Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genes Chromosomes Cancer ; 53(1): 67-77, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24249259

RESUMEN

WTX is a tumor suppressor gene expressed during embryonic development and inactivated in 20-30% of cases of Wilms tumor, the most common pediatric kidney cancer. WTX has been implicated in several cellular processes including Wnt signaling, WT1 transcription, NRF2 degradation, and p53 function. Given that WTX is widely expressed during embryonic development and has been recently shown to regulate mesenchymal precursor cells in several organs, we tested for the potential involvement of WTX in a panel of pediatric tumors and adult sarcomas. A total of 353 tumors were screened for WTX deletions by fluorescence in situ hybridization (FISH). Discrete somatic WTX deletions were identified in two cases, one hepatoblastoma and one embryonal rhabdomyosarcoma, and confirmed by array comparative genomic hybridization. Direct sequencing of the full WTX open reading frame in 24 hepatoblastomas and 21 embryonal rhabdomyosarcomas did not identify additional mutations in these tumor types. The presence of WTX mRNA was confirmed in hepatoblastomas and embryonal rhabdomyosarcomas without WTX deletions by RNA-in situ hybridization. Notably, tumors with evidence of WTX inactivation, Wilms tumor, hepatoblastoma and rhabdomyosarcoma, are primitive tumors that resemble undifferentiated precursor cells and are linked to overgrowth syndromes. These results indicate that WTX inactivation occurs in a wider variety of tumor types than previously appreciated and point to shared pathogenic mechanisms between a subset of pediatric malignancies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Rabdomiosarcoma Embrionario/genética , Proteínas Supresoras de Tumor/genética , Tumor de Wilms/genética , Adulto , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Eliminación de Secuencia
2.
PLoS One ; 8(3): e58555, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472205

RESUMEN

Mouse embryonic stem cells (ESCs) have the potential to differentiate into germ cells (GCs) in vivo and in vitro. Interestingly, XY ESCs can give rise to both male and female GCs in culture, irrespective of the genetic sex. Recent studies showed that ESC-derived primordial GCs contributed to functional gametogenesis in vivo; however, in vitro differentiation techniques have never succeeded in generating mature oocytes from ESCs due to cryptogenic growth arrest during the preantral follicle stages of development. To address this issue, a mouse ESC line, capable of producing follicle-like structures (FLSs) efficiently, was established to investigate their properties using conventional molecular biological methods. The results revealed that the ESC-derived FLSs were morphologically similar to ovarian primary-to-secondary follicles but never formed an antrum; instead, the FLSs eventually underwent abnormal development or cell death in culture, or formed teratomas when transplanted under the kidney capsule in mice. Gene expression analyses demonstrated that the FLSs lacked transcripts for genes essential to late folliculogenesis, including gonadotropin receptors and steroidogenic enzymes, whereas some other genes were overexpressed in FLSs compared to the adult ovary. The E-Cadherin protein, which is involved in cell-to-cell interactions, was also expressed ectopically. Remarkably, it was seen that oocyte-like cells in the FLSs exhibited androgenetic genomic imprinting, which is ordinarily indicative of male GCs. Although the FLSs did not express male GC marker genes, the DNA methyltransferase, Dnmt3L, was expressed at an abnormally high level. Furthermore, the expression of sex determination factors was ambiguous in FLSs as both male and female determinants were expressed weakly. These data suggest that the developmental dysfunction of the ESC-derived FLSs may be attributable to aberrant gene expression and genomic imprinting, possibly associated with uncertain sex determination in culture.


Asunto(s)
Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Células Germinativas/citología , Oocitos/citología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/metabolismo , Medios de Cultivo Condicionados/farmacología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Perfilación de la Expresión Génica , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Folículo Ovárico/metabolismo , Receptores de Gonadotropina/metabolismo , Factores Sexuales
3.
PLoS One ; 7(2): e30996, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355333

RESUMEN

Ovarian cancer is the fifth leading cause of cancer death in women. Ovarian cancers display a high degree of complex genetic alterations involving many oncogenes and tumor suppressor genes. Analysis of the association between genetic alterations and clinical endpoints such as survival will lead to improved patient management via genetic stratification of patients into clinically relevant subgroups. In this study, we aim to define subgroups of high-grade serous ovarian carcinomas that differ with respect to prognosis and overall survival. Genome-wide DNA copy number alterations (CNAs) were measured in 72 clinically annotated, high-grade serous tumors using high-resolution oligonucleotide arrays. Two clinically annotated, independent cohorts were used for validation. Unsupervised hierarchical clustering of copy number data derived from the 72 patient cohort resulted in two clusters with significant difference in progression free survival (PFS) and a marginal difference in overall survival (OS). GISTIC analysis of the two clusters identified altered regions unique to each cluster. Supervised clustering of two independent large cohorts of high-grade serous tumors using the classification scheme derived from the two initial clusters validated our results and identified 8 genomic regions that are distinctly different among the subgroups. These 8 regions map to 8p21.3, 8p23.2, 12p12.1, 17p11.2, 17p12, 19q12, 20q11.21 and 20q13.12; and harbor potential oncogenes and tumor suppressor genes that are likely to be involved in the pathogenesis of ovarian carcinoma. We have identified a set of genetic alterations that could be used for stratification of high-grade serous tumors into clinically relevant treatment subgroups.


