Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 19(9): 1114-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23913124

RESUMO

Although angiogenesis inhibitors have provided substantial clinical benefit as cancer therapeutics, their use is limited by resistance to their therapeutic effects. While ample evidence indicates that such resistance can be influenced by the tumor microenvironment, the underlying mechanisms remain incompletely understood. Here, we have uncovered a paracrine signaling network between the adaptive and innate immune systems that is associated with resistance in multiple tumor models: lymphoma, lung and colon. Tumor-infiltrating T helper type 17 (T(H)17) cells and interleukin-17 (IL-17) induced the expression of granulocyte colony-stimulating factor (G-CSF) through nuclear factor κB (NF-κB) and extracellular-related kinase (ERK) signaling, leading to immature myeloid-cell mobilization and recruitment into the tumor microenvironment. The occurrence of T(H)17 cells and Bv8-positive granulocytes was also observed in clinical tumor specimens. Tumors resistant to treatment with antibodies to VEGF were rendered sensitive in IL-17 receptor (IL-17R)-knockout hosts deficient in T(H)17 effector function. Furthermore, pharmacological blockade of T(H)17 cell function sensitized resistant tumors to therapy with antibodies to VEGF. These findings indicate that IL-17 promotes tumor resistance to VEGF inhibition, suggesting that immunomodulatory strategies could improve the efficacy of anti-angiogenic therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Resistencia a Medicamentos Antineoplásicos , Interleucina-17/metabolismo , Neoplasias/tratamento farmacológico , Neovascularização Patológica/imunologia , Células Th17/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos/imunologia , Antígeno CD11b/metabolismo , Neoplasias Colorretais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/metabolismo , Hormônios Gastrointestinais/metabolismo , Fator Estimulador de Colônias de Granulócitos/biossíntese , Granulócitos/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Linfoma/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , NF-kappa B/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Neuropeptídeos/metabolismo , Comunicação Parácrina , Microambiente Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
2.
Clin Cancer Res ; 19(13): 3681-92, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23685835

RESUMO

PURPOSE: The aim of this study was to identify conserved pharmacodynamic and potential predictive biomarkers of response to anti-VEGF therapy using gene expression profiling in preclinical tumor models and in patients. EXPERIMENTAL DESIGN: Surrogate markers of VEGF inhibition [VEGF-dependent genes or VEGF-dependent vasculature (VDV)] were identified by profiling gene expression changes induced in response to VEGF blockade in preclinical tumor models and in human biopsies from patients treated with anti-VEGF monoclonal antibodies. The potential value of VDV genes as candidate predictive biomarkers was tested by correlating high or low VDV gene expression levels in pretreatment clinical samples with the subsequent clinical efficacy of bevacizumab (anti-VEGF)-containing therapy. RESULTS: We show that VDV genes, including direct and more distal VEGF downstream endothelial targets, enable detection of VEGF signaling inhibition in mouse tumor models and human tumor biopsies. Retrospective analyses of clinical trial data indicate that patients with higher VDV expression in pretreatment tumor samples exhibited improved clinical outcome when treated with bevacizumab-containing therapies. CONCLUSIONS: In this work, we identified surrogate markers (VDV genes) for in vivo VEGF signaling in tumors and showed clinical data supporting a correlation between pretreatment VEGF bioactivity and the subsequent efficacy of anti-VEGF therapy. We propose that VDV genes are candidate biomarkers with the potential to aid the selection of novel indications as well as patients likely to respond to anti-VEGF therapy. The data presented here define a diagnostic biomarker hypothesis based on translational research that warrants further evaluation in additional retrospective and prospective trials.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/farmacologia , Bevacizumab , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neoplasias/genética , Neoplasias/mortalidade , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(15): 6079-84, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23530240

RESUMO

Granulocyte-colony stimulating factor (G-CSF) promotes mobilization of CD11b(+)Gr1(+) myeloid cells and has been implicated in resistance to anti-VEGF therapy in mouse models. High G-CSF production has been associated with a poor prognosis in cancer patients. Here we show that activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. Several growth factors induced G-CSF expression by a MEK-dependent mechanism. Inhibition of G-CSF release with a MEK inhibitor markedly reduced G-CSF production in vitro and synergized with anti-VEGF antibodies to reduce CD11b(+)Ly6G(+) neutrophil mobilization and tumor growth and led to increased survival in animal models of cancer, including a genetically engineered mouse model of pancreatic adenocarcinoma. Analysis of biopsies from pancreatic cancer patients revealed increased phospho-MEK, G-CSF, and Ets expression and enhanced neutrophil recruitment compared with normal pancreata. These results provide insights into G-CSF regulation and on the mechanism of action of MEK inhibitors and point to unique anticancer strategies.


Assuntos
Fator Estimulador de Colônias de Granulócitos/metabolismo , Sistema de Sinalização das MAP Quinases , Neutrófilos/citologia , Proteína Proto-Oncogênica c-ets-2/metabolismo , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Neoplasias/metabolismo , Neovascularização Patológica , Infiltração de Neutrófilos , Proteínas Tirosina Quinases/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
J Pathol ; 227(4): 404-16, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22611017

RESUMO

Inhibiting angiogenesis has become an important therapeutic strategy for cancer treatment but, like other current targeted therapies, benefits experienced for late-stage cancers can be curtailed by inherent refractoriness or by acquired drug resistance, requiring a need for better mechanistic understanding of such effects. Numerous preclinical studies have demonstrated that VEGF pathway inhibitors suppress primary tumour growth and metastasis. However, it has been recently reported that short-term VEGF and VEGFR inhibition can paradoxically accelerate tumour invasiveness and metastasis in certain models. Here we comprehensively compare the effects of both antibody and small molecule receptor tyrosine kinase (RTK) inhibitors targeting the VEGF-VEGFR pathway, using short-term therapy in various mouse models of metastasis. Our findings demonstrate that antibody inhibition of VEGF pathway molecules does not promote metastasis, in contrast to selected small molecule RTK inhibitors at elevated-therapeutic drug dosages. In particular, a multi-targeted RTK inhibitor, sunitinib, which most profoundly potentiated metastasis, also increased lung vascular permeability and promoted tumour cell extravasation. Mechanistically, sunitinib, but not anti-VEGF treatment, attenuated endothelial barrier function in culture and caused a global inhibition of protein tyrosine phosphorylation, including molecules important for maintaining endothelial cell-cell junctions. Together these findings indicate that, rather than a specific consequence of inhibiting the VEGF signalling pathway, pharmacological inhibitors of the VEGF pathway can have dose- and drug class-dependent side-effects on the host vasculature. These findings also advocate for the continued identification of mechanisms of resistance to anti-angiogenics and for therapy development to overcome it.


Assuntos
Inibidores da Angiogênese/classificação , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/classificação , Antineoplásicos/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Benzamidas , Benzenossulfonatos/uso terapêutico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/uso terapêutico , Feminino , Mesilato de Imatinib , Técnicas In Vitro , Indóis/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Niacinamida/análogos & derivados , Compostos de Fenilureia , Piperazinas/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/imunologia , Sorafenibe , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia
5.
J Immunol ; 187(3): 1289-97, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21724995

RESUMO

In TNF-treated cells, TNFR1, TNFR-associated death domain protein (TRADD), Fas-associated death domain protein, and receptor-interacting protein kinase proteins form the signaling complex via modular interaction within their C-terminal death domains. In this paper, we report that the death domain SXXE/D motifs (i.e., S381DHE motif of TNFR1-death domain as well as S215LKD and S296LAE motifs of TRADD-death domain) are phosphorylated, and this is required for stable TNFR1-TRADD complex formation and subsequent activation of NF-κB. Phospho-S215LKD and phospho-S296LAE motifs are also critical to TRADD for recruiting Fas-associated death domain protein and receptor-interacting protein kinase. IκB kinase ß plays a critical role in TNFR1 phosphorylation of S381, which leads to subsequent T cell migration and accumulation. Consistently, we observed in inflammatory bowel disease specimens that TNFR1 was constitutively phosphorylated on S381 in those inflammatory T cells, which had accumulated in high numbers in the inflamed mucosa. Therefore, SXXE/D motifs found in the cytoplasmic domains of many TNFR family members and their adaptor proteins may serve to function as a specific interaction module for the α-helical death domain signal transduction.


Assuntos
Movimento Celular/imunologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/imunologia , Fosfoproteínas/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Subpopulações de Linfócitos T/imunologia , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Motivos de Aminoácidos/imunologia , Sequência de Aminoácidos , Animais , Epitopos de Linfócito T/imunologia , Células HEK293 , Humanos , Mediadores da Inflamação/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Células Jurkat , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fosfoproteínas/fisiologia , Fosforilação/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Proteína de Domínio de Morte Associada a Receptor de TNF/fisiologia
6.
Annu Rev Cell Dev Biol ; 27: 563-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21756109

RESUMO

The formation of the vascular network is an intricate and complex process that is an obligate requirement during vertebrate development. The cardiovascular system is the first organ to develop and reach a functional state, which underscores the crucial role of the vasculature in the developing embryo. The development of the vasculature into highly branched conduits needs to occur in numerous sites and in precise patterns to supply oxygen and nutrients to the rapidly expanding tissue of the embryo. This process is mediated by the coordinated response of vascular endothelial and mural cells to the heterogeneous angiogenic cues provided by tissues and organs, whereas aberrant regulation and coordination of angiogenic signals during development result in lethality, impaired organ development, or disease states. This article reviews the essential signaling pathways required for establishment of the vertebrate vasculature with a major focus on a key regulatory factor, vascular endothelial growth factor (VEGF). We also discuss current knowledge of physiological angiogenic processes as well as their disruptions in pathological processes, particularly tumorigenesis.


Assuntos
Sistema Cardiovascular , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Osso e Ossos/irrigação sanguínea , Osso e Ossos/fisiologia , Sistema Cardiovascular/anatomia & histologia , Sistema Cardiovascular/crescimento & desenvolvimento , Sistema Cardiovascular/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Reprodução , Cicatrização
7.
Nat Rev Cancer ; 10(7): 505-14, 2010 07.
Artigo em Inglês | MEDLINE | ID: mdl-20574450

RESUMO

The cardiovascular system ensures the delivery of nutrients, oxygen, and blood and immune cells to all organs and tissues: it is also responsible for the removal of waste metabolites. The vascular system develops and matures through two tightly regulated processes: vasculogenesis and angiogenesis. Angiogenesis is active only under specific physiological conditions in healthy adults but the vasculature can be aberrantly activated to generate new blood vessels during pathological conditions such as cancer and chronic inflammation. In this Opinion article we discuss the parallels and differences in the angiogenic process under either a physiological or a pathological state, especially tumorigenesis.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica , Animais , Humanos , Neoplasias/patologia
8.
Clin Cancer Res ; 16(3): 775-7, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103657

RESUMO

Although most cancer therapies are directed against tumor cells, an emerging area of cancer therapeutics focuses on targeting cells of the tumor microenvironment. Inhibiting the Src family kinase with dasatinib decreases tumor growth through inhibiting growth of tumor-associated endothelial and myeloid cells.


Assuntos
Proteínas de Fusão bcr-abl/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Células Estromais/efeitos dos fármacos , Tiazóis/farmacologia , Quinases da Família src/antagonistas & inibidores , Animais , Dasatinibe , Células Endoteliais/efeitos dos fármacos , Masculino , Camundongos , Células Mieloides/efeitos dos fármacos , Neoplasias/enzimologia , Neoplasias/patologia
9.
Methods Mol Biol ; 527: 247-55, ix, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19241018

RESUMO

Protein tyrosine phosphorylation plays a central role in cell-signaling and is a focus of biomedical studies and cancer therapy. However, it is still challenging to identify or characterize the coordinated changes of many candidate proteins of one particular pathway or multiple pathways simultaneously. Antibody array is a recently developed approach applied for differential analysis of multiple protein posttranslational modification events in mammalian cells. It is based on the highly specific recognition between the immobilized antibodies on the array and their specific target proteins in a high-throughput screening format. Here we have described in detail two methods for differential analysis of protein tyrosine phosphorylation in cells by (1) using a single fluorescent protein capture format on membrane array and (2) a competitive protein capture method on glass surface array.


Assuntos
Anticorpos Imobilizados/farmacologia , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Análise Serial de Proteínas/métodos , Proteínas Tirosina Quinases/metabolismo , Animais , Anticorpos Fosfo-Específicos/farmacologia , Células Eucarióticas/química , Células Eucarióticas/metabolismo , Humanos , Mamíferos/metabolismo , Fosforilação , Análise Serial de Proteínas/instrumentação , Proteômica/métodos
10.
Mol Cell Biol ; 25(11): 4716-26, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15899873

RESUMO

Ankyrin repeat and SOCS box (ASB) family members have a C-terminal SOCS box and an N-terminal ankyrin-related sequence of variable repeats belonging to the SOCS superfamily. While SH2-domain-bearing SOCS proteins are mainly involved in the negative feedback regulation of the protein tyrosine kinase-STAT pathway in response to a variety of cytokines, the roles of ASB family members remain largely unknown. To investigate ASB functions, we screened for ASB3-interacting factors by using antibody array technology and identified tumor necrosis factor receptor II (TNF-R2) as an ASB3 binding target. ASB3 expression and activities are required for (i) TNF-R2 ubiquitination both in vivo and in vitro, (ii) TNF-R2 proteolysis via the proteasome pathway, and (iii) the inhibition of TNF-R2-mediated Jun N-terminal protein kinase (JNK) activation. While the ankyrin repeats of ASB3 interact with the C-terminal 37 amino acids of TNF-R2, the SOCS box of ASB3 is responsible for recruiting the E3 ubiquitin ligase adaptors Elongins-B/C, leading to TNF-R2 ubiquitination on multiple lysine residues within its C-terminal region. Downregulation of ASB3 expression by a small interfering RNA inhibited TNF-R2 degradation and potentiated TNF-R2-mediated cytotoxicity. The data presented here implicate ASB3 as a negative regulator of TNF-R2-mediated cellular responses to TNF-alpha by direct targeting of TNF-R2 for ubiquitination and proteasome-mediated degradation.


Assuntos
Proteínas de Transporte/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Repetição de Anquirina , Apoptose , Proteínas de Transporte/genética , Linhagem Celular , Regulação para Baixo , Elonguina , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lisina/metabolismo , Dados de Sequência Molecular , Interferência de RNA , Receptores Tipo II do Fator de Necrose Tumoral/genética , Proteínas Supressoras da Sinalização de Citocina , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/agonistas , Fator de Necrose Tumoral alfa/fisiologia
11.
Mol Cell Proteomics ; 3(8): 788-95, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15123764

RESUMO

Previously, we demonstrated that antibodies printed on a solid support were able to detect protein-protein interaction in mammalian cells. Here we further developed the antibody array system for detecting proteins with various post-translational modifications in mammalian cells. In this novel approach, immunoprecipitated proteins were labeled with fluorescent dye followed by incubation over antibody arrays. Targeted proteins, captured by the antibodies immobilized on PVDF membrane or glass slide, were detected by means of near infrared fluorescent scanner or fluorescent microscopy. To demonstrate the application of the antibody arrays in protein post-translational modifications, we profiled protein tyrosine phosphorylation, ubiquitination, and acetylation in mammalian cells under different conditions. Our results indicate that antibody array technology can provide a powerful means of profiling a large number of proteins with different post-translational modifications in cells.


Assuntos
Anticorpos , Análise Serial de Proteínas , Processamento de Proteína Pós-Traducional , Tirosina/metabolismo , Ubiquitina/metabolismo , Células 3T3 , Acetilação , Animais , Linhagem Celular , Imunofluorescência , Corantes Fluorescentes , Perfilação da Expressão Gênica , Células HeLa , Humanos , Camundongos , Fosforilação
12.
J Biol Chem ; 277(49): 47572-80, 2002 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-12270932

RESUMO

Signal transducer and activator of transcription (STAT) proteins are both tyrosine- and serine-phosphorylated, mediating signal transduction and gene regulation. Following gene regulation, STAT activity in the nucleus is then terminated by a nuclear protein phosphatase(s), which remains unidentified. Using novel antibody arrays to screen the Stat1-specific protein phosphatase(s), we identified a SHP-2-Stat1 interaction in the A431 cell nucleus. SHP-2 and Stat1 nuclear localization and their association in response to either epidermal growth factor or interferon-gamma (IFNgamma) were confirmed by immunofluorescent staining and affinity precipitation assays. The SHP-2 C-terminal region containing protein-tyrosine phosphatase activity interacted with the C-terminal SH2 transcriptional activation domain of Stat1. In SHP-2-/- mouse fibroblast cells, Stat1 phosphorylation at both the tyrosine residue Tyr(701) and the serine residue Ser(727) by IFNgamma was enhanced and prolonged. Consistently, purified GST-SHP-2 dephosphorylated Stat1 at both tyrosine and serine residues when immunoprecipitated phospho-Stat1 or a peptide corresponding to the sequence surrounding Tyr(P)(701) or Ser(P)(727) of Stat1 was used as the substrate. Overexpression of SHP-2 in 293T cells inhibited IFNgamma-dependent Stat1 phosphorylation and suppressed Stat1-dependent induction of luciferase activity. Our findings demonstrate that SHP-2 is a dual-specificity protein phosphatase involved in Stat1 dephosphorylation at both tyrosine and serine residues and plays an important role in modulating STAT function in gene regulation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Serina/química , Transativadores/metabolismo , Tirosina/química , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , DNA/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Glutationa Transferase/metabolismo , Humanos , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Luciferases/metabolismo , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Fosforilação , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Fator de Transcrição STAT1 , Homologia de Sequência de Aminoácidos , Serina/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas , Tirosina/metabolismo
13.
J Biol Chem ; 277(34): 30716-23, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12070153

RESUMO

The cytoplasmic region of human epidermal growth factor receptor (EGFR) contains an intrinsic tyrosine kinase (697-955) followed by a 231-residue-long COOH-terminal tail (C-tail), which contains multiple tyrosine residues. To examine the role of the EGFR C-tail in signal transducer and activator of transcription (STAT) activation, a series of EGFR C-tail truncations were constructed. Transient transfection of 293 cells with EGFR lacking the C-tail, i.e. Y974DeltaEGFR or Y992DeltaEGFR, led to EGF-independent or constitutive STAT activation, whereas EGF-dependent STAT activation was restored with truncations made COOH-terminal to the next tyrosine residue, i.e. EGFR-Y1045Delta. Transfection with the-truncated form EGFR-Y954Delta resulted in the loss of STAT activation, suggesting that the sequence between Tyr(974) and Tyr(954) is essential for STAT activation. Phosphopeptide competition analysis revealed multiple tyrosine residues within the C-tail that can act as the docking sites for both Stat1 and Stat3. A region that negatively regulated STAT activation was also identified, extending from Tyr(1114) to Glu(1172), consistent with the ability of this region to recruit a suppressor of cytokine signaling factors SOCS1 and SOCS3. When cotransfected with the full-length EGFR, but not Y992DeltaEGFR, SOCS1 or SOCS3 inhibited STAT activation by EGF in 293 cells. This suggests that both SOCS1 and SOCS3 can negatively regulate EGFR activation, presumably by inducing ubiquitination-dependent EGFR degradation upon ligand binding. These findings may therefore offer clues to how the EGF receptor C-tail regulates STAT activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Receptores ErbB/química , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Repressoras , Transdução de Sinais , Transativadores/metabolismo , Fatores de Transcrição , Sítios de Ligação , Proteínas de Transporte/fisiologia , Receptores ErbB/fisiologia , Humanos , Proteínas/fisiologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...