Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Proteome Res ; 13(6): 2846-55, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24787230

RESUMO

This nonrandomized phase I/II trial assessed the efficacy/tolerability of imatinib plus panitumumab in patients affected by metastatic colorectal cancer (mCRC) after stratification to treatment by selection of activated imatinib drug targets using reverse-phase protein array (RPPA). mCRC patients presenting with a biopsiable liver metastasis were enrolled. Allocation to the experimental and control arms was established using functional pathway activation mapping of c-Kit, PDGFR, and c-Abl phosphorylation by RPPA. The experimental arm received run-in escalation therapy with imatinib followed by panitumumab. The control arm received panitumumab alone. Seven patients were enrolled in the study. For three of the seven patients, sequential pre- and post-treatment biopsies were used to evaluate the effect of the therapeutic compounds on the drug targets and substrates. A decrease in the activation level of the drug targets and downstream substrates was observed in two of three patients. Combination therapy increased the activation of the AKT-mTOR pathway and several receptor tyrosine kinases. This study proposes a novel methodology for stratifying patients to personalized treatment based on the activation level of the drug targets. This workflow provides the ability to monitor changes in the signaling pathways after the administration of targeted therapies and to identify compensatory mechanisms.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Benzamidas/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Piperazinas/farmacologia , Pirimidinas/farmacologia , Adenocarcinoma/secundário , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Análise por Conglomerados , Neoplasias Colorretais/patologia , Estudos de Viabilidade , Humanos , Mesilato de Imatinib , Neoplasias Hepáticas/secundário , Panitumumabe , Seleção de Pacientes , Fosforilação , Projetos Piloto , Piperazinas/uso terapêutico , Medicina de Precisão , Estudos Prospectivos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirimidinas/uso terapêutico , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
J Intern Med ; 274(1): 1-24, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23527888

RESUMO

Systemic therapeutic efficacy is central to determining the outcome of patients with metastatic colorectal cancer (CRC). In these patients, there is a critical need for predictive biomarkers to optimize efficacy whilst minimizing toxicity. The integration of a new generation of molecularly targeted drugs into the treatment of CRC, coupled with the development of sophisticated technologies for individual tumours as well as patient molecular profiling, underlines the potential for personalized medicine. In this review, we focus on the latest progress made within the genomic and proteomic fields, concerning predictive biomarkers for individualized therapy in metastatic CRC.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Terapia de Alvo Molecular/métodos , Medicina de Precisão , Proteômica , Proteínas Proto-Oncogênicas/genética , Animais , Compostos Cromogênicos , Classe I de Fosfatidilinositol 3-Quinases , Neoplasias Colorretais/patologia , Análise Mutacional de DNA , Receptores ErbB/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genômica , Humanos , Hibridização In Situ/métodos , Hibridização in Situ Fluorescente , Comunicação Interdisciplinar , Perda de Heterozigosidade , Terapia de Alvo Molecular/tendências , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Polimorfismo de Nucleotídeo Único , Medicina de Precisão/métodos , Medicina de Precisão/tendências , Valor Preditivo dos Testes , Prognóstico , Proteômica/métodos , Proteômica/tendências , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/genética
3.
Br J Cancer ; 107(6): 947-55, 2012 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-22892392

