Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Oncotarget ; 6(11): 8851-74, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25860930

RESUMO

N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that plays a key role in regulating signaling pathways involved in mediating cancer cell invasion and migration, including those derived from prostate, colon, etc. However, the mechanisms and molecular targets through which NDRG1 reduces cancer cell invasion and migration, leading to inhibition of cancer metastasis, are not fully elucidated. In this investigation, using NDRG1 over-expression models in three tumor cell-types (namely, DU145, PC3MM and HT29) and also NDRG1 silencing in DU145 and HT29 cells, we reveal that NDRG1 decreases phosphorylation of a key proto-oncogene, cellular Src (c-Src), at a well-characterized activating site (Tyr416). NDRG1-mediated down-regulation of EGFR expression and activation were responsible for the decreased phosphorylation of c-Src (Tyr416). Indeed, NDRG1 prevented recruitment of c-Src to EGFR and c-Src activation. Moreover, NDRG1 suppressed Rac1 activity by modulating phosphorylation of a c-Src downstream effector, p130Cas, and its association with CrkII, which acts as a "molecular switch" to activate Rac1. NDRG1 also affected another signaling molecule involved in modulating Rac1 signaling, c-Abl, which then inhibited CrkII phosphorylation. Silencing NDRG1 increased cell migration relative to the control and inhibition of c-Src signaling using siRNA, or a pharmacological inhibitor (SU6656), prevented this increase. Hence, the role of NDRG1 in decreasing cell migration is, in part, due to its inhibition of c-Src activation. In addition, novel pharmacological agents, which induce NDRG1 expression and are currently under development as anti-metastatic agents, markedly increase NDRG1 and decrease c-Src activation. This study leads to important insights into the mechanism involved in inhibiting metastasis by NDRG1 and how to target these pathways with novel therapeutics.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Transdução de Sinais/fisiologia , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Colo/patologia , Proteína Substrato Associada a Crk/fisiologia , Regulação para Baixo , Ativação Enzimática/fisiologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes src , Humanos , Indóis , Masculino , Fosforilação , Neoplasias da Próstata/patologia , Processamento de Proteína Pós-Traducional , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-abl/fisiologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Recombinantes/metabolismo , Sulfonamidas , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia
2.
Cancer Sci ; 106(6): 709-717, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25816892

RESUMO

We have previously reported that an adaptor protein CRK, including CRK-I and CRK-II, plays essential roles in the malignant potential of various aggressive human cancers, suggesting the validity of targeting CRK in molecular targeted therapy of a wide range of cancers. Nevertheless, the role of CRK in human bladder cancer with marked invasion, characterized by distant metastasis and poor prognosis, remains obscure. In the present study, immunohistochemistry indicated a striking enhancement of CRK-I/-II, but not CRK-like, in human bladder cancer tissues compared to normal urothelium. We established CRK-knockdown bladder cancer cells using 5637 and UM-UC-3, which showed a significant decline in cell migration, invasion, and proliferation. It is noteworthy that an elimination of CRK conferred suppressed phosphorylation of c-Met and the downstream scaffold protein Gab1 in a hepatocyte growth factor-dependent and -independent manner. In epithelial-mesenchymal transition-related molecules, E-cadherin was upregulated by CRK elimination, whereas N-cadherin, vimentin, and Zeb1 were downregulated. A similar effect was observed following treatment with c-Met inhibitor SU11274. Depletion of CRK significantly decreased cell proliferation of 5637 and UM-UC-3, consistent with reduced activity of ERK. An orthotopic xenograft model with bioluminescent imaging revealed that CRK knockdown significantly attenuated not only tumor volume but also the number of circulating tumor cells, resulted in a complete abrogation of metastasis. Taken together, this evidence uncovered essential roles of CRK in invasive bladder cancer through the hepatocyte growth factor/c-Met/CRK feedback loop for epithelial-mesenchymal transition induction. Thus, CRK might be a potent molecular target in bladder cancer, particularly for preventing metastasis, leading to the resolution of clinically longstanding critical issues.


Assuntos
Transição Epitelial-Mesenquimal , Fator de Crescimento de Hepatócito/fisiologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Neoplasias da Bexiga Urinária/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Células Neoplásicas Circulantes , Fosforilação , Proteínas Proto-Oncogênicas c-crk/análise
3.
J Clin Invest ; 125(3): 1019-32, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25621495

RESUMO

Effector T cell migration into inflamed sites greatly exacerbates tissue destruction and disease severity in inflammatory diseases, including graft-versus-host disease (GVHD). T cell migration into such sites depends heavily on regulated adhesion and migration, but the signaling pathways that coordinate these functions downstream of chemokine receptors are largely unknown. Using conditional knockout mice, we found that T cells lacking the adaptor proteins CRK and CRK-like (CRKL) exhibit reduced integrin-dependent adhesion, chemotaxis, and diapedesis. Moreover, these two closely related proteins exhibited substantial functional redundancy, as ectopic expression of either protein rescued defects in T cells lacking both CRK and CRKL. We determined that CRK proteins coordinate with the RAP guanine nucleotide exchange factor C3G and the adhesion docking molecule CASL to activate the integrin regulatory GTPase RAP1. CRK proteins were required for effector T cell trafficking into sites of inflammation, but not for migration to lymphoid organs. In a murine bone marrow transplantation model, the differential migration of CRK/CRKL-deficient T cells resulted in efficient graft-versus-leukemia responses with minimal GVHD. Together, the results from our studies show that CRK family proteins selectively regulate T cell adhesion and migration at effector sites and suggest that these proteins have potential as therapeutic targets for preventing GVHD.


Assuntos
Quimiotaxia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Linfócitos T/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Transplante de Medula Óssea , Adesão Celular , Polaridade Celular , Células Cultivadas , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/fisiologia , Transdução de Sinais , Linfócitos T/transplante , Migração Transendotelial e Transepitelial , Proteínas rac1 de Ligação ao GTP/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(34): 12544-9, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25099352

RESUMO

Nervous system injury or disease leads to activation of glia, which govern postinjury responses in the nervous system. Axonal injury in Drosophila results in transcriptional up-regulation of the glial engulfment receptor Draper; there is extension of glial membranes to the injury site (termed activation), and then axonal debris is internalized and degraded. Loss of the small GTPase Rac1 from glia completely suppresses glial responses to injury, but upstream activators remain poorly defined. Loss of the Rac guanine nucleotide exchange factor (GEF) Crk/myoblast city (Mbc)/dCed-12 has no effect on glial activation, but blocks internalization and degradation of debris. Here we show that the signaling molecules downstream of receptor kinase (DRK) and daughter of sevenless (DOS) (mammalian homologs, Grb2 and Gab2, respectively) and the GEF son of sevenless (SOS) (mammalian homolog, mSOS) are required for efficient activation of glia after axotomy and internalization/degradation of axonal debris. At the earliest steps of glial activation, DRK/DOS/SOS function in a partially redundant manner with Crk/Mbc/dCed-12, with blockade of both complexes strongly suppressing all glial responses, similar to loss of Rac1. This work identifies DRK/DOS/SOS as the upstream Rac GEF complex required for glial responses to axonal injury, and demonstrates a critical requirement for multiple GEFs in efficient glial activation after injury and internalization/degradation of axonal debris.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/citologia , Drosophila melanogaster/fisiologia , Proteínas do Olho/fisiologia , Neuroglia/fisiologia , Proteína Son Of Sevenless de Drosófila/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Animais Geneticamente Modificados , Axônios/fisiologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas do Olho/genética , Genes de Insetos , Mutação , Degeneração Neural , Fagossomos/fisiologia , Proteínas Proto-Oncogênicas c-crk/genética , Proteínas Proto-Oncogênicas c-crk/fisiologia , Proteína Son Of Sevenless de Drosófila/genética , Proteínas rac de Ligação ao GTP/genética , Proteínas ras/genética , Proteínas ras/fisiologia
5.
Nat Rev Cancer ; 10(12): 858-70, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21102636

RESUMO

Current evidence highlights the ability of adaptor (or scaffold) proteins to create signalling platforms that drive cellular transformation upon integrin-dependent adhesion and growth factor receptor activation. The understanding of the biological effects that are regulated by these adaptors in tumours might be crucial for the identification of new targets and the development of innovative therapeutic strategies for human cancer. In this Review we discuss the relevance of adaptor proteins in signalling that originates from integrin-mediated cell-extracellular matrix (ECM) adhesion and growth factor stimulation in the context of cell transformation and tumour progression. We specifically underline the contribution of p130 Crk-associated substrate (p130CAS; also known as BCAR1), neural precursor cell expressed, developmentally down-regulated 9 (NEDD9; also known as HEF1), CRK and the integrin-linked kinase (ILK)-pinch-parvin (IPP) complex to cancer, along with the more recently identified p140 Cas-associated protein (p140CAP; also known as SRCIN1).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Integrinas/fisiologia , Neoplasias/etiologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Apoptose , Movimento Celular , Humanos , Invasividade Neoplásica , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Receptor ErbB-2/fisiologia , Fator de Crescimento Transformador beta/fisiologia
6.
Oncogene ; 29(48): 6378-89, 2010 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-20729917

RESUMO

The v-Crk oncogene product consists of two protein interaction modules, a Src homology 2 (SH2) domain and a Src homology 3 (SH3) domain. Overexpression of CrkI, the cellular homolog of v-Crk, transforms mouse fibroblasts, and elevated CrkI expression is observed in several human cancers. The SH2 and SH3 domains of Crk are required for transformation, but the identity of the critical cellular binding partners is not known. A number of candidate Crk SH3-binding proteins have been identified, including the nonreceptor tyrosine kinases c-Abl and Arg, and the guanine nucleotide exchange proteins C3G, SOS1 and DOCK180. The aim of this study is to determine which of these are required for transformation by CrkI. We found that short hairpin RNA-mediated knockdown of C3G or SOS1 suppressed anchorage-independent growth of NIH-3T3 cells overexpressing CrkI, whereas knockdown of SOS1 alone was sufficient to suppress tumor formation by these cells in nude mice. Knockdown of C3G was sufficient to revert morphological changes induced by CrkI expression. By contrast, knockdown of Abl family kinases or their inhibition with imatinib enhanced anchorage-independent growth and tumorigenesis induced by Crk. These results show that SOS1 is essential for CrkI-induced fibroblast transformation, and also reveal a surprising negative role for Abl kinases in Crk transformation.


Assuntos
Transformação Celular Neoplásica , Proteínas Proto-Oncogênicas c-crk/fisiologia , Domínios de Homologia de src , Animais , Apoptose , Fator 2 de Liberação do Nucleotídeo Guanina/fisiologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/fisiologia , Proteínas Proto-Oncogênicas c-crk/química , Proteína SOS1/fisiologia , Transdução de Sinais
7.
Tumour Biol ; 31(1): 59-67, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20237902

RESUMO

Small GTPases, particularly the Rho family, are key regulators of cell motility and migration. Dock180 was well known for the main target of signal adaptor protein Crk and acted as a guanine-nucleotide exchange factor for small GTPase Rac1. In the present study, Dock180 was found to combine primarily with CrkI other than CrkII, and its association with Elmo1 was also demonstrated in ovarian cancer cell SKOV3. To evaluate the role of Dock180 in human ovarian cancer cell, we performed RNAi-mediated knockdown of Dock180 in SKOV3 cells using small interfering RNA expression vector. In Dock180 knockdown cells, we found that Elmo1 expression and Rac1 activity were decreased simultaneously. By contrast, the expressions of both another Crk-combining molecule C3G and Rap1 activity were observed to increase obviously. Accordingly, all Dock180 knockdown cells present with evident change in cell morphology, reduced cell proliferation, and attenuated cell migration. Taken together, these results suggest that signal transfer of Crk/Dock180/Rac1 is implicated in actin cytoskeleton reorganization and thus in the cell proliferation, motility, invasion, and of human ovarian cancer cell line SKOV3.


Assuntos
Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Transdução de Sinais/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Complexo Shelterina , Proteínas de Ligação a Telômeros/fisiologia
8.
Angiogenesis ; 12(4): 339-54, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19760510

RESUMO

Intermittent Hypoxia (IH) that develops in neovascularized solid tumours has been described to positively influence the tumour growth by modulating the behaviour of cancer cells as well as of endothelial cells. However, the molecular mechanisms regulated by IH still remain poorly understood. In this work, the effects of IH were investigated on endothelial cells by a proteomic approach. Protein abundance variations were studied using fluorescent 2D-Differential in Gel Electrophoresis (2D-DIGE). Amongst the proteins of which the abundance varied under IH, NDRG1 and CRK-I/II were identified by mass spectrometry. These proteins have already been described to influence cancer cell migration as well as the angiogenic processes in solid tumours. Since an increase in endothelial cell migration under IH was evidenced in our previous work, the involvement of NDRG1 and CRK-I/II proteins in endothelial cell migration under IH was determined by silencing the expression of both proteins using siRNA. The results revealed that NDRG1 and CRK-I/II are indeed regulators of endothelial cell migration under intermittent hypoxia: silencing of CRK-I/II resulted in an increase in endothelial cell migration, whereas the invalidation of NDRG1 decreased it. These results give news insight regarding the effects of IH on endothelial cell migration and hence on neoangiogenesis.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Hipóxia Celular/fisiologia , Células Endoteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Transformada/efeitos dos fármacos , Linhagem Celular Transformada/metabolismo , Movimento Celular , Esquema de Medicação , Eletroforese em Gel Bidimensional , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Humanos , Células Híbridas/citologia , Células Híbridas/efeitos dos fármacos , Células Híbridas/metabolismo , Processamento de Imagem Assistida por Computador , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Espectrometria de Massas , Oxigênio/administração & dosagem , Oxigênio/farmacologia , Reação em Cadeia da Polimerase/métodos , Proteômica , Proteínas Proto-Oncogênicas c-crk/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-crk/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia
11.
Cell Microbiol ; 9(10): 2497-516, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17848169

RESUMO

The intracellular bacterial pathogen Listeria monocytogenes causes food-borne illnesses leading to gastroenteritis, meningitis or abortion. Listeria induces its internalization into some mammalian cells through binding of the bacterial surface protein InlB to its host receptor, the Met Receptor Tyrosine Kinase. InlB-induced activation of Met stimulates host signal transduction pathways that culminate in cell surface changes driving pathogen engulfment. One mammalian protein with the potential to couple Met to downstream signalling is the adaptor CrkII. CrkII contains an unusual carboxyl-terminal SH3 domain (SH3C) that promotes entry of Listeria. However, binding partners or downstream effectors of SH3C remain unknown. Here, we use RNA interference and overexpression studies to demonstrate that SH3C affects bacterial uptake, at least in part, through stimulation of host phosphatidylinositide (PI) 3-kinase. Experiments with latex beads coated with InlB protein indicated that one potential role of SH3C and PI 3 kinase is to promote changes in the F-actin cytoskeleton necessary for particle engulfment. Taken together, our results indicate that the CrkII SH3C domain engages a cellular ligand that regulates PI 3 kinase activity and host cell surface rearrangements.


Assuntos
Listeria monocytogenes/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-crk/fisiologia , Domínios de Homologia de src , Actinas/fisiologia , Proteínas de Bactérias/metabolismo , Membrana Celular/fisiologia , Cromonas/farmacologia , Endocitose , Ativação Enzimática , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/metabolismo , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais
12.
Curr Med Chem ; 14(12): 1279-89, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17504213

RESUMO

Interferons (IFNs) are a family of pleiotropic cytokines that typically exhibit antiviral, antiproliferative, antitumor, and immunomodulatory properties. While their complex mechanisms of action remain unclear, IFNs are used clinically in the treatment of viral infections, such as hepatitis B and hepatitis C, and remain the primary treatment for a limited number of malignancies, such as melanoma, hairy cell leukemia, and non-Hodgkin's lymphoma and in autoimmune diseases such as multiple sclerosis. IFNs not only regulate somatic cell growth and division but also influence cell survival through the modulation of apoptosis. Paradoxically, IFNs are described to be both pro- and anti-apoptotic in nature. The biological effects of IFNs are primarily mediated via activation of the JAK/STAT pathway, formation of the ISGF3 and STAT1:STAT1 protein complexes, and the subsequent induction of IFN-stimulated genes. However, the activation of JAK/STAT-independent signal transduction pathways also contribute to IFN-mediated responses. To further demonstrate the complexity of the downstream events following stimulation, oligonucleotide microarray studies have shown that in excess of 300 genes are induced following treatment with IFN, some of which are crucial to the induction of apoptosis and cell growth control. In this review we describe the recent advances made in elucidating the various signaling pathways that are activated by IFNs and how these diverse signals contribute to the regulation of cell growth and apoptosis and inhibition of viral replication. Furthermore, we highlight the role of specific signaling molecules and the function(s) of particular IFN-stimulated genes that have been implicated in determining cell fate in response to IFN, as well as the clinical experience of IFN immunotherapy.


Assuntos
Interferons/fisiologia , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/fisiologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/fisiologia , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/fisiologia , Fatores Imunológicos/fisiologia , Proteínas Substratos do Receptor de Insulina , Interferon Tipo I/fisiologia , Interferon gama/fisiologia , Interferons/efeitos adversos , Interferons/imunologia , Interferons/uso terapêutico , Interleucinas/fisiologia , Janus Quinases/fisiologia , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Receptor de Insulina/fisiologia , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais/fisiologia , Viroses/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
13.
Cell Signal ; 19(8): 1662-70, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17399949

RESUMO

HGF, the ligand for the Met receptor tyrosine kinase, is a potent modulator of epithelial-mesenchymal transition and dispersal of epithelial cells, which are processes that play a crucial role in cell motility during normal development and malignant transformation. We and others have shown earlier that the adapter protein CrkII and its associated proteins positively regulate cell migratory events in response to both haptotactic and chemotactic stimuli, including HGF. Here, we demonstrate for the first time that phosphorylation of CrkII serves as a negative feedback loop to regulate motile responses upon Met stimulation. Thus, we found that the treatment of cells with HGF induces tyrosine phosphorylation of CrkII at Y221, which in turn results in inhibition of CrkII signaling via formation of an intramolecular pY221-SH2-domain interaction. Accordingly, expression of a mutant form of CrkII, CrkII-Y221F, which is resistant to phosphorylation at this negative regulatory site, enhanced Met-induced cell motility. Furthermore, we demonstrate here that the Met-induced CrkII phosphorylation depends on the Abl tyrosine kinase activity. As a corollary, we found that Abl inhibitors, such as the STI571 compound, significantly enhanced Met-induced cell motility, but failed to do so in cells that expressed the CrkII-Y221F mutant protein. Taken together, these results demonstrate that the Abl tyrosine kinase functions as a negative regulator of Met-induced cell migration, and that it does so by inducing CrkII phosphorylation at the site Y221.


Assuntos
Movimento Celular/efeitos dos fármacos , Retroalimentação Fisiológica/genética , Genes abl , Metionina/farmacologia , Proteínas Proto-Oncogênicas c-crk/metabolismo , Animais , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Fosforilação , Proteínas Proto-Oncogênicas c-crk/química , Proteínas Proto-Oncogênicas c-crk/genética , Proteínas Proto-Oncogênicas c-crk/fisiologia , Tirosina/metabolismo
14.
Mol Cell Biol ; 26(24): 9442-55, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17030600

RESUMO

Myoblast city (mbc), a member of the CDM superfamily, is essential in the Drosophila melanogaster embryo for fusion of myoblasts into multinucleate fibers. Using germ line clones in which both maternal and zygotic contributions were eliminated and rescue of the zygotic loss-of-function phenotype, we established that mbc is required in the fusion-competent subset of myoblasts. Along with its close orthologs Dock180 and CED-5, MBC has an SH3 domain at its N terminus, conserved internal domains termed DHR1 and DHR2 (or "Docker"), and C-terminal proline-rich domains that associate with the adapter protein DCrk. The importance of these domains has been evaluated by the ability of MBC mutations and deletions to rescue the mbc loss-of-function muscle phenotype. We demonstrate that the SH3 and Docker domains are essential. Moreover, ethyl methanesulfonate-induced mutations that change amino acids within the MBC Docker domain to residues that are conserved in other CDM family members nevertheless eliminate MBC function in the embryo, which suggests that these sites may mediate interactions specific to Drosophila MBC. A functional requirement for the conserved DHR1 domain, which binds to phosphatidylinositol 3,4,5-triphosphate, implicates phosphoinositide signaling in myoblast fusion. Finally, the proline-rich C-terminal sites mediate strong interactions with DCrk, as expected. These sites are not required for MBC to rescue the muscle loss-of-function phenotype, however, which suggests that MBC's role in myoblast fusion can be carried out independently of direct DCrk binding.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Proteínas de Drosophila/fisiologia , Mioblastos/enzimologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Sequência de Aminoácidos , Animais , Fusão Celular , Linhagem Celular , Proteínas do Citoesqueleto/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/enzimologia , Dados de Sequência Molecular , Família Multigênica , Complexos Multiproteicos , Mioblastos/citologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-crk/genética , Proteínas Proto-Oncogênicas c-crk/fisiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais/genética , Domínios de Homologia de src/genética
16.
J Exp Med ; 202(9): 1235-47, 2005 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16275761

RESUMO

CagA protein is a major virulence factor of Helicobacter pylori, which is delivered into gastric epithelial cells and elicits growth factor-like responses. Once within the cells, CagA is tyrosine phosphorylated by Src family kinases and targets host proteins required to induce the cell responses. We show that the phosphorylated CagA binds Crk adaptor proteins (Crk-II, Crk-I, and Crk-L) and that the interaction is important for the CagA-mediated host responses during H. pylori infection. H. pylori-induced scattering of gastric epithelial cells in culture was blocked by overexpression of dominant-negative Crk and by RNA interference-mediated knockdown of endogenous Crk. H. pylori infection of the gastric epithelium induced disruption of E-cadherin/catenin-containing adherens junctions, which was also dependent on CagA/Crk signaling. Furthermore, inhibition of the SoS1/H-Ras/Raf1, C3G/Rap1/B-Raf, or Dock180/Rac1/Wiskott-Aldrich syndrome protein family verprolin homologous protein pathway, all of which are involved downstream of Crk adaptors, greatly diminished the CagA-associated host responses. Thus, CagA targeting of Crk plays a central role in inducing the pleiotropic cell responses to H. pylori infection that cause several gastric diseases, including gastric cancer.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/imunologia , Proteínas Proto-Oncogênicas c-crk/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Adesão Celular/imunologia , Comunicação Celular/imunologia , Linhagem Celular Tumoral , Cães , Receptores ErbB/fisiologia , Mucosa Gástrica/imunologia , Humanos , Fenótipo , Fosfoproteínas/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-crk/metabolismo , Proteínas Proto-Oncogênicas c-met/fisiologia , Transdução de Sinais/imunologia , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA