Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1833(3): 629-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23220047

RESUMO

Transmembrane 4 L six family member 5 (TM4SF5) enhances cell migration and invasion, although how TM4SF5 mechanistically mediates these effects remains unknown. In the study, during efforts to understand TM4SF5-mediated signal transduction, TM4SF5 was shown to bind c-Src and thus hepatoma cell lines expressing TM4SF5 were analyzed for the significance of the interaction in cell invasion. The C-terminus of TM4SF5 bound both inactive c-Src that might be sequestered to certain cellular areas and active c-Src that might form invasive protrusions. Wildtype (WT) TM4SF5 expression enhanced migration and invasive protrusion formation in a c-Src-dependent manner, compared with TM4SF5-null control hepatoma cell lines. However, tailless TM4SF5(ΔC) cells were more efficient than WT TM4SF5 cells, suggesting a negative regulatory role by the C-terminus. TM4SF5 WT- or TM4SF5(ΔC)-mediated formation of invasive protrusions was dependent or independent on serum or epidermal growth factor treatment, respectively, although they both were dependent on c-Src. The c-Src activity of TM4SF5 WT- or TM4SF5(ΔC)-expressing cells correlated with enhanced Tyr845 phosphorylation of epidermal growth factor receptor. Y845F EGFR mutation abolished the TM4SF5-mediated invasive protrusions, but not c-Src phosphorylation. Our findings demonstrate that TM4SF5 modulates c-Src activity during TM4SF5-mediated invasion through a TM4SF5/c-Src/EGFR signaling pathway, differentially along the leading protrusive edges of an invasive cancer cell.


Assuntos
Carcinoma Hepatocelular/patologia , Movimento Celular , Receptores ErbB/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Tirosina/metabolismo , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Adesão Celular , Proliferação de Células , Receptores ErbB/genética , Imunofluorescência , Humanos , Imunoprecipitação , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/genética , Invasividade Neoplásica , Fosforilação , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas pp60(c-src)/genética , RNA Mensageiro/genética , Transdução de Sinais , Células Tumorais Cultivadas
2.
J Cell Sci ; 125(Pt 24): 5960-73, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23077174

RESUMO

Transmembrane 4 L six family member 5 (TM4SF5) plays an important role in cell migration, and focal adhesion kinase (FAK) activity is essential for homeostatic and pathological migration of adherent cells. However, it is unclear how TM4SF5 signaling mediates the activation of cellular migration machinery, and how FAK is activated during cell adhesion. Here, we showed that direct and adhesion-dependent binding of TM4SF5 to FAK causes a structural alteration that may release the inhibitory intramolecular interaction in FAK. In turn, this may activate FAK at the cell's leading edge, to promote migration/invasion and in vivo metastasis. TM4SF5-mediated FAK activation occurred during integrin-mediated cell adhesion. TM4SF5 was localized at the leading edge of the cells, together with FAK and actin-organizing molecules, indicating a signaling link between TM4SF5/FAK and actin reorganization machinery. Impaired interactions between TM4SF5 and FAK resulted in an attenuated FAK phosphorylation (the signaling link to actin organization machinery) and the metastatic potential. Our findings demonstrate that TM4SF5 directly binds to and activates FAK in an adhesion-dependent manner, to regulate cell migration and invasion, suggesting that TM4SF5 is a promising target in the treatment of metastatic cancer.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Quinase 1 de Adesão Focal/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Tetraspaninas/genética , Sequência de Aminoácidos , Animais , Carcinoma Hepatocelular/enzimologia , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Ativação Enzimática , Feminino , Xenoenxertos , Humanos , Neoplasias Hepáticas/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Metástase Neoplásica , Fosforilação , Transdução de Sinais , Tetraspaninas/metabolismo
3.
J Biol Chem ; 287(33): 27499-509, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22761432

RESUMO

Integrin-mediated adhesion to extracellular matrix proteins is dynamically regulated during morphological changes and cell migration. Upon cell adhesion, protein-protein interactions among molecules at focal adhesions (FAs) play major roles in the regulation of cell morphogenesis and migration. Although tyrosine phosphorylation of paxillin is critically involved in adhesion-mediated signaling, the significance of paxillin phosphorylation at Ser-85 and the mechanism by which it regulates cell migration remain unclear. In this study, we examined how Ser-85 phosphorylation of paxillin affects FA formation and cell migration. We found that paxillin phosphorylation at Ser-85 occurred during HeLa cell adhesion to collagen I and was concomitant with tyrosine phosphorylation of both focal adhesion kinase and talin. However, the non-phosphorylatable S85A mutant of paxillin impaired cell spreading, FA turnover, and migration toward collagen I but not toward serum. Furthermore, whereas the (presumably indirect) interaction between paxillin and the C-terminal tail of talin led to dynamic FAs at the cell boundary, S85A paxillin did not bind talin and caused stabilized FAs in the central region of cells. Together, these observations suggest that cell adhesion-dependent Ser-85 phosphorylation of paxillin is important for its interaction with talin and regulation of dynamic FAs and cell migration.


Assuntos
Movimento Celular/fisiologia , Adesões Focais/metabolismo , Paxilina/metabolismo , Substituição de Aminoácidos , Adesão Celular/fisiologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Adesões Focais/genética , Células HeLa , Humanos , Mutação de Sentido Incorreto , Paxilina/genética , Fosforilação/fisiologia , Estrutura Terciária de Proteína , Serina/genética , Serina/metabolismo
4.
Biochem J ; 443(3): 691-700, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22292774

RESUMO

The EMT (epithelial-mesenchymal transition) is involved in fibrosis and cancer, and is regulated by different signalling pathways mediated through soluble factors, actin reorganization and transcription factor actions. Because the tetraspan (also called tetraspanin) TM4SF5 (transmembrane 4 L6 family member 5) is highly expressed in hepatocellular carcinoma and induces EMT, understanding how TM4SF5 expression in hepatocytes is regulated is important. We explored the mechanisms that induce TM4SF5 expression and whether impaired signalling pathways for TM4SF5 expression inhibit the acquisition of mesenchymal cell features, using human and mouse normal hepatocytes. We found that TGFß1 (transforming growth factor ß1)-mediated Smad activation caused TM4SF5 expression and EMT, and activation of the EGFR [EGF (epidermal growth factor) receptor] pathway. Inhibition of EGFR activity following TGFß1 treatment abolished acquisition of EMT, suggesting a link from Smads to EGFR for TM4SF5 expression. Further, TGFß1-mediated EGFR activation and TM4SF5 expression were abolished by EGFR suppression or extracellular EGF depletion. Smad overexpression mediated EGFR activation and TM4SF5 expression in the absence of serum, and EGFR kinase inactivation or EGF depletion abolished Smad-overexpression-induced TM4SF5 and mesenchymal cell marker expression. Inhibition of Smad, EGFR or TM4SF5 using Smad7 or small compounds also blocked TM4SF5 expression and/or EMT. These results indicate that TGFß1- and growth factor-mediated signalling activities mediate TM4SF5 expression leading to acquisition of mesenchymal cell features, suggesting that TM4SF5 induction may be involved in the development of liver pathologies.


Assuntos
Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Linhagem Celular Tumoral , Humanos
5.
J Biol Chem ; 285(46): 36021-31, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20829364

RESUMO

Protein-protein interactions and/or signaling activities at focal adhesions, where integrin-mediated adhesion to extracellular matrix occurs, are critical for the regulation of adhesion-dependent cellular functions. Although the phosphorylation and activities of focal adhesion molecules have been intensively studied, the effects of the O-GlcNAc modification of their Ser/Thr residues on cellular functions have been largely unexplored. We investigated the effects of O-GlcNAc modification on actin reorganization and morphology of rat insulinoma INS-1 cells after glucosamine (GlcN) treatment. We found that paxillin, a key adaptor molecule in focal adhesions, could be modified by O-GlcNAc in INS-1 cells treated with GlcN and in pancreatic islets from mice treated with streptozotocin. Ser-84/85 in human paxillin appeared to be modified by O-GlcNAc, which was inversely correlated to Ser-85 phosphorylation (Ser-83 in rat paxillin). Integrin-mediated adhesion signaling inhibited the GlcN treatment-enhanced O-GlcNAc modification of paxillin. Adherent INS-1 cells treated with GlcN showed restricted protrusions, whereas untreated cells showed active protrusions for multiple-elongated morphologies. Upon GlcN treatment, expression of a triple mutation (S83A/S84A/S85A) resulted in no further restriction of protrusions. Together these observations suggest that murine pancreatic ß cells may have restricted actin organization upon GlcN treatment by virtue of the O-GlcNAc modification of paxillin, which can be antagonized by a persistent cell adhesion process.