RESUMO

BACKGROUND: High BCAR4 and ERBB2 mRNA levels in primary breast cancer associate with tamoxifen resistance and poor patient outcome. We determined whether BCAR4 expression sensitises breast cancer cells to lapatinib, and identifies a subgroup of patients who possibly may benefit from ERBB2-targeted therapies despite having tumours with low ERBB2 expression. METHODS: Proliferation assays were applied to determine the effect of BCAR4 expression on lapatinib treatment. Changes in cell signalling were quantified with reverse-phase protein microarrays. Quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) of ERBB2 and BCAR4 was performed in 1418 primary breast cancers. Combined BCAR4 and ERBB2 mRNA levels were evaluated for association with progression-free survival (PFS) in 293 oestrogen receptor-α (ER)-positive patients receiving tamoxifen as first-line monotherapy for recurrent disease. RESULTS: BCAR4 expression strongly sensitised ZR-75-1 and MCF7 breast cancer cells to the combination of lapatinib and antioestrogens. Lapatinib interfered with phosphorylation of ERBB2 and its downstream mediators AKT, FAK, SHC, STAT5, and STAT6. Reverse transcriptase-PCR analysis showed that 27.6% of the breast cancers were positive for BCAR4 and 22% expressed also low levels of ERBB2. The clinical significance of combining BCAR4 and ERBB2 mRNA status was underscored by the finding that the group of patients having BCAR4-positive/ERBB2-low-expressing cancers had a shorter PFS on tamoxifen treatment than the BCAR4-negative group. CONCLUSION: This study shows that BCAR4 expression identifies a subgroup of ER-positive breast cancer patients without overexpression of ERBB2 who have a poor outcome and might benefit from combined ERBB2-targeted and antioestrogen therapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Moduladores de Receptor Estrogênico/uso terapêutico , Terapia de Alvo Molecular/métodos , Quinazolinas/uso terapêutico , RNA não Traduzido/metabolismo , Receptor ErbB-2/metabolismo , Tamoxifeno/uso terapêutico , Adulto , Idoso , Antineoplásicos/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Biomarcadores Tumorais/análise , Neoplasias da Mama/mortalidade , Proliferação de Células , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lapatinib , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/farmacologia , RNA Longo não Codificante , RNA Mensageiro/metabolismo , RNA não Traduzido/efeitos dos fármacos , RNA não Traduzido/genética , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/genética , Receptores de Estrogênio/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
4.
Br J Cancer ; 105(11): 1759-67, 2011 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22045189

RESUMO

BACKGROUND: Recent studies suggest that cancer stem cells (CSCs) mediate chemoresistance, but interestingly, only a small percentage of cells in a resistant tumour are CSCs; this suggests that non-CSCs survive by other means. We hypothesised that chemoresistant colorectal cancer (CRC) cells generate soluble factors that enhance survival of chemonaive tumour cells. METHODS: Chemoresistant CRC cells were generated by serial passage in oxaliplatin (Ox cells). Conditioned media (CM) was collected from parental and oxaliplatin-resistant (OxR) cells. CRC cells were treated with CM and growth and survival were assessed. Tumour growth rates were determined in nude mice after cells were treated with CM. Mass spectrometry (MS) identified proteins in CM. Reverse phase protein microarray assays determined signalling effects of CM in parental cells. RESULTS: Oxaliplatin-resistant CM increased survival of chemo-naive cells. CSC CM also increased growth of parental cells. Parental and OxR mixed tumours grew larger than tumours composed of parental or OxR cells alone. Mass spectrometry detected unique survival-promoting factors in OxR CM compared with parental CM. Cells treated with OxR CM demonstrated early phosphorylation of EGFR and MEK1, with later upregulation of total Akt .We identified progranulin as a potential mediator of chemoresistance. CONCLUSION: Chemoresistant tumour cells and CSCs may promote resistance through soluble factors that mediate survival in otherwise chemosensitive tumour cells.


Assuntos
Efeito Espectador/fisiologia , Neoplasias Colorretais/patologia , Células-Tronco Neoplásicas/patologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Meios de Cultura , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HCT116 , Células HT29 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Masculino , Espectrometria de Massas/métodos , Camundongos , Camundongos Nus , Análise em Microsséries/métodos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Fosforilação , Progranulinas , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Regulação para Cima
5.
J Theor Biol ; 279(1): 29-43, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21439301

RESUMO

In a previous paper we introduced a method called augmented sparse reconstruction (ASR) that identifies links among nodes of ordinary differential equation networks, given a small set of observed trajectories with various initial conditions. The main purpose of that technique was to reconstruct intracellular protein signaling networks. In this paper we show that a recursive augmented sparse reconstruction generates artificial networks that are homologous to a large, reference network, in the sense that kinase inhibition of several reactions in the network alters the trajectories of a sizable number of proteins in comparable ways for reference and reconstructed networks. We show this result using a large in-silico model of the epidermal growth factor receptor (EGF-R) driven signaling cascade to generate the data used in the reconstruction algorithm. The most significant consequence of this observed homology is that a nearly optimal combinatorial dosage of kinase inhibitors can be inferred, for many nodes, from the reconstructed network, a result potentially useful for a variety of applications in personalized medicine.


Assuntos
Proteínas/metabolismo , Transdução de Sinais , Algoritmos , Inibidores de Proteínas Quinases/metabolismo , Padrões de Referência
6.
J Proteome Res ; 9(12): 6678-88, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20945832

RESUMO

Mass spectrometry (MS) is an attractive alternative to quantification of proteins by immunoassays, particularly for protein biomarkers of clinical relevance. Reliable quantification requires that the MS-based assays are robust, selective, and reproducible. Thus, the development of standardized protocols is essential to introduce MS into clinical research laboratories. The aim of this study was to establish a complete workflow for assessing the transferability and reproducibility of selected reaction monitoring (SRM) assays between clinical research laboratories. Four independent laboratories in North America, using identical triple-quadrupole mass spectrometers (Quantum Ultra, Thermo), were provided with standard protocols and instrumentation settings to analyze unknown samples and internal standards in a digested plasma matrix to quantify 51 peptides from 39 human proteins using a multiplexed SRM assay. The interlaboratory coefficient of variation (CV) was less than 10% for 25 of 39 peptides quantified (12 peptides were not quantified based upon hydrophobicity) and exhibited CVs less than 20% for the remaining peptides. In this report, we demonstrate that previously developed research platforms for SRM assays can be improved and optimized for deployment in clinical research environments.


Assuntos
Técnicas de Laboratório Clínico/normas , Espectrometria de Massas/métodos , Peptídeos/análise , Proteínas/análise , Sequência de Aminoácidos , Humanos , Espectrometria de Massas/instrumentação , América do Norte , Peptídeos/normas , Proteínas/normas , Padrões de Referência , Reprodutibilidade dos Testes
7.
Science ; 269(5231): 1721-3, 1995 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-7569900

RESUMO

Activation of early response genes by interferons (IFNs) requires tyrosine phosphorylation of STAT (signal transducers and activators of transcription) proteins. It was found that the serine-threonine kinase mitogen-activated protein kinase (MAPK) [specifically, the 42-kilodalton MAPK or extracellular signal-regulated kinase 2 (ERK2)] interacted with the alpha subunit of IFN-alpha/beta receptor in vitro and in vivo. Treatment of cells with IFN-beta induced tyrosine phosphorylation and activation of MAPK and caused MAPK and Stat1 alpha to coimmunoprecipitate. Furthermore, expression of dominant negative MAPK inhibited IFN-beta-induced transcription. Therefore, MAPK appears to regulate IFN-alpha and IFN-beta activation of early response genes by modifying the Jak-STAT signaling cascade.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Interferon beta/farmacologia , Receptores de Interferon/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Sítios de Ligação , Células Cultivadas , Ativação Enzimática , Humanos , Interferon-alfa/farmacologia , Proteínas de Membrana , Proteína Quinase 1 Ativada por Mitógeno , Fosforilação , Receptor de Interferon alfa e beta , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT1 , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Tirosina/metabolismo
8.
Science ; 261(5129): 1730-3, 1993 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-8378773

RESUMO

Interferon-alpha (IFN-alpha) and IFN-gamma regulate gene expression by tyrosine phosphorylation of several transcription factors that have the 91-kilodalton (p91) protein of interferon-stimulated gene factor-3 (ISGF-3) as a common component. Interferon-activated protein complexes bind enhancers present in the promoters of early response genes such as the high-affinity Fc gamma receptor gene (Fc gamma RI). Treatment of human peripheral blood monocytes or basophils with interleukin-3 (IL-3), IL-5, IL-10, or granulocyte-macrophage colony-stimulating factor (GM-CSF) activated DNA binding proteins that recognized the IFN-gamma response region (GRR) located in the promoter of the Fc gamma RI gene. Although tyrosine phosphorylation was required for the assembly of each of these GRR binding complexes, only those formed as a result of treatment with IFN-gamma or IL-10 contained p91. Instead, complexes activated by IL-3 or GM-CSF contained a tyrosine-phosphorylated protein of 80 kilodaltons. Induction of Fc gamma RI RNA occurred only with IFN-gamma and IL-10, whereas pretreatment of cells with GM-CSF or IL-3 inhibited IFN-gamma induction of Fc gamma RI RNA. Thus, several cytokines other than interferons can activate putative transcription factors by tyrosine phosphorylation.


Assuntos
Citocinas/farmacologia , Proteínas de Ligação a DNA/metabolismo , Monócitos/metabolismo , Transativadores , Fatores de Transcrição/metabolismo , Tirosina/metabolismo , Sequência de Bases , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon gama/farmacologia , Interleucina-10/farmacologia , Interleucina-3/farmacologia , Interleucinas/farmacologia , Dados de Sequência Molecular , Fosforilação , Regiões Promotoras Genéticas , Receptores de IgG/genética , Receptores de IgG/metabolismo , Fator de Transcrição STAT1
9.
J Theor Biol ; 253(3): 616-22, 2008 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-18513748

RESUMO

Until recently, the low-abundance (LA) range of the serum proteome was an unexplored reservoir of diagnostic information. Today it is increasingly appreciated that a diagnostic goldmine of LA biomarkers resides in the blood stream in complexed association with more abundant higher molecular weight carrier proteins such as albumin and immunoglobulins. As we now look to the possibility of harvesting these LA biomarkers more efficiently through engineered nano-scale particles, mathematical approaches are needed in order to reveal the mechanisms by which blood carrier proteins act as molecular 'mops' for LA diagnostic cargo, and the functional relationships between bound LA biomarker concentrations and other variables of interest such as biomarker intravasation and clearance rates and protein half-lives in the bloodstream. Here we show, by simple mathematical modeling, how the relative abundance of large carrier proteins and their longer half-lives in the bloodstream work together to amplify the total blood concentration of these tiny biomarkers. The analysis further suggests that alterations in the production of biomarkers lead to gradual rather than immediate changes in biomarker levels in the blood circulation. The model analysis also points to the characteristics of artificial nano-particles that would render them more efficient harvesters of tumor biomarkers in the circulation, opening up possibilities for the early detection of curable disease, rather than simply better detection of advanced disease.


Assuntos
Biomarcadores/sangue , Proteínas de Transporte/sangue , Modelos Biológicos , Biomarcadores/metabolismo , Meia-Vida , Humanos , Rim/metabolismo , Ligação Proteica , Proteoma
10.
J Theor Biol ; 255(1): 40-52, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18706918

RESUMO

The problem of reconstructing and identifying intracellular protein signaling and biochemical networks is of critical importance in biology. We propose a mathematical approach called augmented sparse reconstruction for the identification of links among nodes of ordinary differential equation (ODE) networks, given a small set of observed trajectories with various initial conditions. As a test case, the method is applied to the epidermal growth factor receptor (EGFR) driven signaling cascade, a well-studied and clinically important signaling network. Our method builds a system of representation from a collection of trajectory integrals, selectively attenuating blocks of terms in the representation. The system of representation is then augmented with random vectors, and l(1) minimization is used to find sparse representations for the dynamical interactions of each node. After showing the performance of our method on a model of the EGFR protein network, we sketch briefly the potential future therapeutic applications of this approach.


Assuntos
Algoritmos , Simulação por Computador , Mapeamento de Interação de Proteínas/métodos , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Humanos , Modelos Biológicos , Ligação Proteica
11.
Mol Cell Biol ; 16(4): 1419-24, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8657115

RESUMO

Previous studies have indicated that the expression of viral oncoproteins, cell transformation, or phorbol ester treatment of cells can inhibit alpha/beta interferon (IFN-alpha/beta)-induced gene expression. The mechanisms by which these promoters of cell growth exert their inhibitory effects vary, but in most instances they involve a disruption of the IFN-alpha/beta-induced transcription complex ISGF3 such that the DNA-binding component of this complex (the 48-kDa ISGF3gamma protein) does not bind to the interferon-stimulated response element (ISRE). In this report, we demonstrated that phorbol ester treatment of human peripheral blood monocytes dramatically inhibits activation of IFN-alpha/B-stimulated early response genes but by a mechanism which does not involve abrogation of the ISRE binding of ISGF3gamma. Phorbol ester treatment of monocytes inhibited IFN alpha-stimulated tyrosine phosphorylation of the transcription factors Stat1alpha, Stat2, and Stat3 and of the tyrosine kinase Tyk2 but had no effect on IFN-gamma activation of Stat1alpha. IFNalpha-stimulated tyrosine phosphorylation of Jak1 and the alpha subunit of the IFN-alpha receptor were unaffected by phorbol 12-myristate 13-acetate (PMA). Moreover, PMA caused the dephosphorylation of Tyk2 but not of Jak1, which was activated by IFN. Pretreatment of cells with vanadate prevented the effects of PMA with regard to PMA-induced Tyk2 dephosphorylation. These observations suggest that PMA exerts its inhibitory effects by activation of a tyrosine phosphatase which selectively regulates Tyk2 but not Jak1 activity.


Assuntos
Interferon-alfa/antagonistas & inibidores , Interferon gama/antagonistas & inibidores , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Sequência de Bases , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Humanos , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Dados de Sequência Molecular , Monócitos/metabolismo , Fosforilação , Receptores de Interferon/metabolismo , Fatores de Transcrição/metabolismo
12.
Mol Cell Biol ; 14(2): 1477-86, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8289823

RESUMO

Members of the interferon regulatory factor (IRF) family of DNA binding transcription factors have roles in growth regulation, antiviral responses, and transcriptional induction of interferon (IFN)-activated early response genes. The IRF family member ISGF3 gamma is the DNA binding component of IFN-stimulated gene factor 3 (ISGF3), a multicomponent complex responsible for the stimulation of IFN-alpha-responsive genes. IFN-alpha-stimulated formation of ISGF3 and subsequent gene expression can be inhibited by phorbol esters or expression of the adenovirus E1A protein. We have investigated IFN signaling in human malignant tumor cell lines of the lung, colon, ovary, cervix, and hematopoietic organs and found some of these cells to be defective for IFN-alpha-induced formation of ISGF3. In many cases, an inhibitory activity termed transcriptional knockout (TKO) correlated with nonresponsiveness. TKO purified from a human papillomavirus-negative cervical carcinoma cell line has a molecular size of 19 kDa. The purified protein interacted with the ISGF3 gamma component of ISGF3, preventing binding of ISGF3 to DNA. Purified TKO displaced ISGF3 from its DNA binding site in vitro and prevented ISGF3 gamma, IRF-1, and IRF-2 from interacting with the IFN-stimulated response element. Partially purified TKO can also directly interact with ISGF3 gamma in the absence of DNA. This protein may be involved with the development of malignancies and the inability of IFN to exert its antiproliferative and antiviral effects.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Fatores de Transcrição/biossíntese , Células Tumorais Cultivadas/metabolismo , Sequência de Bases , Linhagem Celular , Células Cultivadas , Neoplasias do Colo , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon alfa-2 , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Neoplasias Pulmonares , Masculino , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Neoplasias Ovarianas , Proteínas Recombinantes , Pele , Fatores de Transcrição/metabolismo , Neoplasias do Colo do Útero
13.
Mol Cell Biol ; 12(10): 4486-95, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1406637

RESUMO

Phorbol esters activate the expression of a variety of early-response genes through protein kinase C-dependent pathways. In addition, phorbol esters may promote cell growth by the inhibition of expression of cellular gene products regulated by antiproliferative agents such as interferons (IFN)s. In human diploid fibroblasts, phorbol 12-myristate 13-acetate (PMA) selectively inhibits the IFN-alpha-induced cellular gene ISG54. Using transient transfection assays, we have delineated two elements in the promoter of this gene that are necessary for the inhibitory actions of PMA. These elements include (i) the IFN-stimulated response element (ISRE) which is necessary for IFN-alpha-induced cellular gene expression, and (ii) an element located near the site of transcription initiation. IFN-alpha treatment resulted in the rapid induction of ISGF3, a multisubunit transcription factor which binds to the ISRE. PMA caused a substantial reduction in IFN alpha-induced ISGF3 in both nuclear and cytoplasmic extracts, as determined by electrophoretic mobility shift assays with the ISRE as a probe. In vitro reconstitution experiments revealed that IFN-alpha activation of the ISGF3 alpha component of ISGF3 was not affected by PMA. Further experiments were consistent with the possibility that PMA regulated the activity of a cellular factor which competed with ISGF3 gamma for binding of the activated ISGF3 alpha polypeptides. Electrophoretic mobility shift assays using the cap site of ISG54 as a probe demonstrated the formation of a specific complex whose DNA binding activity was not affected by treatment of cells with PMA or IFN-alpha. Competitive inhibition studies were consistent with the DNA-protein complex at the cap site of ISG54 containing proteins with DNA binding sites in common with those which also interact with the ISRE. These data suggest a unique regulatory mechanism by which phorbol esters can modulate IFN signaling.


Assuntos
Interferon-alfa/antagonistas & inibidores , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Sequência de Bases , Linhagem Celular , Colforsina/farmacologia , DNA , Proteínas de Ligação a DNA/metabolismo , Fibroblastos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon-alfa/fisiologia , Dados de Sequência Molecular , Transdução de Sinais/fisiologia , Transfecção
14.
Mol Cell Biol ; 14(3): 2113-8, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7509451

RESUMO

Binding of growth hormone (GH) and erythropoietin (EPO) to their respective receptors results in receptor clustering and activation of tyrosine kinases that initiate a cascade of events resulting not only in the rapid tyrosine phosphorylation of several proteins but also in the induction of early-response genes. In this report, we show that GH and EPO induce the tyrosine phosphorylation of cellular proteins with molecular masses of 93 kDa and of 91 and 84 kDa, respectively, and that these proteins form DNA-binding complexes which recognize an enhancer that has features in common with several rapidly induced genes such as c-fos. Assembly of the protein complexes required tyrosine phosphorylation, which occurred within minutes after addition of ligand. The activated complexes translocated from the cytoplasm to the nucleus. The protein activated by GH is antigenically similar to p91, a protein common to several transcription complexes that are activated by interferons and other cytokines. In contrast, the proteins activated by EPO are distinct from p91. These findings establish the outlines for a cytokine-induced intracellular signaling pathway, which begins with ligand-induced receptor clustering that activates one or more tyrosine kinases. These data are the first to demonstrate that GH- and EPO-activated tyrosine-phosphorylated proteins can specifically recognize a well-defined enhancer and therefore provide a mechanism for rapidly transducing signals from the membrane to the nucleus.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Eritropoetina/fisiologia , Regulação da Expressão Gênica , Hormônio do Crescimento/fisiologia , Fosfoproteínas/metabolismo , Tirosina/análogos & derivados , Sequência de Bases , Linhagem Celular , Desoxirribonucleoproteínas/química , Humanos , Técnicas In Vitro , Dados de Sequência Molecular , Peso Molecular , Oligodesoxirribonucleotídeos/química , Fosfotirosina , Proteínas Tirosina Quinases/fisiologia , RNA Mensageiro/genética , Transdução de Sinais , Tirosina/metabolismo
15.
Mol Cell Biol ; 17(7): 3833-40, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199317

RESUMO

Activation of early response genes by interferons (IFNs) and other cytokines requires tyrosine phosphorylation of a family of transcription factors termed signal transducers and activators of transcription (Stats). The Janus family of tyrosine kinases (Jak1, Jak2, Jak3, and Tyk2) is required for cytokine-induced tyrosine phosphorylation and dimerization of the Stat proteins. In order for IFNs to stimulate maximal expression of Stat1alpha-regulated genes, phosphorylation of a serine residue in the carboxy terminus by mitogen-activated protein kinase (MAPK) is also required. In HeLa cells, both IFN-beta and oncostatin M (OSM) stimulated MAPK and Raf-1 enzyme activity, in addition to Stat1 and Stat3 tyrosine phosphorylation. OSM stimulation of Raf-1 correlated with GTP loading of Ras, whereas IFN-beta activation of Raf-1 was Ras independent. IFN-beta- and OSM-induced Raf-1 activity could be coimmunoprecipitated with either Jak1 or Tyk2. Furthermore, HeLa cells lacking Jak1 displayed no activation of STAT1alpha, STAT3, and Raf-1 by IFN-beta or OSM and also demonstrated no increase in the relative level of GTP-bound p21ras in response to OSM. The requirement for Jak1 for IFN-beta- and OSM-induced activation of Raf-1 was also seen in Jak1-deficient U4A fibrosarcoma cells. Interestingly, basal MAPK, but not Raf-1, activity was constitutively enhanced in Jak1-deficient HeLa cells. Transient expression of Jak1 in both Jak-deficient HeLa cells and U4A cells reconstituted the ability of IFN-beta and OSM to activate Raf-1 and decreased the basal activity of MAPK, while expression of a kinase-inactive form of the protein showed no effect. Moreover, U4A cells selected for stable expression of Jak1, or COS cells transiently expressing Jak1 or Tyk2 but not Jak3, exhibited enhanced Raf-1 activity. Therefore, it appears that Jak1 is required for Raf-1 activation by both IFN-beta and OSM. These results provide evidence for a link between the Jaks and the Raf/MAPK signaling pathways.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Interferon beta/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno , Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Células COS , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Células HeLa , Humanos , Janus Quinase 1 , MAP Quinase Quinase 1 , Oncostatina M , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-raf , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transdução de Sinais , TYK2 Quinase , Transativadores/metabolismo , Células Tumorais Cultivadas
16.
Int J Immunopathol Pharmacol ; 19(3): 517-24, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17026836

RESUMO

Peroxiredoxins (Prx) comprise an extended family of small antioxidant proteins which conserve a thioredoxin-dependent catalytic function that can contribute to cell protection from reactive oxygen species (ROS). ROS generation is one of the deleterious intracellular effects of ionizing radiation, but the role of Prx during radiation treatment has not been extensively explored. Present experiments measure effects of ionizing radiation on expression of human Prx types I (PAGA), II (NKEF-B) and IV (AOE372) in human myeloid leukemia cells (K562). Prx gene transcription was analyzed by amplifying with RT-PCR cDNAs complementary to each Prx-specific coding sequence and by identifying the derived products with Southern blotting procedure. Transcripts of GAPDH were used as the endogenous standard for semi-quantitative comparisons. No consistent increase in Prx gene expression was detected at time intervals up to 72 h after gamma radiation doses that caused cell cycle arrest and nuclear damage (maximum 20 Gy). Immunoblots also were consistent with a prolonged expression or stability of the Prx I/II proteins. Similarly, a cytotoxic concentration of the oxidant hemin, which stimulates rapid hemoglobinization of K562 cells, caused no induction of Prx gene expression. Our results indicate a high Prx stability in human radio-resistant leukemia cells.


Assuntos
Regulação Leucêmica da Expressão Gênica/efeitos da radiação , Leucemia Mieloide/genética , Peroxidases/genética , Humanos , Células K562 , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Estresse Oxidativo , Peroxirredoxinas , Tolerância a Radiação
17.
Cancer Res ; 60(22): 6293-7, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11103786

RESUMO

Annexin I protein expression was evaluated in patient-matched longitudinal study sets of laser capture microdissected normal, premalignant, and invasive epithelium from human esophageal squamous cell cancer and prostatic adenocarcinoma. In 25 esophageal cases (20 by Western blot and 5 by immunohistochemistry) and 17 prostate cases (3 by Western blot and 14 by immunohistochemistry), both tumor types showed either complete loss or a dramatic reduction in the level of annexin I protein expression compared with patient-matched normal epithelium (P < or = 0.05). Moreover, by using Western blot analysis of laser capture microdissected, patient-matched longitudinal study sets of both tumor types, the loss of protein expression occurred in premalignant lesions. Concordance of this result with immunohistochemical analysis suggests that annexin I may be an essential component for maintenance of the normal epithelial phenotype. Additional studies investigating the mechanism(s) and functional consequences of annexin I protein loss in tumor cells are warranted.


Assuntos
Adenocarcinoma/metabolismo , Anexina A1/biossíntese , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias da Próstata/metabolismo , Anexina A1/metabolismo , Western Blotting , Dissecação/métodos , Epitélio/metabolismo , Esôfago/metabolismo , Humanos , Imuno-Histoquímica , Estudos Longitudinais , Masculino , Lesões Pré-Cancerosas/metabolismo , Próstata/metabolismo
18.
Oncogene ; 35(13): 1643-56, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26165839

RESUMO

Resistance to therapies targeting the estrogen pathway remains a challenge in the treatment of estrogen receptor-positive breast cancer. To address this challenge, a systems biology approach was used. A library of small interfering RNAs targeting an estrogen receptor (ER)- and aromatase-centered network identified 46 genes that are dispensable in estrogen-dependent MCF7 cells, but are selectively required for the survival of estrogen-independent MCF7-derived cells and multiple additional estrogen-independent breast cancer cell lines. Integration of this information identified a tumor suppressor gene TOB1 as a critical determinant of estrogen-independent ER-positive breast cell survival. Depletion of TOB1 selectively promoted G1 phase arrest and sensitivity to AKT and mammalian target of rapmycin (mTOR) inhibitors in estrogen-independent cells but not in estrogen-dependent cells. Phosphoproteomic profiles from reverse-phase protein array analysis supported by mRNA profiling identified a significant signaling network reprogramming by TOB1 that differed in estrogen-sensitive and estrogen-resistant cell lines. These data support a novel function for TOB1 in mediating survival of estrogen-independent breast cancers. These studies also provide evidence for combining TOB1 inhibition and AKT/mTOR inhibition as a therapeutic strategy, with potential translational significance for the management of patients with ER-positive breast cancers.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Estrogênios/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Supressoras de Tumor/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células MCF-7 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo
19.
Oncogene ; 35(19): 2529-41, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26387536

RESUMO

Previously, it has been shown that pancreatic ductal adenocarcinoma (PDA) tumors exhibit high levels of hypoxia, characterized by low oxygen pressure (pO2) and decreased O2 intracellular perfusion. Chronic hypoxia is strongly associated with resistance to cytotoxic chemotherapy and chemoradiation in an understudied phenomenon known as hypoxia-induced chemoresistance. The hypoxia-inducible, pro-oncogenic, serine-threonine kinase PIM1 (Proviral Integration site for Moloney murine leukemia virus 1) has emerged as a key regulator of hypoxia-induced chemoresistance in PDA and other cancers. Although its role in therapeutic resistance has been described previously, the molecular mechanism behind PIM1 overexpression in PDA is unknown. Here, we demonstrate that cis-acting AU-rich elements (ARE) present within a 38-base pair region of the PIM1 mRNA 3'-untranslated region mediate a regulatory interaction with the mRNA stability factor HuR (Hu antigen R) in the context of tumor hypoxia. Predominantly expressed in the nucleus in PDA cells, HuR translocates to the cytoplasm in response to hypoxic stress and stabilizes the PIM1 mRNA transcript, resulting in PIM1 protein overexpression. A reverse-phase protein array revealed that HuR-mediated regulation of PIM1 protects cells from hypoxic stress through phosphorylation and inactivation of the apoptotic effector BAD and activation of MEK1/2. Importantly, pharmacological inhibition of HuR by MS-444 inhibits HuR homodimerization and its cytoplasmic translocation, abrogates hypoxia-induced PIM1 overexpression and markedly enhances PDA cell sensitivity to oxaliplatin and 5-fluorouracil under physiologic low oxygen conditions. Taken together, these results support the notion that HuR has prosurvival properties in PDA cells by enabling them with growth advantages in stressful tumor microenvironment niches. Accordingly, these studies provide evidence that therapeutic disruption of HuR's regulation of PIM1 may be a key strategy in breaking an elusive chemotherapeutic resistance mechanism acquired by PDA cells that reside in hypoxic PDA microenvironments.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Proteína Semelhante a ELAV 1/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-pim-1/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular , Fluoruracila/farmacologia , Humanos , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Oxigênio/metabolismo , Proto-Oncogene Mas , RNA Mensageiro/metabolismo
20.
Endocr Relat Cancer ; 12(2): 263-72, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15947101

RESUMO

Metastatic lesions occur in up to 36% of patients with pheochromocytoma. Currently there is no way to reliably detect or predict which patients are at risk for metastatic pheochromocytoma. Thus, the discovery of biomarkers that could distinguish patients with benign disease from those with metastatic disease would be of great clinical value. Using surface-enhanced laser desorption ionization protein chips combined with high-resolution mass spectrometry, we tested the hypothesis that pheochromocytoma pathologic states can be reflected as biomarker information within the low molecular weight (LMW) region of the serum proteome. LMW protein profiles were generated from the serum of 67 pheochromocytoma patients from four institutions and analyzed by two different bioinformatics approaches employing pattern recognition algorithms to determine if the LMW component of the circulatory proteome contains potentially useful discriminatory information. Both approaches were able to identify combinations of LMW molecules which could distinguish all metastatic from all benign pheochromocytomas in a separate blinded validation set. In conclusion, for this study set low molecular mass biomarker information correlated with pheochromocytoma pathologic state using blinded validation. If confirmed in larger validation studies, efforts to identify the underlying diagnostic molecules by sequencing would be warranted. In the future, measurement of these biomarkers could be potentially used to improve the ability to identify patients with metastatic disease.


Assuntos
Neoplasias das Glândulas Suprarrenais/diagnóstico , Biomarcadores Tumorais/sangue , Proteínas de Neoplasias/sangue , Feocromocitoma/diagnóstico , Proteoma/análise , Adolescente , Neoplasias das Glândulas Suprarrenais/patologia , Adulto , Idoso , Criança , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Peso Molecular , Metástase Neoplásica , Feocromocitoma/patologia , Proteômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA