetic events that predispose to synchronous bilateral Wilms tumor (BWT). We performed whole exome or whole genome sequencing, total-strand RNA-seq, and DNA methylation analysis using germline and/or tumor samples from 68 patients with BWT from St. Jude Children's Research Hospital and the Children's Oncology Group. We found that 25/61 (41%) of patients evaluated harbored pathogenic or likely pathogenic germline variants, with WT1 (14.8%), NYNRIN (6.6%), TRIM28 (5%) and the BRCA-related genes (5%) BRCA1, BRCA2, and PALB2 being most common. Germline WT1 variants were strongly associated with somatic paternal uniparental disomy encompassing the 11p15.5 and 11p13/WT1 loci and subsequent acquired pathogenic CTNNB1 variants. Somatic coding variants or genome-wide copy number alterations were almost never shared between paired synchronous BWT, suggesting that the acquisition of independent somatic variants leads to tumor formation in the context of germline or early embryonic, post-zygotic initiating events. In contrast, 11p15.5 status (loss of heterozygosity, loss or retention of imprinting) was shared among paired synchronous BWT in all but one case. The predominant molecular events for BWT predisposition include pathogenic germline variants or post-zygotic epigenetic hypermethylation at the 11p15.5 H19/ICR1 locus (loss of imprinting). This study demonstrates that post-zygotic somatic mosaicism for 11p15.5 hypermethylation/loss of imprinting is the single most common initiating molecular event predisposing to BWT. Evidence of somatic mosaicism for 11p15.5 loss of imprinting was detected in leukocytes of a cohort of BWT patients and long-term survivors, but not in unilateral Wilms tumor patients and long-term survivors or controls, further supporting the hypothesis that post-zygotic 11p15.5 alterations occurred in the mesoderm of patients who go on to develop BWT. Due to the preponderance of BWT patients with demonstrable germline or early embryonic tumor predisposition, BWT exhibits a unique biology when compared to unilateral Wilms tumor and therefore warrants continued refinement of its own treatment-relevant biomarkers which in turn may inform directed treatment strategies in the future.

16.
Nat Commun ; 14(1): 8006, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110397

RESUMEN

Developing synchronous bilateral Wilms tumor suggests an underlying (epi)genetic predisposition. Here, we evaluate this predisposition in 68 patients using whole exome or genome sequencing (n = 85 tumors from 61 patients with matched germline blood DNA), RNA-seq (n = 99 tumors), and DNA methylation analysis (n = 61 peripheral blood, n = 29 non-diseased kidney, n = 99 tumors). We determine the predominant events for bilateral Wilms tumor predisposition: 1)pre-zygotic germline genetic variants readily detectable in blood DNA [WT1 (14.8%), NYNRIN (6.6%), TRIM28 (5%), and BRCA-related genes (5%)] or 2)post-zygotic epigenetic hypermethylation at 11p15.5 H19/ICR1 that may require analysis of multiple tissue types for diagnosis. Of 99 total tumor specimens, 16 (16.1%) have 11p15.5 normal retention of imprinting, 25 (25.2%) have 11p15.5 copy neutral loss of heterozygosity, and 58 (58.6%) have 11p15.5 H19/ICR1 epigenetic hypermethylation (loss of imprinting). Here, we ascertain the epigenetic and genetic modes of bilateral Wilms tumor predisposition.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Niño , Humanos , Tumor de Wilms/genética , Tumor de Wilms/patología , Genotipo , Metilación de ADN/genética , ADN , Neoplasias Renales/genética , Neoplasias Renales/patología , Epigénesis Genética , Impresión Genómica
17.
Cell Rep Med ; 3(6): 100644, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35617957

RESUMEN

Over the last decade, sequencing of primary tumors has clarified the genetic underpinnings of Wilms tumor but has not affected therapy, outcome, or toxicity. We now sharpen our focus on relapse samples from the umbrella AREN03B2 study. We show that over 40% of relapse samples contain mutations in SIX1 or genes of the MYCN network, drivers of progenitor proliferation. Not previously seen in large studies of primary Wilms tumors, DIS3 and TERT are now identified as recurrently mutated. The analysis of primary-relapse tumor pairs suggests that 11p15 loss of heterozygosity (and other copy number changes) and mutations in WT1 and MLLT1 typically occur early, but mutations in SIX1, MYCN, and WTX are late developments in some individuals. Most strikingly, 75% of relapse samples had gain of 1q, providing strong conceptual support for studying circulating tumor DNA in clinical trials to better detect 1q gain earlier and monitor response.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Niño , Genes del Tumor de Wilms , Proteínas de Homeodominio/genética , Humanos , Neoplasias Renales/genética , Proteína Proto-Oncogénica N-Myc/genética , Recurrencia Local de Neoplasia/genética , Tumor de Wilms/genética
18.
Ann Surg ; 251(3): 555-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20142733

RESUMEN

OBJECTIVE: To determine the event-free survival (EFS) and overall survival (OS) of children with very low risk Wilms tumor (VLRWT) treated with surgery only. BACKGROUND: Previous studies suggested that postoperative chemotherapy had not improved the prognosis of children with VLRWT. A total of 77 children <24 months of age with small (<550 g) Stage I favorable histology Wilms tumors were treated with surgery only. This study was closed based on stopping rules to ensure that the 2-year EFS was > or =90%. METHODS: A total of 77 children were assessed for EFS and OS. Of these patients, 21 enrolled at the time of closure were recalled, treated with dactinomycin and vincristine (regimen EE4A), and censored for analysis thereafter. About 111 children subsequently treated with EE4A were available for comparison. RESULTS: Median follow-up of surviving patients was 8.2 years for surgery only (range, 1.9-11.8 years) and 5.2 years for the EE4A group (range, 1.6-8.9 years). The estimated 5-year EFS for surgery only was 84% (95% confidence interval [CI]: 73%, 91%); for the EE4A patients it was 97% (95% CI: 92%, 99%, P = 0.002). One death was observed in each treatment group. The estimated 5-year OS was 98% (95% CI: 87%, 99%) for surgery only and 99% (95% CI: 94%, 99%) for EE4A (P = 0.70). CONCLUSION: The surgery-only EFS was lower than anticipated but, coupled with a much higher than anticipated salvage rate of the chemotherapy naive patients whose disease recurred, led to an observed long-term OS equivalent to that seen with 2-drug chemotherapy. This approach to the treatment of patients with VLRWT eliminates the toxic side-effects of chemotherapy for a large majority of patients. A follow-up study is underway to confirm these findings.


Asunto(s)
Neoplasias Renales/cirugía , Tumor de Wilms/cirugía , Humanos , Lactante , Neoplasias Renales/mortalidad , Factores de Riesgo , Tasa de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Tumor de Wilms/mortalidad
19.
Pediatr Blood Cancer ; 53(7): 1349-51, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19653292

RESUMEN

Frasier syndrome is characterized by a 46 XY disorder of sex development, nephropathy, and increased risk for gonadoblastoma due to Wilms tumor 1(WT1) mutation in the donor splice site of intron-9, resulting in the splice form +KTS. Germ cell tumors and gonadoblastomas have been reported previously in Frasier syndrome. We present the clinical, radiological, and genetic (WT1 mutation analysis) of a 46 XY phenotypic female with Frasier syndrome with bilateral gonadoblastoma with dysgerminoma who developed pilocytic astrocytoma.


Asunto(s)
Astrocitoma/genética , Disgerminoma/genética , Síndrome de Frasier/genética , Genes del Tumor de Wilms , Gonadoblastoma/genética , Neoplasias Hipotalámicas/genética , Síndromes Neoplásicos Hereditarios/genética , Mutación Puntual , Sitios de Empalme de ARN/genética , Astrocitoma/complicaciones , Astrocitoma/patología , Astrocitoma/cirugía , Niño , Disgerminoma/patología , Disgerminoma/cirugía , Femenino , Disgenesia Gonadal 46 XY/genética , Gonadoblastoma/patología , Gonadoblastoma/cirugía , Hemianopsia/etiología , Humanos , Neoplasias Hipotalámicas/complicaciones , Neoplasias Hipotalámicas/patología , Neoplasias Hipotalámicas/cirugía , Masculino , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Múltiples/patología , Neoplasias Primarias Múltiples/cirugía , Fenotipo , Proteinuria/genética , Trastornos del Habla/etiología
20.
Genes Chromosomes Cancer ; 47(6): 461-70, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18311776

RESUMEN

Wilms tumor is genetically heterogeneous, and until recently only one Wilms tumor gene was known, WT1 at 11p13. However, WT1 is altered in only approximately 20% of Wilms tumors. Recently a novel gene, WTX at Xq11.1, was reported to be mutated in Wilms tumors. No overlap between tumors with mutations in WTX and WT1 was noted, suggesting that WT1 and WTX mutations could account for the genetic basis of roughly half of Wilms tumors. To assess the frequency of WTX mutations and their relationship to WT1 mutations in a larger (n = 125) panel of Wilms tumors which had been thoroughly assessed for mutations in WT1, we conducted a complete mutational analysis of WTX that included sequencing of the entire coding region and quantitative PCR to identify deletions of the WTX gene. Twenty-three (18.4%) tumors carried a total of 24 WTX mutations, a lower WTX mutation frequency than that previously observed. Surprisingly, we observed an equivalent frequency of WTX mutations in tumors with mutations in either or both WT1 and CTNNB1 (20.0%) and tumors with no mutation in either WT1 or CTNNB1 (17.5%). WTX has been reported to play a role in the WNT/beta-catenin signaling pathway, and, interestingly, WTX deletion/truncation mutations appeared to be rare in tumors carrying exon 3 mutations of CTNNB1, encoding beta-catenin. Our findings indicate that WT1 and WTX mutations occur with similar frequency, that they partially overlap in Wilms tumors, and that mutations in WT1, WTX, and CTNNB1 underlie the genetic basis of about one-third of Wilms tumors.


Asunto(s)
Genes del Tumor de Wilms , Neoplasias Renales/genética , Mutación , Proteínas Supresoras de Tumor/genética , Tumor de Wilms/genética , beta Catenina/genética , Proteínas Adaptadoras Transductoras de Señales , Niño , Preescolar , Análisis Mutacional de ADN , Exones/genética , Femenino , Humanos , Lactante , Neoplasias Renales/epidemiología , Masculino , Mutación Puntual , Transducción de Señal/genética , Tumor de Wilms/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA