Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Drug Metab Dispos ; 49(3): 179-187, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33376147

RESUMO

CYP2D6 is a major drug metabolizing enzyme with a buried active site. Channels leading to the active site from various enzyme surfaces are believed to facilitate ligand egress and access to the active site. The present study used molecular dynamics (MD) and in vitro studies with CYP2D6*1 and a Trp75-to-Ala mutant to examine channel gating in CYP2D6 by Trp75. MD simulations measured energy landscapes of Trp75 conformations and simulated substrate passage within channel 2b using bufuralol as a model substrate. Trp75 alternated between multiple stable states that supported substrate transport along channel 2b with low-energy barriers between states (∼ -1 kcal/mol). Trp75 conformations were stabilized primarily by hydrogen bonding between Trp75 and Glu222, Asn226, Ala225, or Gln72. Energy barriers were low between Trp75 conformations, allowing Trp75 to easily move between various conformations over time and to function in both binding to and moving substrates in the 2b channel of CYP2D6. Michaelis-Menten kinetic studies completed with purified enzyme in a reconstituted system showed overall reduced enzyme efficiency for metabolism of bufuralol and dextromethorphan by the Trp75Ala mutant compared with CYP2D6*1. In stopped-flow measurements, k off for dextromethorphan was decreased in the absence of Trp75. Our results support a role for Trp75 in substrate shuttling to the active site of CYP2D6. SIGNIFICANCE STATEMENT: Using combined molecular dynamics and in vitro assays, this study shows for the first time a role for Trp75 as a channel entrance gating residue in the mechanism of substrate binding/unbinding in CYP2D6. Energy landscapes derived from molecular dynamics were used to quantitate the strength of gating, and kinetics assays showed the impact on enzyme efficiency and k off of a Trp75Ala mutation.


Assuntos
Citocromo P-450 CYP2D6/metabolismo , Ativação do Canal Iônico/fisiologia , Triptofano/metabolismo , Animais , Cristalografia por Raios X/métodos , Citocromo P-450 CYP2D6/química , Etanolaminas/metabolismo , Etanolaminas/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Estrutura Secundária de Proteína , Ratos , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia , Triptofano/química
2.
Drug Metab Dispos ; 47(6): 567-573, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30952677

RESUMO

Rolapitant [(Varubi), 5S,8S)-8-[[(1R)-1-[3,5 bis(trifluoromethyl phenyl]ethoxy]methyl]-8-phenyl-1,7-diazaspiro[4.5]decan-2-one] is a high-affinity NK1 receptor antagonist that was approved in September 2015 as a treatment for nausea and vomiting caused by chemotherapy. In vivo rolapitant moderately inhibits CYP2D6 for at least 7 days after one 180 mg dose. Due to the long inhibition time, we investigated rolapitant as a possible mechanism-based inactivator of CYP2D6. Rolapitant docked in the active site of CYP2D6 and displayed type I binding to CYP2D6 with a K s value of 1.2 ± 0.4 µM. However, in NADPH-, time-, and concentration-dependent assays of CYP2D6 activity, no evidence for mechanism-based inactivation and no metabolites of rolapitant were observed. Stopped-flow binding studies yielded a kon /koff (K d) value of 6.2 µM. The IC50 value for rolapitant inhibition of CYP2D6 activity was 24 µM, suggesting that inhibition is not due to tight binding of rolapitant to CYP2D6. By Lineweaver-Burk analysis, rolapitant behaved as a mixed, reversible inhibitor. The K i values of 20 and 34 µM were determined by Dixon analysis, with bufuralol and dextromethorphan as reporter substrates, respectively, and drug-drug interaction modeling did not predict the reported in vivo inhibition. The interaction of rolapitant with CYP2D6 was also examined in 1 microsecond molecular dynamics simulations. Rolapitant adopted multiple low-energy binding conformations near the active site, but at distances not consistent with metabolism. Given these findings, we do not see evidence that rolapitant is a mechanism-based inactivator. Moreover, the reversible inhibition of CYP2D6 by rolapitant may not fully account for the moderate inhibition described in vivo.


Assuntos
Inibidores do Citocromo P-450 CYP2D6/uso terapêutico , Citocromo P-450 CYP2D6/metabolismo , Compostos de Espiro/uso terapêutico , Domínio Catalítico/fisiologia , Dextrometorfano/uso terapêutico , Interações Medicamentosas/fisiologia , Etanolaminas/uso terapêutico , Humanos
3.
Kidney Int ; 88(5): 1057-69, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26083655

RESUMO

The study of kidney cancer pathogenesis and its treatment has been limited by the scarcity of genetically defined animal models. The FLCN gene that codes for the protein folliculin, mutated in Birt-Hogg-Dubé syndrome, presents a new target for mouse modeling of kidney cancer. Here we developed a kidney-specific knockout model by disrupting the mouse Flcn in the proximal tubules, thus avoiding homozygous embryonic lethality or neonatal mortality, and eliminating the requirement of loss of heterozygosity for tumorigenesis. This knockout develops renal cysts and early onset (6 months) of multiple histological subtypes of renal neoplasms featuring high tumor penetrance. Although the majority of the tumors were chromophobe renal cell carcinomas in affected mice under 1 year of age, papillary renal cell carcinomas predominated in the kidneys of older knockout mice. This renal neoplasia from cystic hyperplasia at 4 months to high-grade renal tumors by 16 months represented the progression of tumorigenesis. The mTOR and TGF-ß signalings were upregulated in Flcn-deficient tumors, and these two activated pathways may synergetically cause renal tumorigenesis. Treatment of knockout mice with the mTOR inhibitor rapamycin for 10 months led to the suppression of tumor growth. Thus, our model recapitulates human Birt-Hogg-Dubé kidney tumorigenesis, provides a valuable tool for further study of Flcn-deficient renal tumorigenesis, and tests new drugs/approaches to their treatment.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Cistos/patologia , Modelos Animais de Doenças , Neoplasias Renais/genética , Neoplasias Renais/patologia , Túbulos Renais Proximais/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética , Animais , Antibióticos Antineoplásicos/uso terapêutico , Carcinogênese/genética , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Cistos/genética , Hiperplasia/patologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Genes Cancer ; 4(7-8): 247-60, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24167653

RESUMO

Hepatitis B virus (HBV) is a well-known cause of hepatocellular carcinoma (HCC), but the regulators effectively driving virus production and HCC progression remain unclear. By using genetically engineered mouse models, we show that overexpression of hepatocyte growth factor (HGF) accelerated HCC progression, supporting the genomic analysis that an up-regulated HGF signature is associated with poor prognosis in HBV-positive HCC patients. We show that for both liver regeneration and spontaneous HCC development there is an inclusive requirement for MET expression, and when HGF induces autocrine activation the tumor displays sensitivity to a small-molecule Met inhibitor. Our results demonstrate that HGF is a driver of HBV-induced HCC progression and may serve as an effective biomarker for Met-targeted therapy. MET inhibitors are entering clinical trials against cancer, and our data provide a molecular basis for targeting the Met pathway in hepatitis B-induced HCC.

5.
Invest Ophthalmol Vis Sci ; 54(10): 6472-80, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24003089

RESUMO

PURPOSE: To identify inflammatory cytokines significantly elevated and independent of VEGF levels in the vitreous of proliferative diabetic retinopathy (PDR) patients that may serve as novel diagnostic factors or therapeutic targets. METHODS: Thirty-nine cytokines and chemokines were measured from the vitreous of 72 patients undergoing vitrectomy (29 controls and 43 PDR) via a magnetic bead-based immunoassay. Patient information, including sex, age, history of smoking, cancer diagnosis and treatment, and presence of diabetes and hypertension were also collected. Univariate and multivariate logistic regression analyses were performed to assess the association of cytokine concentrations and patient demographics with disease. RESULTS: Nineteen cytokines were significantly elevated in the vitreous of PDR patients compared with controls, including five novel cytokines that have not previously been associated with PDR: sCD40L, GM-CSF, IFNα2, IL-12p40, and MCP-3. Sixteen cytokines were found to be statistically independent of VEGF. Of these, 14 show a statistically significant interaction with VEGF, while two do not. With regards to patient demographics, age and hypertension were statistically significant risk factors with the odds of disease decreasing with increasing age and increasing 3-fold for hypertensive patients. CONCLUSIONS: This is the first report of a comprehensive multiplex analysis to identify novel VEGF-independent cytokines associated with PDR. Of the 39 inflammatory cytokines tested, 16 are predictive of disease risk, independent of VEGF levels. These PDR-associated cytokines represent potential targets in the treatment of PDR, both in conjunction with anti-VEGF therapy, as well as for patients that are nonresponders to such therapy.


Assuntos
Citocinas/metabolismo , Retinopatia Diabética/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vitreorretinopatia Proliferativa/metabolismo , Corpo Vítreo/química , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Retinopatia Diabética/complicações , Retinopatia Diabética/cirurgia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vitrectomia , Vitreorretinopatia Proliferativa/etiologia , Vitreorretinopatia Proliferativa/cirurgia
6.
Mol Cancer Ther ; 12(9): 1701-14, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804705

RESUMO

Angiosarcoma is a rare neoplasm of endothelial origin that has limited treatment options and poor five-year survival. As a model for human angiosarcoma, we studied primary cells and tumorgrafts derived from canine hemangiosarcoma (HSA), which is also an endothelial malignancy with similar presentation and histology. Primary cells isolated from HSA showed constitutive extracellular signal-regulated kinase (ERK) activation. The mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor CI-1040 reduced ERK activation and the viability of primary cells derived from visceral, cutaneous, and cardiac HSA in vitro. HSA-derived primary cells were also sensitive to sorafenib, an inhibitor of B-Raf and multireceptor tyrosine kinases. In vivo, CI-1040 or PD0325901 decreased the growth of cutaneous cell-derived xenografts and cardiac-derived tumorgrafts. Sorafenib decreased tumor size in both in vivo models, although cardiac tumorgrafts were more sensitive. In human angiosarcoma, we noted that 50% of tumors stained positively for phosphorylated ERK1/2 and that the expression of several MEK-responsive transcription factors was upregulated. Our data showed that MEK signaling is essential for the growth of HSA in vitro and in vivo and provided evidence that the same pathways are activated in human angiosarcoma. This indicates that MEK inhibitors may form part of an effective therapeutic strategy for the treatment of canine HSA or human angiosarcoma, and it highlights the use of spontaneous canine cancers as a model of human disease.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Difenilamina/análogos & derivados , Hemangiossarcoma/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Animais , Difenilamina/farmacologia , Modelos Animais de Doenças , Cães , Ensaios de Seleção de Medicamentos Antitumorais , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Hemangiossarcoma/tratamento farmacológico , Hemangiossarcoma/metabolismo , Hemangiossarcoma/veterinária , Humanos , Camundongos , Camundongos Nus , Niacinamida/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sorafenibe , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Res ; 73(7): 2044-51, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23365135

RESUMO

Sustained activation of the stress-regulated transcription factor NRF2 (NFE2L2) is a prominent feature of many types of cancer, implying that mutations driving NRF2 may be important to tumor progression. In hereditary type 2 papillary renal cell carcinoma (PRCC2, also known as hereditary leiomyomatosis and renal cell cancer), NRF2 activation is a direct consequence of the accumulation of intracellular fumarate, a result of fumarate hydratase (FH) inactivation, but it is not clear how NRF2 may be activated in sporadic forms of PRCC2. Here we show that somatic mutations in NRF2, CUL3, and SIRT1 are responsible for driving the NRF2 activation phenotype in sporadic PRCC2. Transcriptome sequencing revealed the expression pattern of mutant alleles of NRF2, CUL3, and SIRT1 and also confirmed NRF2 activation in clinical specimens. Our results show a convergence in somatic mutations in sporadic PRCC2 with FH mutation in hereditary PRCC2.


Assuntos
Carcinoma de Células Renais/genética , Proteínas Culina/genética , Neoplasias Renais/genética , Mutação/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Sequência de Aminoácidos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Células Cultivadas , Proteínas Culina/metabolismo , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Homologia de Sequência de Aminoácidos , Sirtuína 1/metabolismo
8.
Pediatr Res ; 73(4 Pt 1): 450-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23269123

RESUMO

BACKGROUND: Gene expression in archived newborn blood spots remaining from newborn screening may reflect pathophysiological disturbances useful in understanding the etiology of cerebral palsy (CP). METHODS: We quantified the expression of gene sets representing four physiological pathways hypothesized to contribute to CP in archived unfrozen residual newborn blood spot specimens from 53 children with CP and 53 age-, gender-, and gestational age-matched controls. We selected four empirical and three canonical gene sets representing the inflammatory, hypoxic, coagulative, and thyroidal pathways and examined mRNA expression using an 8 × 60,000 oligonucleotide microarray. The log2 fold change of gene expression between matched cases and controls was analyzed using the generally applicable gene set enrichment method. RESULTS: The empirical inflammatory and empirical hypoxic gene sets were significantly downregulated in term-born CP cases (n = 33) as compared with matched controls (P = 0.0007 and 0.0009, respectively), whereas both gene sets were significantly upregulated (P =0.0055 and 0.0223, respectively) in preterm-born CP cases (n = 20). The empirical thyroidal gene set was significantly upregulated in preterm-born CP cases (P = 0.0023). CONCLUSION: The newborn blood spot transcriptome can serve as a platform for investigating distinctive gene expression patterns in children who later develop CP.


Assuntos
Paralisia Cerebral/genética , Teste em Amostras de Sangue Seco , Perfilação da Expressão Gênica , Testes Genéticos , Triagem Neonatal/métodos , Adolescente , Estudos de Casos e Controles , Paralisia Cerebral/sangue , Paralisia Cerebral/diagnóstico , Criança , Pré-Escolar , Feminino , Redes Reguladoras de Genes , Marcadores Genéticos , Predisposição Genética para Doença , Idade Gestacional , Humanos , Recém-Nascido , Masculino , Fenótipo , Reação em Cadeia da Polimerase , Valor Preditivo dos Testes , Reprodutibilidade dos Testes
9.
Int J Mol Sci ; 13(8): 9599-9608, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22949818

RESUMO

Screening newborns for treatable serious conditions is mandated in all US states and many other countries. After screening, Guthrie cards with residual blood (whole spots or portions of spots) are typically stored at ambient temperature in many facilities. The potential of archived dried blood spots (DBS) for at-birth molecular studies in epidemiological and clinical research is substantial. However, it is also challenging as analytes from DBS may be degraded due to preparation and storage conditions. We previously reported an improved assay for obtaining global RNA gene expression from blood spots. Here, we evaluated sex-specific gene expression and its preservation in DBS using oligonucleotide microarray technology. We found X inactivation-specific transcript (XIST), lysine-specific demethylase 5D (KDM5D) (also known as selected cDNA on Y, homolog of mouse (SMCY)), uncharacterized LOC729444 (LOC729444), and testis-specific transcript, Y-linked 21 (TTTY21) to be differentially-expressed by sex of the newborn. Our finding that trait-specific RNA gene expression is preserved in unfrozen DBS, demonstrates the technical feasibility of performing molecular genetic profiling using such samples. With millions of DBS potentially available for research, we see new opportunities in using newborn molecular gene expression to better understand molecular pathogenesis of perinatal diseases.


Assuntos
Biomarcadores/análise , Coleta de Amostras Sanguíneas , Paralisia Cerebral/genética , Perfilação da Expressão Gênica , Triagem Neonatal , Adolescente , Animais , Estudos de Casos e Controles , Paralisia Cerebral/sangue , Paralisia Cerebral/diagnóstico , Criança , Pré-Escolar , Feminino , Histona Desmetilases/genética , Humanos , Recém-Nascido , Masculino , Camundongos , Antígenos de Histocompatibilidade Menor , Análise de Sequência com Séries de Oligonucleotídeos , RNA Longo não Codificante/genética , Reação em Cadeia da Polimerase em Tempo Real
10.
BMC Res Notes ; 5: 370, 2012 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-22824328

RESUMO

BACKGROUND: High-throughput methods that ascribe a cellular or physiological function for each gene product are useful to understand the roles of genes that have not been extensively characterized by molecular or genetic approaches. One method to infer gene function is "guilt-by-association", in which the expression pattern of a poorly characterized gene is shown to co-vary with the expression of better-characterized genes. The function of the poorly characterized gene is inferred from the known function(s) of the well-described genes. For example, genes co-expressed with transcripts that vary during the cell cycle, development, environmental stresses, and with oncogenesis have been implicated in those processes. FINDINGS: While examining the expression characteristics of several poorly characterized genes, we noted that we could associate each of the genes with a cellular phenotype by correlating individual gene expression changes with gene set enrichment scores from individual samples. We evaluated the effectiveness of this approach using a modest sized gene expression data set (expO) and a compendium of gene expression phenotypes (MSigDBv3.0). We found the transcripts that correlated best with enrichment in mitochondrial and lysosomal gene sets were mostly related to those processes (89/100 and 44/50, respectively). The reciprocal evaluation, ranking gene sets according to correlation of enrichment with an individual gene's expression, also reflected known associations for prominent genes in the biomedical literature (16/19). In evaluating the model, we also found that 4% of the genome encodes proteins that are associated with small molecule and small peptide signal transduction gene sets, implicating a large number of genes in both internal and external environmental sensing. CONCLUSIONS: Our results show that this approach is useful to infer functions of disparate sets of genes. This method mirrors the biological experimental approaches used by others to associate individual genes with defined gene expression changes. Moreover, the approach can be used beyond discovering genes related to a cellular process to discover meaningful expression phenotypes from a compendium that are associated with a given gene. The effectiveness, versatility, and breadth of this approach make possible its application in a variety of contexts and with a variety of downstream analyses.


Assuntos
Genes Mitocondriais , Genoma Humano , Modelos Logísticos , Mitocôndrias/genética , Modelos Genéticos , RNA Mensageiro/genética , Algoritmos , Bases de Dados Genéticas , Expressão Gênica , Perfilação da Expressão Gênica , Interação Gene-Ambiente , Estudo de Associação Genômica Ampla , Humanos , Lisossomos/genética
11.
Cancer Res ; 72(17): 4361-71, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22805307

RESUMO

The pituitary tumor transforming gene (PTTG1) is a recently discovered oncogene implicated in malignant progression of both endocrine and nonendocrine malignancies. Clear cell renal cell carcinoma (ccRCC) is cytogenetically characterized by chromosome 3p deletions that harbor the ccRCC-related von Hippel-Lindau, PBRM1, BAP1, and SETD2 tumor suppressor genes, along with chromosome 5q amplifications where the significance has been unclear. PTTG1 localizes to the chromosome 5q region where amplifications occur in ccRCC. In this study, we report a functional role for PTTG1 in ccRCC tumorigenesis. PTTG1 was amplified in ccRCC, overexpressed in tumor tissue, and associated with high-grade tumor cells and poor patient prognosis. In preclinical models, PTTG1 ablation reduced tumorigenesis and invasion. An analysis of gene expression affected by PTTG1 indicated an association with invasive and metastatic disease. PTTG1-dependent expression of the RhoGEF proto-oncogene ECT2 was observed in a number of ccRCC cell lines. Moreover, ECT2 expression correlated with PTTG1 expression and poor clinical features. Together, our findings reveal features of PTTG1 that are consistent with its identification of an oncogene amplified on chromsome 5q in ccRCC, where it may offer a novel therapeutic target of pathologic significance in this disease.


Assuntos
Carcinoma de Células Renais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/genética , Proteínas de Neoplasias/genética , Animais , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Cromossomos Humanos Par 5 , Análise por Conglomerados , Amplificação de Genes , Dosagem de Genes , Expressão Gênica , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Proto-Oncogene Mas , Securina , Transplante Heterólogo
12.
Chin J Cancer ; 31(9): 413-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22776233

RESUMO

Biallelic inactivation of fumarate hydratase(FH) causes type 2 papillary renal cell carcinoma (PRCC2), uterine fibroids, and cutaneous leimyomas, a condition known as hereditary leiomyomatosis and renal cell cancer(HLRCC). The most direct effect of FH inactivation is intracellular fumarate accumulation. A majority of studies on FH inactivation over the past decade have focused on the theory that intracellular fumarate stabilizes hypoxia-inducible factor 1α(HIF1A) through competitive inhibition of HIF prolyl hydroxylases. Recently, a competing theory that intracellular fumarate activates nuclear factor (erythroid-derived 2)-like 2(NRF2) through post-translational modification of its negative regulator. Kelch-like ECH-associated protein 1(KEAP1) has emerged from a computational modeling study and mouse model studies. This review dissects the origin of these two governing theories and highlights the presence of chromatin-structure-regulated targets of transcription factors, which we refer to as "cryptic targets" of transcription factors. One such cryptic target is heme oxygenase I(HMOX1), the expression of which is known to be modulated by the gene product of SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4, also known as BRG1).


Assuntos
Fumarato Hidratase/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias Renais/metabolismo , Leiomiomatose/metabolismo , Fator 2 Relacionado a NF-E2 , Síndromes Neoplásicas Hereditárias/metabolismo , Animais , DNA Helicases/metabolismo , Fumarato Hidratase/metabolismo , Fumaratos/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Neoplasias Renais/genética , Leiomiomatose/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Síndromes Neoplásicas Hereditárias/genética , Proteínas Nucleares/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Processamento de Proteína Pós-Traducional , Neoplasias Cutâneas , Fatores de Transcrição/metabolismo , Neoplasias Uterinas
13.
PLoS One ; 7(6): e38955, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701735

RESUMO

Epigenetic silencing is one of the mechanisms leading to inactivation of a tumor suppressor gene, either by DNA methylation or histone modification in a promoter regulatory region. Mitogen inducible gene 6 (MIG-6), mainly known as a negative feedback inhibitor of the epidermal growth factor receptor (EGFR) family, is a tumor suppressor gene that is associated with many human cancers. To determine if MIG-6 is inactivated by epigenetic alteration, we identified a group of human lung cancer and melanoma cell lines in which its expression is either low or undetectable and studied the effects of methylation and of histone deacetylation on its expression. The DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza-dC) induced MIG-6 expression in melanoma cell lines but little in lung cancer lines. By contrast, the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) induced MIG-6 expression in lung cancer lines but had little effect in melanoma lines. However, the MIG-6 promoter itself did not appear to be directly affected by either methylation or histone deacetylation, indicating an indirect regulatory mechanism. Luciferase reporter assays revealed that a short segment of exon 1 in the MIG-6 gene is responsible for TSA response in the lung cancer cells; thus, the MIG-6 gene can be epigenetically silenced through an indirect mechanism without having a physical alteration in its promoter. Furthermore, our data also suggest that MIG-6 gene expression is differentially regulated in lung cancer and melanoma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Azacitidina/análogos & derivados , Metilases de Modificação do DNA/antagonistas & inibidores , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Azacitidina/farmacologia , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ilhas de CpG/genética , Primers do DNA/genética , Decitabina , Epigênese Genética/genética , Humanos , Luciferases , Análise em Microsséries , Dados de Sequência Molecular , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Proteínas Supressoras de Tumor/genética
14.
Nat Genet ; 44(6): 690-3, 2012 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-22561520

RESUMO

Opisthorchis viverrini-related cholangiocarcinoma (CCA), a fatal bile duct cancer, is a major public health concern in areas endemic for this parasite. We report here whole-exome sequencing of eight O. viverrini-related tumors and matched normal tissue. We identified and validated 206 somatic mutations in 187 genes using Sanger sequencing and selected 15 genes for mutation prevalence screening in an additional 46 individuals with CCA (cases). In addition to the known cancer-related genes TP53 (mutated in 44.4% of cases), KRAS (16.7%) and SMAD4 (16.7%), we identified somatic mutations in 10 newly implicated genes in 14.8-3.7% of cases. These included inactivating mutations in MLL3 (in 14.8% of cases), ROBO2 (9.3%), RNF43 (9.3%) and PEG3 (5.6%), and activating mutations in the GNAS oncogene (9.3%). These genes have functions that can be broadly grouped into three biological classes: (i) deactivation of histone modifiers, (ii) activation of G protein signaling and (iii) loss of genome stability. This study provides insight into the mutational landscape contributing to O. viverrini-related CCA.


Assuntos
Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/parasitologia , Colangiocarcinoma/genética , Colangiocarcinoma/parasitologia , Fasciolíase/complicações , Adulto , Idoso , Proteínas de Ligação a DNA/genética , Exoma , Feminino , Humanos , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Mutação , Receptores Imunológicos/genética , Análise de Sequência de DNA
15.
Curr Oncol Rep ; 14(3): 240-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22535504

RESUMO

Renal cell carcinoma (RCC) is the most common type of renal cancer in adults. RCC is notoriously resistant to current therapies suggesting the need to improve our knowledge and create more effective therapies. The molecular genetic defects that occur in RCC are extensive and complex ranging from single DNA changes, to large chromosomal defects, to signature disruptions in the transcription of hundreds of genes. These changes are often shared within each histological RCC subtype, illustrating their significance to the disease phenotype. This review presents an overview of the genetic abnormalities that occur within the most common subtypes of RCC. We discuss the recent molecular findings that have advanced our understanding of the somatic architecture of renal tumors and their impact on disease therapeutics.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Mutação/genética , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/terapia , Análise Citogenética , Perfilação da Expressão Gênica , Humanos , Neoplasias Renais/patologia , Neoplasias Renais/terapia , Transcriptoma
16.
Cancer Cell ; 20(4): 511-23, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22014576

RESUMO

Fumarate hydratase (FH) mutation causes hereditary type 2 papillary renal cell carcinoma (PRCC2). The main effect of FH mutation is fumarate accumulation. The current paradigm posits that the main consequence of fumarate accumulation is HIF-α stabilization. Paradoxically, FH mutation differs from other HIF-α stabilizing mutations, such as VHL and SDH mutations, in its associated tumor types. We identified that fumarate can directly up-regulate antioxidant response element (ARE)-controlled genes. We demonstrated that aldo-keto reductase family 1 member B10 (AKR1B10) is an ARE-controlled gene and is up-regulated upon FH knockdown as well as in FH null cell lines. AKR1B10 overexpression is also a prominent feature in both hereditary and sporadic PRCC2. This phenotype better explains the similarities between hereditary and sporadic PRCC2.


Assuntos
Antioxidantes/metabolismo , Carcinoma de Células Renais/genética , Fumarato Hidratase/genética , Neoplasias Renais/genética , Aldeído Redutase/genética , Aldeído Redutase/metabolismo , Aldo-Ceto Redutases , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Fumarato Hidratase/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína 1 Associada a ECH Semelhante a Kelch , Neoplasias Renais/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Fenótipo , RNA Mensageiro/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/fisiologia
17.
Sci Signal ; 4(180): ra44, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21730326

RESUMO

The antimalaria drug chloroquine has been used as an anti-inflammatory agent for treating systemic lupus erythematosus and rheumatoid arthritis. We report that chloroquine promoted the transrepression of proinflammatory cytokines by the glucocorticoid receptor (GR). In a mouse collagen-induced arthritis model, chloroquine enhanced the therapeutic effects of glucocorticoid treatment. By inhibiting lysosome function, chloroquine synergistically activated glucocorticoid signaling. Lysosomal inhibition by either bafilomycin A1 (an inhibitor of the vacuolar adenosine triphosphatase) or knockdown of transcription factor EB (TFEB, a master activator of lysosomal biogenesis) mimicked the effects of chloroquine. The abundance of the GR, as well as that of the androgen receptor and estrogen receptor, correlated with changes in lysosomal biogenesis. Thus, we showed that glucocorticoid signaling is regulated by lysosomes, which provides a mechanistic basis for treating inflammation and autoimmune diseases with a combination of glucocorticoids and lysosomal inhibitors.


Assuntos
Artrite Experimental/tratamento farmacológico , Cloroquina/uso terapêutico , Glucocorticoides/metabolismo , Lisossomos/efeitos dos fármacos , Transdução de Sinais , Animais , Antirreumáticos , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Cloroquina/farmacologia , Citocinas , Glucocorticoides/uso terapêutico , Inflamação , Lisossomos/metabolismo , Lisossomos/fisiologia , Camundongos , Receptores de Glucocorticoides
18.
Cancer Res ; 71(15): 5225-34, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642374

RESUMO

In recent years, several molecularly targeted therapies have been approved for clear cell renal cell carcinoma (ccRCC), a highly aggressive cancer. Although these therapies significantly extend overall survival, nearly all patients with advanced ccRCC eventually succumb to the disease. To identify other molecular targets, we profiled gene expression in 90 ccRCC patient specimens for which tumor grade information was available. Gene set enrichment analysis indicated that cell-cycle-related genes, in particular, Polo-like kinase 1 (PLK1), were associated with disease aggressiveness. We also carried out RNAi screening to identify kinases and phosphatases that when inhibited could prevent cell proliferation. As expected, RNAi-mediated knockdown of PLK1 and other cell-cycle kinases was sufficient to suppress ccRCC cell proliferation. The association of PLK1 in both disease aggression and in vitro growth prompted us to examine the effects of a small-molecule inhibitor of PLK1, BI 2536, in ccRCC cell lines. BI 2536 inhibited the proliferation of ccRCC cell lines at concentrations required to inhibit PLK1 kinase activity, and sustained inhibition of PLK1 by BI 2536 led to dramatic regression of ccRCC xenograft tumors in vivo. Taken together, these findings highlight PLK1 as a rational therapeutic target for ccRCC.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/enzimologia , Proteínas de Ciclo Celular/genética , Perfilação da Expressão Gênica , Neoplasias Renais/enzimologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/genética , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Pteridinas/uso terapêutico , Interferência de RNA , Animais , Antineoplásicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral/efeitos dos fármacos , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/genética , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/fisiologia , Pteridinas/farmacologia , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Quinase 1 Polo-Like
19.
PLoS One ; 6(2): e17165, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21365009

RESUMO

Mitogen-activated protein kinase kinases (MKK or MEK) 1 and 2 are usually treated as redundant kinases. However, in assessing their relative contribution towards ERK-mediated biologic response investigators have relied on tests of necessity, not sufficiency. In response we developed a novel experimental model using lethal toxin (LeTx), an anthrax toxin-derived pan-MKK protease, and genetically engineered protease resistant MKK mutants (MKKcr) to test the sufficiency of MEK signaling in melanoma SK-MEL-28 cells. Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK1cr. Microarray analysis revealed non-overlapping downstream transcriptional targets of MEK1 and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL-28 cells expressing MKK1cr but not MEK2cr. These results indicate in SK-MEL-28 cells MEK1 and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. We conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK1 conditionally compensates for loss of MEK2 only in the presence of other MKK.


Assuntos
Proliferação de Células , MAP Quinase Quinase 2/fisiologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Animais , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Células CHO , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/genética , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Proliferação de Células/efeitos dos fármacos , Análise por Conglomerados , Cricetinae , Cricetulus , Perfilação da Expressão Gênica , Humanos , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Melanoma/genética , Análise em Microsséries , Invasividade Neoplásica , Mutação Puntual/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Cutâneas/genética , Células Tumorais Cultivadas
20.
Am J Pathol ; 178(2): 853-60, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281817

RESUMO

Molecular pathways associated with pathogenesis of sporadic papillary renal cell carcinoma (PRCC), the second most common form of kidney cancer, are poorly understood. We analyzed primary tumor specimens from 35 PRCC patients treated by nephrectomy via gene expression analysis and tissue microarrays constructed from an additional 57 paraffin-embedded PRCC samples via immunohistochemistry. Gene products were validated and further studied by Western blot analyses using primary PRCC tumor samples and established renal cell carcinoma cell lines, and potential associations with pathologic variables and survival in 27 patients with follow-up information were determined. We show that the expression of E2-EPF ubiquitin carrier protein, which targets the principal negative regulator of hypoxia-inducible factor (HIF), von Hippel-Lindau protein, for proteasome-dependent degradation, is markedly elevated in the majority of PRCC tumors exhibiting increased HIF1α expression, and is associated with poor prognosis. In addition, we identified multiple hypoxia-responsive elements within the E2-EPF promoter, and for the first time we demonstrated that E2-EPF is a hypoxia-inducible gene directly regulated via HIF1. These findings reveal deregulation of the oxygen-sensing pathway impinging on the positive feedback mechanism of HIF1-mediated regulation of E2-EPF in PRCC.


Assuntos
Enzimas de Conjugação de Ubiquitina/metabolismo , Sequência de Bases , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/patologia , Hipóxia Celular , Progressão da Doença , Feminino , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Dados de Sequência Molecular , Mutação/genética , Prognóstico , Elementos de Resposta/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...