Asunto(s)
Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/mortalidad , Variaciones en el Número de Copia de ADN/genética , Marcadores Genéticos/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Mapeo Cromosómico , Cistadenocarcinoma Seroso/terapia , Femenino , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/terapia , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia
4.
Neuro Oncol ; 14(2): 132-44, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22067563

RESUMEN

The clinicopathological heterogeneity of glioblastoma (GBM) and the various genetic and phenotypic subtypes in GBM stem cells (GSCs) are well described. However, the relationship between GSCs and the corresponding primary tumor from which they were isolated is poorly understood. We have established GSC-enriched neurosphere cultures from 15 newly diagnosed GBM specimens and examined the relationship between the histopathological and genomic features of GSC-derived orthotopic xenografts and those of the respective patient tumors. GSC-initiated xenografts recapitulate the distinctive cytological hallmarks and diverse histological variants associated with the corresponding patient GBM, including giant cell and gemistocytic GBM, and primitive neuroectodermal tumor (PNET)-like components. This indicates that GSCs generate tumors that preserve patient-specific disease phenotypes. The majority of GSC-derived intracerebral xenografts (11 of 15) demonstrated a highly invasive behavior crossing the midline, whereas the remainder formed discrete nodular and vascular masses. In some cases, GSC invasiveness correlated with preoperative MRI, but not with the status of PI3-kinase/Akt pathways or O(6)-methylguanine methyltransferase expression. Genome-wide screening by array comparative genomic hybridization and fluorescence in situ hybridization revealed that GSCs harbor unique genetic copy number aberrations. GSCs acquiring amplifications of the myc family genes represent only a minority of tumor cells within the original patient tumors. Thus, GSCs are a genetically distinct subpopulation of neoplastic cells within a GBM. These studies highlight the value of GSCs for preclinical modeling of clinically relevant, patient-specific GBM and, thus, pave the way for testing novel anti-GSC/GBM agents for personalized therapy.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Células Madre Neoplásicas/patología , Animales , Neoplasias Encefálicas/metabolismo , Diferenciación Celular/fisiología , Femenino , Genotipo , Glioblastoma/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Invasividad Neoplásica/patología , Células Madre Neoplásicas/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
5.
Cancer Cell ; 20(6): 810-7, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22137795

RESUMEN

Tumor heterogeneity has been implicated in tumor growth and progression as well as resistance to therapy. We present an example of genetic heterogeneity in human malignant brain tumors in which multiple closely related driver genes are amplified and activated simultaneously in adjacent intermingled cells. We have observed up to three different receptor tyrosine kinases (EGFR, MET, PDGFRA) amplified in single tumors in different cells in a mutually exclusive fashion. Each subpopulation was actively dividing, and the genetic changes resulted in protein production, and coexisting subpopulations shared common early genetic mutations indicating their derivation from a single precursor cell. The stable coexistence of different clones within the same tumor will have important clinical implications for tumor resistance to targeted therapies.


Asunto(s)
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Glioblastoma/genética , Mosaicismo , Proteínas Proto-Oncogénicas c-met/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Anciano , Secuencia de Bases , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Receptores ErbB/metabolismo , Femenino , Heterogeneidad Genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
PLoS One ; 6(12): e28585, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194861

RESUMEN

Mutations at codon 641 of EZH2 are recurrent in germinal center B cell lymphomas, and the most common variants lead to altered EZH2 enzymatic activity and enhanced tri-methylation of histone H3 at lysine 27, a repressive chromatin modification. As an initial step toward screening patients for cancer genotype-directed therapy, we developed a screening assay for EZH2 codon 641 mutations amenable for testing formalin-fixed clinical specimens, based on the sensitive SNaPshot single nucleotide extension technology. We detected EZH2 mutations in 12/55 (22%) follicular lymphomas (FL), 5/35 (14%) diffuse large B cell lymphomas with a germinal center immunophenotype (GCB-DLBCL), and 2/11 (18%) high grade B cell lymphomas with concurrent rearrangements of BCL2 and MYC. No EZH2 mutations were detected in cases of Burkitt lymphoma (0/23). EZH2 mutations were frequently associated with the presence of BCL2 rearrangement (BCL2-R) in both the FL (28% of BCL-R cases versus 0% of BCL2-WT cases, p<0.05) and GCB-DLBCL groups (33% of BCL2-R cases versus 4% of BCL2-WT cases, p<0.04), and across all lymphoma types excluding BL (27% of BCL2-R cases versus 3% of BCL2-WT cases, p<0.003). We confirmed gain-of-function activity for all previously reported EZH2 codon 641 mutation variants. Our findings suggest that EZH2 mutations constitute an additional genetic "hit" in many BCL2-rearranged germinal center B cell lymphomas. Our work may be helpful in the selection of lymphoma patients for future trials of pharmacologic agents targeting EZH2 and EZH2-regulated pathways.


Asunto(s)
Codón/genética , Proteínas de Unión al ADN/genética , Reordenamiento Génico/genética , Centro Germinal/metabolismo , Linfoma de Células B/genética , Mutación/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factores de Transcripción/genética , Animales , Western Blotting , Proteína Potenciadora del Homólogo Zeste 2 , Fibroblastos/metabolismo , Centro Germinal/patología , Humanos , Ratones , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Complejo Represivo Polycomb 2 , Polimorfismo de Nucleótido Simple/genética
7.
Pharmacol Res ; 64(3): 203-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21605676

RESUMEN

We previously characterized the l-Ser analog #290, H(tBut)-l-Ser-O-Methyl·HCl, as a novel inhibitor of osteoclastogenesis which functions in both mouse and human cells. Here, we assessed the activity of #290 in animal models of osteoporosis and rheumatoid arthritis. Treatment of animals with #290 both prevented bone loss and led to the recovery of lost bone in osteoporotic mice. When inflammatory arthritis was induced in SKG mice, #290 treatment suppressed arthritis scores and significantly prevented the destruction of calcaneous bones. Additionally, #290 reciprocally modulated the mammalian target of rapamycin (mTOR) pathway in osteoclasts and osteoblasts in vitro, suggesting a dual effect on bone homeostasis. Our results demonstrate that #290 is a potential novel therapeutic tool for the treatment and/or study of diseases associated with bone destruction.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Lípidos/química , Lípidos/uso terapéutico , Osteoporosis/tratamiento farmacológico , Serina/análogos & derivados , Células 3T3 , Animales , Artritis Reumatoide/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/patología , Huesos/citología , Huesos/efectos de los fármacos , Huesos/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoporosis/patología , Serina/química , Serina/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo
8.
Cancer Res ; 70(6): 2158-64, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20215515

RESUMEN

In a genome-wide screen of 684 cancer cell lines, we identified homozygous intragenic microdeletions involving genes encoding components of the apical-basal cell polarity complexes. Among these, PARD3 is disrupted in cell lines and primary tumors from squamous carcinomas and glioblastomas. Reconstituting PARD3 expression in both cell types restores tight junctions and retards contact-dependent proliferation. Searching specifically for small intragenic microdeletions using high-resolution genomic arrays may be complementary to other genomic deletion screens and resequencing efforts in identifying new tumor suppressor genes.


Asunto(s)
Eliminación de Gen , Neoplasias/genética , Proteínas Adaptadoras Transductoras de Señales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Genoma Humano , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Hibridación Fluorescente in Situ , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Neoplasias/metabolismo , Neoplasias/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
9.
Cancer Res ; 69(21): 8231-5, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19843854

RESUMEN

Uterine leiomyosarcoma (ULMS) is a rare gynecologic malignancy with a low survival rate. Currently, there is no effective treatment for ULMS. Infrequent occurrences of human ULMS hamper the understanding of the initiation and progression of the disease, thereby limiting the ability to develop efficient therapies. To elucidate the roles of the p53 and BRCA1 tumor suppressor genes in gynecologic malignancies, we generated mice in which p53 and/or BRCA1 can be conditionally deleted using anti-Müllerian hormone type II receptor (Amhr2)-driven Cre recombinase. We showed that conditional deletion of p53 in mice results in the development of uterine tumors that resemble human ULMS and that concurrent deletion of p53 and BRCA1 significantly accelerates the progression of these tumors. This finding led to our hypothesis that BRCA1 may play a role in human ULMS development. Consistent with this hypothesis, we showed that the BRCA1 protein is absent in 29% of human ULMS and that BRCA1 promoter methylation is the likely mechanism of BRCA1 downregulation. These data indicate that the loss of BRCA1 function may be an important step in the progression of ULMS. Our findings provide a rationale for investigating therapies that target BRCA1 deficiency in ULMS.


Asunto(s)
Proteína BRCA1/fisiología , Leiomiosarcoma/metabolismo , Neoplasias Uterinas/metabolismo , Animales , Hormona Antimülleriana/genética , Metilación de ADN , Regulación hacia Abajo , Femenino , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Leiomiosarcoma/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Proteína p53 Supresora de Tumor/fisiología , Neoplasias Uterinas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...