Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Stem Cell Reports ; 6(5): 772-783, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27132888

RESUMO

Human male germ cell tumors (GCTs) are derived from primordial germ cells (PGCs). The master pluripotency regulator and neuroectodermal lineage effector transcription factor SOX2 is repressed in PGCs and the seminoma (SEM) subset of GCTs. The mechanism of SOX2 repression and its significance to GC and GCT development currently are not understood. Here, we show that SOX2 repression in SEM-derived TCam-2 cells is mediated by the Polycomb repressive complex (PcG) and the repressive H3K27me3 chromatin mark that are enriched at its promoter. Furthermore, SOX2 repression in TCam-2 cells can be abrogated by recruitment of the constitutively expressed H3K27 demethylase UTX to the SOX2 promoter through retinoid signaling, leading to expression of neuronal and other lineage genes. SOX17 has been shown to initiate human PGC specification, with its target PRDM1 suppressing mesendodermal genes. Our results are consistent with a role for SOX2 repression in normal germline development by suppressing neuroectodermal genes.


Assuntos
Neoplasias Embrionárias de Células Germinativas/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXF/genética , Seminoma/genética , Neoplasias Testiculares/genética , Linhagem da Célula/genética , Cromatina/genética , Regulação Neoplásica da Expressão Gênica , Células Germinativas/patologia , Histona Desmetilases/genética , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/patologia , Proteínas Nucleares/genética , Proteínas do Grupo Polycomb/genética , Regiões Promotoras Genéticas , Seminoma/patologia , Neoplasias Testiculares/patologia
2.
Leuk Lymphoma ; 57(4): 899-908, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26294112

RESUMO

Genomic copy number alterations (CNAs) in diffuse large B-cell lymphoma (DLBCL) have roles in disease pathogenesis, but overall clinical relevance remains unclear. Herein, an unbiased algorithm was uniformly applied across three genome profiling datasets comprising 392 newly-diagnosed DLBCL specimens that defined 32 overlapping CNAs, involving 36 minimal common regions (MCRs). Scoring criteria were established for 50 aberrations within the MCRs while considering peak gains/losses. Application of these criteria to independent datasets revealed novel candidate genes with coordinated expression, such as CNOT2, potentially with pathogenic roles. No one single aberration significantly associated with patient outcome across datasets, but genomic complexity, defined by imbalance in more than one MCR, significantly portended adverse outcome in two of three independent datasets. Thus, the standardized scoring of CNAs currently developed can be uniformly applied across platforms, affording robust validation of genomic imbalance and complexity in DLBCL and overall clinical utility as biomarkers of patient outcome.


Assuntos
Variação Genética , Genômica , Linfoma Difuso de Grandes Células B/genética , Locos de Características Quantitativas , Anticorpos Monoclonais Murinos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Aberrações Cromossômicas , Hibridização Genômica Comparativa , Ciclofosfamida/uso terapêutico , Variações do Número de Cópias de DNA , Doxorrubicina/uso terapêutico , Feminino , Regulação Neoplásica da Expressão Gênica , Testes Genéticos , Genômica/métodos , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/mortalidade , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo de Nucleotídeo Único , Prednisona/uso terapêutico , Prognóstico , Rituximab , Transdução de Sinais , Transcriptoma , Vincristina/uso terapêutico
3.
PLoS One ; 10(12): e0142846, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26624623

RESUMO

Germ Cell Tumors (GCT) have a high cure rate, but we currently lack the ability to accurately identify the small subset of patients who will die from their disease. We used a combined genomic and expression profiling approach to identify genomic regions and underlying genes that are predictive of outcome in GCT patients. We performed array-based comparative genomic hybridization (CGH) on 53 non-seminomatous GCTs (NSGCTs) treated with cisplatin based chemotherapy and defined altered genomic regions using Circular Binary Segmentation. We identified 14 regions associated with two year disease-free survival (2yDFS) and 16 regions associated with five year disease-specific survival (5yDSS). From corresponding expression data, we identified 101 probe sets that showed significant changes in expression. We built several models based on these differentially expressed genes, then tested them in an independent validation set of 54 NSGCTs. These predictive models correctly classified outcome in 64-79.6% of patients in the validation set, depending on the endpoint utilized. Survival analysis demonstrated a significant separation of patients with good versus poor predicted outcome when using a combined gene set model. Multivariate analysis using clinical risk classification with the combined gene model indicated that they were independent prognostic markers. This novel set of predictive genes from altered genomic regions is almost entirely independent of our previously identified set of predictive genes for patients with NSGCTs. These genes may aid in the identification of the small subset of patients who are at high risk of poor outcome.


Assuntos
Perfilação da Expressão Gênica , Genômica , Modelos Estatísticos , Neoplasias Embrionárias de Células Germinativas/diagnóstico , Neoplasias Embrionárias de Células Germinativas/genética , Hibridização Genômica Comparativa , Variações do Número de Cópias de DNA , Intervalo Livre de Doença , Humanos
4.
Stem Cells ; 33(2): 367-77, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25336442

RESUMO

The predominant view of pluripotency regulation proposes a stable ground state with coordinated expression of key transcription factors (TFs) that prohibit differentiation. Another perspective suggests a more complexly regulated state involving competition between multiple lineage-specifying TFs that define pluripotency. These contrasting views were developed from extensive analyses of TFs in pluripotent cells in vitro. An experimentally validated, genome-wide repertoire of the regulatory interactions that control pluripotency within the in vivo cellular contexts is yet to be developed. To address this limitation, we assembled a TF interactome of adult human male germ cell tumors (GCTs) using the Algorithm for the Accurate Reconstruction of Cellular Pathways (ARACNe) to analyze gene expression profiles of 141 tumors comprising pluripotent and differentiated subsets. The network (GCT(Net)) comprised 1,305 TFs, and its ingenuity pathway analysis identified pluripotency and embryonal development as the top functional pathways. We experimentally validated GCT(Net) by functional (silencing) and biochemical (ChIP-seq) analysis of the core pluripotency regulatory TFs POU5F1, NANOG, and SOX2 in relation to their targets predicted by ARACNe. To define the extent of the in vivo pluripotency network in this system, we ranked all TFs in the GCT(Net) according to sharing of ARACNe-predicted targets with those of POU5F1 and NANOG using an odds-ratio analysis method. To validate this network, we silenced the top 10 TFs in the network in H9 embryonic stem cells. Silencing of each led to downregulation of pluripotency and induction of lineage; 7 of the 10 TFs were identified as pluripotency regulators for the first time.


Assuntos
Algoritmos , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neoplasias Embrionárias de Células Germinativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Linhagem Celular Tumoral , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco Pluripotentes/patologia , Fatores de Transcrição/genética
5.
J Urol ; 193(5): 1479-85, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25498568

RESUMO

PURPOSE: Accurate discrimination of benign oncocytoma and malignant renal cell carcinoma is useful for planning appropriate treatment strategies for patients with renal masses. Classification of renal neoplasms solely based on histopathology can be challenging, especially the distinction between chromophobe renal cell carcinoma and oncocytoma. In this study we develop and validate an algorithm based on genomic alterations for the classification of common renal neoplasms. MATERIALS AND METHODS: Using TCGA renal cell carcinoma copy number profiles and the published literature, a classification algorithm was developed and scoring criteria were established for the presence of each genomic marker. As validation, 191 surgically resected formalin fixed paraffin embedded renal neoplasms were blindly submitted to targeted array comparative genomic hybridization and classified according to the algorithm. CCND1 rearrangement was assessed by fluorescence in situ hybridization. RESULTS: The optimal classification algorithm comprised 15 genomic markers, and involved loss of VHL, 3p21 and 8p, and chromosomes 1, 2, 6, 10 and 17, and gain of 5qter, 16p, 17q and 20q, and chromosomes 3, 7 and 12. On histological rereview (leading to the exclusion of 3 specimens) and using histology as the gold standard, 58 of 62 (93%) clear cell, 51 of 56 (91%) papillary and 33 of 34 (97%) chromophobe renal cell carcinomas were classified correctly. Of the 36 oncocytoma specimens 33 were classified as oncocytoma (17 by array comparative genomic hybridization and 10 by array comparative genomic hybridization plus fluorescence in situ hybridization) or benign (6). Overall 93% diagnostic sensitivity and 97% specificity were achieved. CONCLUSIONS: In a clinical diagnostic setting the implementation of genome based molecular classification could serve as an ancillary assay to assist in the histological classification of common renal neoplasms.


Assuntos
Adenoma Oxífilo/classificação , Adenoma Oxífilo/genética , Algoritmos , Carcinoma de Células Renais/classificação , Carcinoma de Células Renais/genética , Genômica , Córtex Renal , Neoplasias Renais/classificação , Neoplasias Renais/genética , Hibridização Genômica Comparativa , Humanos , Hibridização in Situ Fluorescente
6.
J Exp Med ; 211(7): 1379-91, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-24913233

RESUMO

Loss of cell cycle controls is a hallmark of cancer and has a well-established role in aggressive B cell malignancies. However, the role of such lesions in indolent follicular lymphoma (FL) is unclear and individual lesions have been observed with low frequency. By analyzing genomic data from two large cohorts of indolent FLs, we identify a pattern of mutually exclusive (P = 0.003) genomic lesions that impair the retinoblastoma (RB) pathway in nearly 50% of FLs. These alterations include homozygous and heterozygous deletions of the p16/CDKN2a/b (7%) and RB1 (12%) loci, and more frequent gains of chromosome 12 that include CDK4 (29%). These aberrations are associated with high-risk disease by the FL prognostic index (FLIPI), and studies in a murine FL model confirm their pathogenic role in indolent FL. Increased CDK4 kinase activity toward RB1 is readily measured in tumor samples and indicates an opportunity for CDK4 inhibition. We find that dual CDK4 and BCL2 inhibitor treatment is safe and effective against available models of FL. In summary, frequent RB pathway lesions in indolent, high-risk FLs indicate an untapped therapeutic opportunity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfoma Folicular , Proteína do Retinoblastoma , Animais , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Humanos , Linfoma Folicular/tratamento farmacológico , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Linfoma Folicular/patologia , Masculino , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo
7.
Stem Cells Dev ; 23(10): 1149-56, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24383669

RESUMO

MicroRNAs (miRNAs) regulate myriad biological processes; however, their role in cell fate choice is relatively unexplored. Pluripotent NT2/D1 embryonal carcinoma cells differentiate into an epithelial/smooth muscle phenotype when treated with bone morphogenetic protein-2 (BMP-2). To identify miRNAs involved in epithelial cell development, we performed miRNA profiling of NT2/D1 cells treated with BMP-2 at 6, 12, and 24 h, and on days 6 and 10. Integration of the miRNA profiling data with previously obtained gene expression profiling (GEP) data of NT2/D1 cells treated with BMP-2 at the same time points identified miR-18b and miR-518b as the top two miRNAs with the highest number of up-regulated predicted targets with known functions in epithelial lineage development. Silencing of miR-18b and miR-518b in NT2/D1 cells revealed several up-regulated TFs with functions in epithelial lineage development; among these, target prediction programs identified FOXN1 as the only direct target of both miRNAs. FOXN1 has previously been shown to play an important role in keratinocyte differentiation and epithelial cell proliferation. NT2/D1 and H9 human embryonic stem cells with silenced miR-18b and miR-518b showed up-regulation of FOXN1 and the epithelial markers CDH1, EPCAM, KRT19, and KRT7. A 3'UTR luciferase assay confirmed FOXN1 to be a target of the two miRNAs, and up-regulation of FOXN1 in NT2/D1 cells led to the expression of epithelial markers. Overexpression of the two miRNAs in BMP-2-treated NT2/D1 cells led to down-regulation of FOXN1 and epithelial lineage markers. These results show that miR-18b and miR-518b are upstream controllers of FOXN1-directed epithelial lineage development.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/metabolismo , Fatores de Transcrição Forkhead/biossíntese , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/metabolismo , Linhagem Celular , Proliferação de Células/fisiologia , Regulação para Baixo/fisiologia , Células Epiteliais/citologia , Fatores de Transcrição Forkhead/genética , Humanos , MicroRNAs/genética , Células-Tronco Pluripotentes/citologia , Regulação para Cima/fisiologia
8.
Leuk Lymphoma ; 55(4): 920-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24047479

RESUMO

Array comparative genomic hybridization (aCGH) has yet to be fully leveraged in a prognostic setting in chronic lymphocytic leukemia (CLL). Genomic imbalance was assessed in 288 CLL specimens using a targeted array. Based on 20 aberrations in a hierarchical manner, all 228 treatment-naive specimens were classified into a group with poor outcome (20.6%) exhibiting at least one aberration that was univariately associated with adverse outcome (gain: 2p, 3q, 8q, 17q, loss: 7q, 8p, 11q, 17p, 18p), good outcome (32.5%) showing 13q14 loss without any of the other 10 aberrations (gain: 1p, 7p, 12, 18p, 18q, 19, loss: 4p, 5p, 6q, 7p) or intermediate outcome (remainder). The three groups were significantly separated with respect to time to first treatment and overall survival (p < 0.001), and validation of the stratification scheme was performed in two independent datasets. Gain of 3q and 8q, and 17p loss were determined to be independent unfavorable prognostic biomarkers. TP53, NOTCH1 and SF3B1 mutations correlated with the presence of one poor outcome aCGH marker, at a considerably higher frequency than when only considering poor risk aberrations routinely detected by fluorescence in situ hybridization (FISH). These data support genomic imbalance evaluation in CLL by aCGH to assist in risk stratification.


Assuntos
Aberrações Cromossômicas , Genômica , Leucemia Linfocítica Crônica de Células B/genética , Biomarcadores Tumorais/genética , Deleção Cromossômica , Cromossomos Humanos Par 13 , Hibridização Genômica Comparativa , Conjuntos de Dados como Assunto , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Hibridização in Situ Fluorescente , Leucemia Linfocítica Crônica de Células B/diagnóstico , Leucemia Linfocítica Crônica de Células B/mortalidade , Masculino , Mutação , Estadiamento de Neoplasias , Avaliação de Resultados da Assistência ao Paciente , Prognóstico
9.
Endocrine ; 44(2): 489-95, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23435613

RESUMO

Parathyroid cancer is a rare, clinically aggressive cause of primary hyperparathyroidism, and whether these malignancies generally evolve from pre-existing benign adenomas or arise de novo is unclear. Furthermore, while inactivation of the CDC73 (HRPT2) tumor suppressor gene, encoding parafibromin, is a major contributor, other genes essential to parathyroid carcinogenesis remain unknown. We sought to identify genomic regions potentially harboring such oncogenes or tumor suppressor genes, and to gain insight into the origins and molecular relationship of malignant versus benign parathyroid tumors. We performed genome-wide copy-number and loss of heterozygosity analysis using Affymetrix 50K SNP mapping arrays and/or comparative genomic hybridization on 16 primary parathyroid carcinomas, local recurrences or distant metastases, and matched normal controls, from 10 individuals. Recurrent regions of allelic loss were observed on chromosomes 1p, 3, and 13q suggesting that key parathyroid tumor suppressor genes are located in these chromosomal locations. Recurrent allelic gains were seen on chromosomes 1q and 16, suggesting the presence of parathyroid oncogenes on these chromosomes. Importantly, the most common alteration in benign parathyroid adenomas, loss of 11q, was not found as a recurrent change in the malignant parathyroid tissues. Molecular allelotyping using highly polymorphic microsatellite markers provided further confirmation that the prevalence of 11q loss is markedly and significantly lower in carcinomas as compared with adenomas. Our observations provide molecular support for the concept that sporadic parathyroid cancer usually arises de novo, rather than evolving from a pre-existing typical benign adenoma. Furthermore, these results help direct future investigation to ultimately determine which of the candidate genes in these chromosomal locations make significant contributions to the molecular pathogenesis of parathyroid cancer.


Assuntos
Adenoma/genética , Desequilíbrio Alélico/genética , Carcinoma/genética , Neoplasias das Paratireoides/genética , Adenoma/complicações , Adulto , Idoso , Carcinoma/complicações , Estudos de Coortes , Hibridização Genômica Comparativa , Variações do Número de Cópias de DNA , Feminino , Humanos , Hiperparatireoidismo Primário/complicações , Hiperparatireoidismo Primário/genética , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias das Paratireoides/complicações , Polimorfismo de Nucleotídeo Único , Adulto Jovem
10.
Oncogene ; 32(14): 1752-60, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22665059

RESUMO

The mechanism by which renal cell carcinoma (RCC) colonizes the lung microenvironment during metastasis remains largely unknown. To investigate this process, we grafted human RCC cells with varying lung metastatic potential in mice. Gene expression profiling of the mouse lung stromal compartment revealed a signature enriched for neutrophil-specific functions that was induced preferentially by poorly metastatic cells. Analysis of the gene expression signatures of tumor cell lines showed an inverse correlation between metastatic activity and the levels of a number of chemokines, including CXCL5 and IL8. Enforced depletion of CXCL5 and IL8 in these cell lines enabled us to establish a functional link between lung neutrophil infiltration, secretion of chemokines by cancer cells and metastatic activity. We further show that human neutrophils display a higher cytotoxic activity against poorly metastatic cells compared with highly metastatic cells. Together, these results support a model in which neutrophils recruited to the lung by tumor-secreted chemokines build an antimetastatic barrier with loss of neutrophil chemokines in tumor cells acting as a critical rate-limiting step during lung metastatic seeding.


Assuntos
Carcinoma de Células Renais/prevenção & controle , Quimiocinas/metabolismo , Neoplasias Renais/prevenção & controle , Neoplasias Pulmonares/prevenção & controle , Neutrófilos/metabolismo , Adulto , Animais , Apoptose , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células , Quimiocinas/antagonistas & inibidores , Quimiocinas/genética , Progressão da Doença , Imunofluorescência , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Cell ; 147(3): 554-64, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22036564

RESUMO

Insights into cancer genetics can lead to therapeutic opportunities. By cross-referencing chromosomal changes with an unbiased genetic screen we identify the ephrin receptor A7 (EPHA7) as a tumor suppressor in follicular lymphoma (FL). EPHA7 is a target of 6q deletions and inactivated in 72% of FLs. Knockdown of EPHA7 drives lymphoma development in a murine FL model. In analogy to its physiological function in brain development, a soluble splice variant of EPHA7 (EPHA7(TR)) interferes with another Eph-receptor and blocks oncogenic signals in lymphoma cells. Consistent with this drug-like activity, administration of the purified EPHA7(TR) protein produces antitumor effects against xenografted human lymphomas. Further, by fusing EPHA7(TR) to the anti-CD20 antibody (rituximab) we can directly target this tumor suppressor to lymphomas in vivo. Our study attests to the power of combining descriptive tumor genomics with functional screens and reveals EPHA7(TR) as tumor suppressor with immediate therapeutic potential.


Assuntos
Genes Supressores de Tumor , Linfoma Folicular/metabolismo , Receptor EphA7/metabolismo , Animais , Anticorpos Monoclonais Murinos/uso terapêutico , Linhagem Celular Tumoral , Cromossomos Humanos Par 6 , Genômica , Humanos , Linfoma Folicular/tratamento farmacológico , Linfoma Folicular/genética , Masculino , Camundongos , Transplante de Neoplasias , Interferência de RNA , Rituximab , Transplante Heterólogo
12.
BJU Int ; 104(9 Pt B): 1334-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19840009

RESUMO

Germ cell tumours (GCTs) represent the leading cause of cancer-related morbidity and mortality in young men aged 18-35 years. Transformation of the cell of origin results in tumours with several unique properties. GCTs are characterized by gain of the short arm of chromosome 12 in almost all cases, a frequency of genomic alteration not seen in any other solid tumours. GCTs are truly pluripotent, giving rise to cells of somatic and extra-embryonic lineages, which results in tumours with a spectrum of differentiation that rivals that seen in normal embryogenesis and development. Despite the presence of genomic instability and many oncogenic changes, GCTs are highly curable, even in the metastatic setting, due to their extreme sensitivity to cisplatin-based chemotherapy. In this review we highlight some of the molecular events associated with the genesis, differentiation and chemotherapeutic response of these tumours, and discuss how these alterations are linked with biological features unique to germ cells.


Assuntos
Antineoplásicos/uso terapêutico , Cromossomos Humanos Par 12/genética , Cisplatino/uso terapêutico , Neoplasias Embrionárias de Células Germinativas/genética , Células-Tronco Pluripotentes/efeitos dos fármacos , Neoplasias Testiculares/genética , Adolescente , Adulto , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/patologia , Adulto Jovem
13.
J Clin Oncol ; 27(31): 5240-7, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19770384

RESUMO

PURPOSE: Germ cell tumor (GCT) is the most common malignancy in young adult men. Currently, patients are risk-stratified on the basis of clinical presentation and serum tumor markers. The introduction of molecular markers could improve outcome prediction. PATIENTS AND METHODS: Expression profiling was performed on 74 nonseminomatous GCTs (NSGCTs) from cisplatin-treated patients (ie, training set) and on 34 similarly treated patients with NSGCTs (ie, validation set). A gene classifier was developed by using prediction analysis for microarrays (PAM) for the binary end point of 5-year overall survival (OS). A predictive score was developed for OS by using the univariate Cox model. RESULTS: In the training set, PAM identified 140 genes that predicted 5-year OS (cross-validated classification rate, 60%). The PAM model correctly classified 90% of patients in the validation set. Patients predicted to have good outcome had significantly longer survival than those with poor predicted outcome (P < .001). For the OS end point, a 10-gene model had a predictive accuracy (ie, concordance index) of 0.66 in the training set and a concordance index of 0.83 in the validation set. Dichotomization of the samples on the basis of the median score resulted in significant differences in survival (P = .002). For both end points, the gene-based predictor was an independent prognostic factor in a multivariate model that included clinical risk stratification (P < .01 for both). CONCLUSION: We have identified gene expression signatures that accurately predict outcome in patients with GCTs. These predictive genes should be useful for the prediction of patient outcome and could provide novel targets for therapeutic intervention.


Assuntos
Biomarcadores Tumorais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Neoplasias Embrionárias de Células Germinativas/genética , Adolescente , Adulto , Idoso , Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/mortalidade , Prognóstico , Resultado do Tratamento , Adulto Jovem
14.
Leuk Lymphoma ; 49(11): 2170-80, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19021061

RESUMO

The principal curative agent in the front-line treatment of patients with diffuse large B-cell lymphoma (DLBCL) is the anthracycline, doxorubicin. To define pathways that may have a functional role in the response of DLBCL in vivo to doxorubicin-based therapies, seven DLBCL cell lines were treated with doxorubicin and the cellular response evaluated. Expression profiling of responses revealed changes in levels of genes consistent with discrete pathway activation that were confirmed functionally. The two most sensitive cell lines (Ly3 and Ly10) displayed activation of the TP53 pathway but not in the remaining five (Ly1, Ly2, Ly4, Ly7 and Ly8), where TP53 mutations were identified. In this latter group, a G2/M delay was invoked. NF-kappaB pathway activation was evident in Ly1 which with Ly4 displayed the most chemoresistant response. Treatment of Ly1 after doxorubicin with the proteasomic inhibitor, bortezomib, additively increased the cytotoxic effect of doxorubicin. Chemoresistance of Ly4 was associated with loss of chromosome 2 (0-9 Mbp) that in vivo was highly correlated with adverse outcome. Thus, the response of DLBCL in vivo and in vitro is defined by several distinct molecular and genetic pathways which is, perhaps, not surprising given the heterogeneous clinical, morphologic and genetic nature of DLBCL.


Assuntos
Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Biologia de Sistemas , Biomarcadores Tumorais , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Resistência a Medicamentos/genética , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Humanos , Linfoma Difuso de Grandes Células B/genética , NF-kappa B/metabolismo , Prognóstico , Pirazinas/farmacologia , Biologia de Sistemas/métodos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
15.
Genes Chromosomes Cancer ; 47(1): 43-55, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17943972

RESUMO

Germ cell tumors (GCTs) are the most common solid malignancy in young adult men, but the genes and genomic regions involved in their etiology are not fully defined. We report here an investigation of DNA copy number changes in GCTs using 1 Mb BAC arrays. As expected, 12p gain was the defining genomic alteration, occurring in 72/74 GCTs. Parallel expression profiling of these tumors identified potential oncogenes from gained regions (LYN and RAB25) and potential tumor suppressor genes in regions of loss (SYNPO2, TTC12, IGSF4, and EPB41L3). Notably, we observed specific genomic alterations associated with histology, including gain of 17p11.2-q21.32 and loss of 2p25.3 in embryonal carcinoma, gain of 8p23.3-12 and loss of 5p15.33-35.3, 11q23.1-25, and 13q12.11-34 in seminoma, and gain of 1q31.3-42.3, 3p, 14q11.2-32.33, and 20q and loss of 8q11.1-23.1 in yolk sac tumors (YST). Many significant genes that mapped to these regions had previously been associated with specific histologies, such as EOMES (chr3) and BMP2 (chr20) in YST and SPRY2 (chr13) and SOX17 (chr8) in seminomas. Additionally, our results suggest a model in which histologic differentiation of GCTs may drive genomic evolution.


Assuntos
Diferenciação Celular/genética , Evolução Molecular , Genoma Humano , Germinoma/genética , Neoplasias Testiculares/genética , Adulto , Cromossomos Artificiais Bacterianos , Feminino , Dosagem de Genes , Perfilação da Expressão Gênica , Humanos , Masculino , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos
16.
Cytogenet Genome Res ; 118(2-4): 337-44, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18000388

RESUMO

We analyzed a cohort of 61 follicular lymphomas (FL) with an abnormal G-banded karyotype by spectral karyotyping (SKY) to better define the chromosome instability associated with the t(14;18)(q32;q21) positive and negative subsets of FL and histologic grade. In more than 70% of the patients, SKY provided additional cytogenetic information and up to 40% of the structural abnormalities were revised. The six most frequent breakpoints in both SKY and G-banding analyses were 14q32, 18q21, 3q27, 1q11-q21, 6q11-q15 and 1p36 (15-77%). SKY detected nine additional sites (1p11-p13, 2p11-p13, 6q21, 8q24, 6q21, 9p13, 10q22-q24, 12q11-q13 and 17q11-q21) at an incidence of >10%. In addition to the known recurring translocations, t(14;18)(q32;q21) [70%], t(3;14)(q27;q32) [10%], t(1;14)(q21;q32) [5%] and t(8;14)(q24;q32) [2%] and their variants, 125 non-IG gene translocations were identified of which four were recurrent within this series. In contrast to G-banding analysis, SKY revealed a greater degree of karyotypic instability in the t(14;18) (q32;q21) negative subset compared to the t(14;18)(q32;q21) positive subset. Translocations of 3q27 and gains of chromosome 1 were significantly more frequent in the former subset. SKY also allowed a better definition of chromosomal imbalances, thus 37% of the deletions detected by G-banding were shown to be unbalanced translocations leading to gain of genetic material. The majority of recurring (>10%) imbalances were detected at a greater (2-3 fold) incidence by SKY and several regions were narrowed down, notably at gain 2p13-p21, 2q11-q21, 2q31-q37, 12q12-q15, 17q21-q25 and 18q21. Chromosomal abnormalities among the different histologic grades were consistent with an evolution from low to high grade disease and breaks at 6q11-q15 and 8q24 and gain of 7/7q and 8/8q associated significantly with histologic progression. This study also indicates that in addition to gains and losses, non-IG gene translocations involving 1p11-p13, 1p36, 1q11-q21, 8q24, 9p13, and 17q11-q21 play an important role in the histologic progression of FL with t(14;18)(q32;q21) and t(3q27).


Assuntos
Instabilidade Cromossômica , Cromossomos Humanos Par 14 , Cromossomos Humanos Par 18 , Linfoma Folicular/genética , Translocação Genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Bandeamento Cromossômico , Progressão da Doença , Feminino , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Linfoma Folicular/patologia , Masculino , Pessoa de Meia-Idade
17.
J Urol ; 177(4): 1264-8; discussion 1268, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17382702

RESUMO

PURPOSE: We identified differentially expressed genes associated with response to pegylated interferon-alpha treatment in patients with renal cell carcinoma. MATERIALS AND METHODS: We performed expression profiling on renal cell carcinoma specimens isolated from 23 patients with metastatic disease who were subsequently treated with interferon. Significance Analysis for Microarrays software was used to identify genes that were differentially expressed between patients with partial response compared to those with disease progression. RESULTS: A candidate gene approach looking at VHL and known target genes did not identify any genes whose expression correlated with patient response. A global analysis of approximately 54,000 probe sets identified 4 genes that had expression correlated with response. Reverse transcriptase-polymerase chain reaction analysis of 2 of these genes confirmed that they were more highly expressed in tumors from patients who responded to interferon-alpha. Interestingly, both of these genes mapped to 4q31-32, a region that has been implicated as the site of a potential tumor suppressor gene in renal cell carcinoma. CONCLUSIONS: We have identified 4 genes (3 uncharacterized and 1 known) that may prove useful in predicting response to interferon-alpha treatment in patients with renal cell carcinoma.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Interferon-alfa/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Humanos
18.
Genes Chromosomes Cancer ; 46(4): 373-84, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17243165

RESUMO

Cervical cancer (CC) cells exhibit complex karyotypic alterations, which is consistent with deregulation of numerous critical genes in its formation and progression. To characterize this karyotypic complexity at the molecular level, we used cDNA array comparative genomic hybridization (aCGH) to analyze 29 CC cases and identified a number of over represented and deleted genes. The aCGH analysis revealed at least 17 recurrent amplicons and six common regions of deletions. These regions contain several known tumor-associated genes, such as those involved in transcription, apoptosis, cytoskeletal remodeling, ion-transport, drug metabolism, and immune response. Using the fluorescence in situ hybridization (FISH) approach we demonstrated the presence of high-level amplifications at the 8q24.3, 11q22.2, and 20q13 regions in CC cell lines. To identify amplification-associated genes that correspond to focal amplicons, we examined one or more genes in each of the 17 amplicons by Affymetrix U133A expression arrays and semiquantitative reverse-transcription PCR (RT-PCR) in 31 CC tumors. This analysis exhibited frequent and robust upregulated expression in CC relative to normal cervix for genes EPHB2 (1p36), CDCA8 (1p34.3), AIM2 (1q22-23), RFC4, MUC4, and HRASLS (3q27-29), SKP2 (5p12-13), CENTD3 (5q31.3), PTK2, RECQL4 (8q24), MMP1 and MMP13 (11q22.2), AKT1 (14q32.3), ABCC3 (17q21-22), SMARCA4 (19p13.3) LIG1 (19q13.3), UBE2C (20q13.1), SMC1L1 (Xp11), KIF4A (Xq12), TMSNB (Xq22), and CSAG2 (Xq28). Thus, the gene dosage and expression profiles generated here have enabled the identification of focal amplicons characteristic for the CC genome and facilitated the validation of relevant genes in these amplicons. These data, thus, form an important step toward the identification of biologically relevant genes in CC pathogenesis. This article contains Supplementary Material available at http://www.interscience.wiley.com/jpages/1045-2257/suppmat.


Assuntos
Amplificação de Genes , Dosagem de Genes , Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias do Colo do Útero/genética , Linhagem Celular Tumoral , Feminino , Humanos
19.
Stem Cells ; 25(3): 771-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17138961

RESUMO

Human embryonal carcinoma (EC) cell lines exhibit considerable heterogeneity in their levels of pluripotency. Thus, NT2/D1 cells differentiate into neural lineages upon exposure to all-trans retinoic acid (ATRA) and non-neural epithelial lineages upon exposure to bone morphogenetic protein-2 (BMP-2). In contrast, 27X-1 cells differentiate into extra-embryonic endodermal (ExE) cells upon treatment with either morphogen. To understand the molecular basis for the differential responses of the two cell lines, we performed gene expression profiling at the undifferentiated EC cell line state to identify constitutive differences in gene expression. NT2/D1 cells preferentially expressed transcripts associated with neurectodermal development, whereas 27X-1 cells expressed high levels of transcripts associated with mesendodermal characteristics. We then determined temporal expression profiles of 27X-1 cells during ExE differentiation upon treatment with ATRA and BMP-2 and compared the data with changes in gene expression observed during BMP-2- and ATRA-induced differentiation of NT2/D1 cells. ATRA and BMP-2 induced distinct sets of transcription factors and phenotypic markers in the two EC cell lines, underlying distinct lineage choices. Although 27X-1 differentiation yielded comprehensive gene expression profiles of parietal endodermal lineages, we were able to use the combined analysis of 27X-1 data with data derived from yolk sac tumors for the identification of transcripts associated with visceral endoderm formation. Our results demonstrate constitutive differences in the levels of pluripotency between NT2/D1 and 27X-1 cells that correlate with lineage potential. This study also demonstrates that EC cells can serve as robust models to investigate early lineage choices during both embryonic and extra-embryonic human development.


Assuntos
Carcinoma Embrionário/embriologia , Carcinoma Embrionário/genética , Regulação Neoplásica da Expressão Gênica , Oligopeptídeos/genética , Carcinoma Embrionário/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Cinética , Morfogênese , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tretinoína/farmacologia
20.
Clin Cancer Res ; 12(24): 7215-20, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17189392

RESUMO

Inheritance of a defective copy of the von Hippel-Lindau (VHL) gene leads to the most common cause of inherited renal cell carcinoma (RCC). In addition, most patients with sporadic RCC have aberrant VHL. In the absence of VHL, hypoxia-inducible factor alpha accumulates, leading to production of several growth factors, including vascular endothelial growth factor and platelet-derived growth factor. We review here the biology of RCC and how a combination of proximal and distal block of VHL/hypoxia-inducible factor alpha pathway by novel targeted agents, including sunitinib, sorafenib, bevacizumab, everolimus, and temsirolimus, has led to significant improvements in progression-free survival.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Proteína Supressora de Tumor Von Hippel-Lindau/antagonistas & inibidores , Benzenossulfonatos/uso terapêutico , Pesquisa Biomédica/tendências , Inibidores Enzimáticos/química , Everolimo , Humanos , Indóis/uso terapêutico , Modelos Biológicos , Niacinamida/análogos & derivados , Compostos de Fenilureia , Piridinas/uso terapêutico , Pirróis/uso terapêutico , Transdução de Sinais , Sirolimo/análogos & derivados , Sirolimo/uso terapêutico , Sorafenibe , Sunitinibe , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...