Assuntos
Acetilglucosamina/metabolismo , Actinas/metabolismo , Glucosamina/farmacologia , Paxilina/metabolismo , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Humanos , Imuno-Histoquímica , Insulinoma/metabolismo , Insulinoma/patologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Microscopia de Fluorescência , Mutação , Paxilina/genética , Fosforilação/efeitos dos fármacos , Ratos , Serina/genética , Serina/metabolismo , Estreptozocina
6.
Biochim Biophys Acta ; 1803(8): 975-82, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20399237

RESUMO

Transmembrane 4 L six family member 5 (TM4SF5) causes epithelial-mesenchymal transition (EMT) for aberrant cell proliferation. However, the effects of TM4SF5 expression on cell cycle are unknown so far. In this study, using hepatocytes that either ectopically or endogenously express TM4SF5 and human hepatocarcinoma tissues, the role of TM4SF5 in G1/S phase progression was examined. We found that TM4SF5 expression accelerated G1/S phase progression with facilitated cyclin D1 and E expression and Rb phosphorylation. Furthermore, TM4SF5 enhanced trafficking of CDK4 and cyclin D1 into the nucleus and induced complex formation between them. However, TM4SF5-facilitated G1/S phase progression was blocked by silencing of p27Kip1 using siRNA or by infection of active RhoA. Pharmacological inhibition of ROCK accelerated the G1/S phase progression of control TM4SF5-unexpressing cells. Altogether, these observations suggest that TM4SF5 accelerates G1/S phase progression with facilitated CDK4/cyclin D1 entry into the nucleus, which might be supported by TM4SF5-mediated actin reorganization through cytosolic p27Kip1 expression and Rho GTPase activity.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fase G1/fisiologia , Proteínas de Membrana/metabolismo , Fase S/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas de Membrana/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética
7.
Blood ; 113(8): 1845-55, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19036703

RESUMO

Tetraspan TM4SF5 is highly expressed in a diverse number of tumor types. Here we explore the mechanistic roles of TM4SF5 in angiogenesis. We found that TM4SF5 overexpression correlates with vascular endothelial growth factor (VEGF) expression in SNU449 hepatocytes and with vessel formation in clinical hepatocarcinoma samples. Conditioned media from TM4SF5-expressing cells enhanced viability and tube formation of primary human umbilical vein endothelial cells, and outgrowth of endothelial cells from aorta ring segments, which was abolished by treatment with an anti-VEGF antibody. TM4SF5 retained integrin alpha(5) on the cell surface for VEGF induction, and preincubation with anti-integrin alpha(5) antibody abolished TM4SF5-mediated VEGF expression and secretion. TM4SF5-mediated effects required integrin alpha(5), c-Src, and signal transducer and activator of transcription 3 (STAT3). In addition, tumors from nude mice injected with TM4SF5-expressing cells and from clinical human hepatocarcinoma tissues showed enhanced integrin alpha(5) expression, vessel formation, and signaling activity, which were inhibited by administration of anti-integrin alpha(5) or -VEGF antibody. This study suggests that TM4SF5 facilitates angiogenesis of neighboring endothelial cells through VEGF induction, mediated by cooperation between TM4SF5 and integrin alpha(5) of epithelial cells.


Assuntos
Carcinoma Hepatocelular/irrigação sanguínea , Integrina alfa5/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticorpos/farmacologia , Aorta/citologia , Proteína Tirosina Quinase CSK , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Integrina alfa5/imunologia , Neoplasias Hepáticas/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Quinases da Família src
8.
J Cell Biochem ; 111(1): 59-66, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20506553

RESUMO

Overexpression of transmembrane 4 L six family member 5 (TM4SF5), a four-transmembrane L6 family member, causes aberrant cell proliferation and angiogenesis, but the roles of TM4SF5 in migration, invasion, and tumor metastasis remain unknown. Using in vitro hepatocarcinoma cells that ectopically or endogenously express TM4SF5 and in vivo mouse systems, roles of TM4SF5 in metastatic potentials were examined. We found that TM4SF5 expression facilitated migration, invadopodia formation, MMP activation, invasion, and eventually lung metastasis in nude mice, but suppression of TM4SF5 with its shRNA blocked the effects. Altogether, TM4SF5-mediated migration and invasion suggest that TM4SF5 may be therapeutically targeted to deal with TM4SF5-mediated hepatocellular cancers.


Assuntos
Carcinoma Hepatocelular , Movimento Celular/fisiologia , Hepatócitos , Neoplasias Hepáticas , Proteínas de Membrana/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Feminino , Hepatócitos/citologia , Hepatócitos/patologia , Hepatócitos/fisiologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica
9.
Hepatology ; 49(4): 1316-25, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19177595

RESUMO

UNLABELLED: We previously reported that the four-transmembrane L6 family member 5 (TM4SF5) was highly expressed in hepatocarcinoma, induced morphological elongation and epithelial-mesenchymal transition, and caused abnormal cell growth in multilayers in vitro and tumor formation in vivo. In this study, we identified a synthetic compound, 4'-(p-toluenesulfonylamido)-4-hydroxychalcone (TSAHC) that antagonized both the TM4SF5-mediated multilayer growth and TM4SF5-enhanced migration/invasion. TSAHC treatment induced multilayer-growing cells to grow in monolayers, recovering contact inhibition without accompanying apoptosis, and inhibited chemotactic migration and invasion. Tumor formation in nude mice injected with TM4SF5-expressing cells and the growth of cells expressing endogenous TM4SF5, but not of TM4SF5-null cells, was suppressed by treatment with TSAHC, but not by treatment with its analogs. The structure-activity relationship indicated the significance of 4'-p-toluenesulfonylamido and 4-hydroxy groups for the anti-TM4SF5 effects of TSAHC. Point mutations of the putative N-glycosylation sites abolished the TM4SF5-specific TSAHC responsiveness. CONCLUSION: These observations suggest that TM4SF5-enhanced tumorigenic proliferation and metastatic potential can be blocked by TSAHC, likely through targeting the extracellular region of TM4SF5, which is important for protein-protein interactions.


Assuntos
Carcinoma Hepatocelular/metabolismo , Chalcona/análogos & derivados , Chalconas/farmacologia , Hepatócitos/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Sulfonamidas/farmacologia , Animais , Testes de Carcinogenicidade , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalcona/farmacologia , Inibição de Contato/efeitos dos fármacos , Glicosilação , Humanos , Proteínas de Membrana/genética , Camundongos , Fenótipo , Mutação Puntual , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade
10.
Carcinogenesis ; 30(11): 1872-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19789264

RESUMO

Four-transmembrane L6 family member 5 (TM4SF5) and its homolog L6, a tumor antigen, form a four-transmembrane L6 family. TM4SF5 expression causes uncontrolled cell proliferation and angiogenesis. Although other genuine transmembrane 4 superfamily (TM4SF) members co-operate with integrins for cell migration, roles of TM4SF5 in the cellular spreading and migration are unknown. Using hepatocarcinoma cell clones that ectopically express TM4SF5, we found that cross talks via an extracellular interaction between TM4SF5 and integrin alpha2 in collagen type I environment inhibited integrin alpha2 functions such as spreading on and migration toward collagen I, which were recovered by suppression of TM4SF5 or structural disturbance of its second extracellular loop using a peptide or mutagenesis. Altogether, the observations suggest that TM4SF5 in hepatocytes negatively regulates integrin alpha2 function via an interaction between the extracellular loop 2 of TM4SF5 and integrin alpha2 during cell spreading on and migration through collagen I environment.


Assuntos
Integrina alfa2/metabolismo , Neoplasias Hepáticas/genética , Proteínas de Membrana/metabolismo , Antígenos de Neoplasias/metabolismo , Movimento Celular , Células Clonais , Colágeno Tipo I/metabolismo , Matriz Extracelular , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Integrina alfa2/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/genética , Invasividade Neoplásica/genética , Ligação Proteica , Estrutura Terciária de Proteína , Receptor Cross-Talk
11.
Exp Cell Res ; 314(11-12): 2238-48, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18570920

RESUMO

O-GlcNAc transferase (OGT)-mediated modification of protein Ser/Thr residues with O-GlcNAc influences protein activity, similar to the effects of phosphorylation. The anti-apoptotic Akt1 is both activated by phosphorylation and modified with O-GlcNAc. However, the nature and significance of the Akt1 O-GlcNAc modification is unknown. The relationship of O-GlcNAc modification and phosphorylation at Akt1 Ser473 was examined with respect to apoptosis of murine beta-pancreatic cells. Glucosamine treatment induced apoptosis, which correlated with enhanced O-GlcNAc modification of Akt1 and concomitant reduction in Ser473 phosphorylation. Pharmacological inhibition of OGT or O-GlcNAcase revealed an inverse correlation between O-GlcNAc modification and Ser473 phosphorylation of Akt1. MALDI-TOF/TOF mass spectrometry analysis of Akt1 immunoprecipitates from glucosamine-treated cells, but not untreated controls, showed a peptide containing S473/T479 that was presumably modified with O-GlcNAc. Furthermore, in vitro O-GlcNAc-modification analysis of wildtype and mutant Akt1 revealed that S473 was targeted by recombinant OGT. A S473A Akt1 mutant demonstrated reduced basal and glucosamine-induced Akt1 O-GlcNAc modification compared with wildtype Akt1. Furthermore, wildtype Akt1, but not the S473A mutant, appeared to be associated with OGT following glucosamine treatment. Together, these observations suggest that Akt1 Ser473 may undergo both phosphorylation and O-GlcNAc modification, and the balance between these may regulate murine beta-pancreatic cell fate.


Assuntos
Acetilglucosamina/metabolismo , Apoptose/fisiologia , Células Secretoras de Insulina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina/metabolismo , Aloxano/metabolismo , Animais , Linhagem Celular , Glucosamina/metabolismo , Glucose/metabolismo , Humanos , Hiperglicemia , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Manitol/metabolismo , Camundongos , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Treonina/metabolismo
12.
Oncotarget ; 6(25): 21655-74, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26091349

RESUMO

The adhesion properties of cells are involved in tumor metastasis. Although KRS at the plasma membrane is shown important for cancer metastasis, additionally to canonical roles of cytosolic KRS in protein translation, how KRS and its downstream effectors promote the metastatic migration remains unexplored. Disseminative behaviors (an earlier metastatic process) of colon cancer cell spheroids embedded in 3D collagen gels were studied with regards to cell adhesion properties, and relevance in KRS(-/+) knocked-down animal and clinical colon cancer tissues. Time-lapse imaging revealed KRS-dependent cell dissemination from the spheroids, whereas KRS-suppressed spheroids remained static due to the absence of outbound movements supported by cell-extracellular matrix (ECM) adhesion. While keeping E-cadherin at the outward disseminative cells, KRS caused integrin-involved intracellular signaling for ERK/c-Jun, paxillin, and cell-ECM adhesion-mediated signaling to modulate traction force for crawling movement. KRS-suppressed spheroids became disseminative following ERK or paxillin re-expression. The KRS-dependent intracellular signaling activities correlated with the invasiveness in clinical colon tumor tissues and in KRS(-/+) knocked-down mice tissues. Collectively, these observations indicate that KRS at the plasma membrane plays new roles in metastatic migration as a signaling inducer, and causes intracellular signaling for cancer dissemination, involving cell-cell and cell-ECM adhesion, during KRS-mediated metastasis.


Assuntos
Colágeno Tipo I/metabolismo , Neoplasias do Colo/enzimologia , Lisina-tRNA Ligase/metabolismo , Animais , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Citosol/metabolismo , Matriz Extracelular/metabolismo , Feminino , Transferência Ressonante de Energia de Fluorescência , Técnica Indireta de Fluorescência para Anticorpo , Células HCT116 , Humanos , Camundongos , Metástase Neoplásica , Paxilina/metabolismo , Fosforilação , Biossíntese de Proteínas , Transdução de Sinais
13.
Cancer Biol Ther ; 11(3): 330-6, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21099346

RESUMO

Two separate clinical studies of advanced hepatocarcinoma patients recently reported that the multikinase inhibitor sorafenib (nexavar) could extend survival of the patients only by 2-3 months. We also previously demonstrated that 4'-(p-toluenesulfonylamido)-4-hydroxychalcone (TSAHC) blocks the multilayer growth and migration mediated by TM4SF5, which is highly expressed in approximately 80% of Korean hepatocarcinoma patients. Therefore, we wondered how TSAHC might be different from sorafenib to deal with hepatocarcinoma in terms of the therapeutic characteristics including specificity for TM4SF5. TM4SF5 is previously shown to mediate tumorigenesis through cytosolic p27Kip1-mediated inactivation of RhoA, epithelial-mesenchymal transition, multilayer growth, migration, invasion, and tumor angiogenesis. In this study, TSAHC and two derivatives showed similar antagonistic activities against TM4SF5-mediated signaling and multilayer growth in vitro and anti-tumorigenic activity even in early stages of TM4SF5-mediated tumor formation in nude mice. Meanwhile, sorafenib was only effective much later in tumorigenesis in vivo and affected in vitro proliferation in a TM4SF5-independent manner. Altogether, these observations suggest that TSAHC may be a promising anti-tumorigenic reagent, especially against TM4SF5-mediated hepatocarcinoma.


Assuntos
Antineoplásicos/farmacologia , Benzenossulfonatos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Chalcona/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Proteínas de Membrana/metabolismo , Piridinas/farmacologia , Sulfonamidas/farmacologia , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Chalcona/metabolismo , Chalcona/farmacologia , Descoberta de Drogas , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Niacinamida/análogos & derivados , Compostos de Fenilureia , Transdução de Sinais , Sorafenibe , Sulfonamidas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA