Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Rev Mol Cell Biol ; 11(9): 633-43, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20729930

RESUMO

Cell migration affects all morphogenetic processes and contributes to numerous diseases, including cancer and cardiovascular disease. For most cells in most environments, movement begins with protrusion of the cell membrane followed by the formation of new adhesions at the cell front that link the actin cytoskeleton to the substratum, generation of traction forces that move the cell forwards and disassembly of adhesions at the cell rear. Adhesion formation and disassembly drive the migration cycle by activating Rho GTPases, which in turn regulate actin polymerization and myosin II activity, and therefore adhesion dynamics.


Assuntos
Movimento Celular , Citoesqueleto/metabolismo , Animais , Adesão Celular , Miosina Tipo II/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
2.
Ann Surg Oncol ; 23(6): 1993-2000, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26847682

RESUMO

PURPOSE: Most patients with pancreatic ductal adenocarcinoma (PDAC) die within 5 years following resection plus adjuvant gemcitabine (Gem) from outgrowth of occult metastases. We hypothesized that inhibition of the KRAS pathway with the MEK inhibitor trametinib would inhibit the outgrowth of occult liver metastases in a preclinical model. METHODS: Liver metastases harvested from two patients with PDAC (Tumors 608, 366) were implanted orthotopically in mice. Tumor cell lines were derived and transduced with lentiviruses encoding luciferase and injected into spleens of mice generating microscopic liver metastases. Growth kinetics of liver metastases were measured with bioluminescent imaging and time-to-progression (TTP), progression-free survival (PFS), and overall survival (OS) were determined. RESULTS: Trametinib (0.3 mg/kg BID) significantly prolonged OS versus control (Tumor 608: 114 vs. 43 days, p < 0.001; Tumor 366: not reached vs. 167 days, p = 0.0488). In vivo target validation demonstrated trametinib significantly reduced phosphorylated-ERK and expression of the ERK-responsive gene DUSP6. In a randomized, preclinical trial, mice were randomized to: (1) control, (2) adjuvant Gem (100 mg/kg IP, Q3 days) × 7 days followed by surveillance, or (3) adjuvant Gem followed by trametinib. Sequential Gem-trametinib significantly decreased metastatic cell outgrowth and increased TTP and PFS. CONCLUSIONS: Treatment of mice bearing micrometastases with trametinib significantly delayed tumor outgrowth by effectively inhibiting KRAS-MEK-ERK signaling. In a randomized, preclinical, murine trial adjuvant sequential Gem followed by trametinib inhibited occult metastatic cell outgrowth in the liver and increased PFS versus adjuvant Gem alone. An adjuvant trial of sequential Gem-trametinib is being planned in patients with resected PDAC.


Assuntos
Carcinoma Ductal Pancreático/prevenção & controle , Neoplasias Hepáticas/prevenção & controle , Neoplasias Pancreáticas/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Piridonas/farmacologia , Pirimidinonas/farmacologia , Animais , Carcinoma Ductal Pancreático/secundário , Humanos , Neoplasias Hepáticas/secundário , Camundongos , Neoplasias Pancreáticas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nat Cell Biol ; 4(4): E97-100, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11944043

RESUMO

Cell migration is an integrated process that requires the continuous, coordinated formation and disassembly of adhesions. These processes are complex and require a regulated interaction of numerous molecules, and the activation of specific signalling pathways. Even though understanding these processes is challenging, important insights are beginning to emerge, and the technology to facilitate significant advances in this area is now in place.


Assuntos
Técnicas Citológicas , Transdução de Sinais , Animais , Adesão Celular , Movimento Celular , Guanosina Trifosfato/metabolismo , Humanos , Proteínas rho de Ligação ao GTP/metabolismo
4.
Nat Cell Biol ; 6(2): 154-61, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14743221

RESUMO

Cell migration is a complex, highly regulated process that involves the continuous formation and disassembly of adhesions (adhesion turnover). Adhesion formation takes place at the leading edge of protrusions, whereas disassembly occurs both at the cell rear and at the base of protrusions. Despite the importance of these processes in migration, the mechanisms that regulate adhesion formation and disassembly remain largely unknown. Here we develop quantitative assays to measure the rate of incorporation of molecules into adhesions and the departure of these proteins from adhesions. Using these assays, we show that kinases and adaptor molecules, including focal adhesion kinase (FAK), Src, p130CAS, paxillin, extracellular signal-regulated kinase (ERK) and myosin light-chain kinase (MLCK) are critical for adhesion turnover at the cell front, a process central to migration.


Assuntos
Adesão Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Movimento Celular/fisiologia , Proteína Substrato Associada a Crk , Proteínas do Citoesqueleto/genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Quinase de Cadeia Leve de Miosina/genética , Paxilina , Fosfoproteínas/genética , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína p130 Retinoblastoma-Like , Quinases da Família src/genética
5.
Am J Physiol Cell Physiol ; 297(4): C814-22, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19587216

RESUMO

Atherogenesis involves activation of NF-kappaB in endothelial cells by fluid shear stress. Because this pathway involves integrins, we investigated the involvement of focal adhesion kinase (FAK). We found that FAK was not required for flow-stimulated translocation of the p65 NF-kappaB subunit to the nucleus but was essential for phosphorylation of p65 on serine 536 and induction of ICAM-1, an NF-kappaB-dependent gene. NF-kappaB activation by TNF-alpha or hydrogen peroxide was FAK independent. Events upstream of NF-kappaB, including integrin activation, Rac activation, reactive oxygen production, and degradation of IkappaB, were FAK independent. FAK therefore regulates NF-kappaB phosphorylation and transcriptional activity in response to flow by a novel mechanism.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , NF-kappa B/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Peróxido de Hidrogênio/farmacologia , Quinase I-kappa B/metabolismo , Integrinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Fosforilação , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Estresse Mecânico , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas rac de Ligação ao GTP/metabolismo
6.
Breast Cancer Res ; 11(4): R52, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19630967

RESUMO

INTRODUCTION: Signal transducer and activator of transcription (STAT) 5b is a transcription factor involved in pro-proliferative and pro-survival signaling in a number of solid tumors, including breast cancer. The contribution of STAT5b to breast cancer cell motility has not been explored. This work aims to elucidate the role of STAT5b in breast cancer cell migration. METHODS: STAT5b was knocked down by using siRNA in two aggressive, highly migratory breast cancer cell lines (BT-549 and MDA-MB-231), and transwell migration assays were performed to determine the importance of STAT5b for their migration. Knockdown-rescue experiments were used to validate the specificity of STAT5b knockdown and to determine which regions/functions of STAT5b are necessary for its role in migration. Live-cell imaging of wound healing and spreading was carried out to examine cell morphology and motility after STAT5b knockdown. RESULTS: Knockdown of STAT5b, but not STAT5a, inhibited migration of BT-549 and MDA-MB-231 breast cancer cells to serum by 60% to 80%, and inhibited migration equally over a range of serum concentrations (0.1% to 10% serum). Migratory inhibition upon STAT5b knockdown could be rescued by reintroduction of wild-type STAT5b, as well as Y699F- and dominant-negative STAT5b mutants, but not an SH2 domain defective R618K-STAT5b mutant. beta1- integrin-mediated migration of breast cancer cells to fibronectin was inhibited with STAT5b knockdown, and loss of STAT5b correlated with loss of directional migration and formation of multiple, highly contractile protrusions upon attachment to fibronectin. CONCLUSIONS: The data presented here demonstrate that STAT5b is integral to breast cancer cell migration and identify a novel, SH2-dependent function of STAT5b in regulating beta1-integrin-mediated migration of highly aggressive breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Integrina beta1/fisiologia , Invasividade Neoplásica/fisiopatologia , Proteínas de Neoplasias/fisiologia , Fator de Transcrição STAT5/fisiologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/patologia , Movimento Celular/fisiologia , Feminino , Fibronectinas/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Neoplasias/química , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Cicatrização , Domínios de Homologia de src
7.
J Cell Biol ; 166(2): 225-35, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15263018

RESUMO

Shigella, the causative agent of bacillary dysentery, invades epithelial cells in a process involving Src tyrosine kinase signaling. Cortactin, a ubiquitous actin-binding protein present in structures of dynamic actin assembly, is the major protein tyrosine phosphorylated during Shigella invasion. Here, we report that RNA interference silencing of cortactin expression, as does Src inhibition in cells expressing kinase-inactive Src, interferes with actin polymerization required for the formation of cellular extensions engulfing the bacteria. Shigella invasion induced the recruitment of cortactin at plasma membranes in a tyrosine phosphorylation-dependent manner. Overexpression of wild-type forms of cortactin or the adaptor protein Crk favored Shigella uptake, and Arp2/3 binding-deficient cortactin derivatives or an Src homology 2 domain Crk mutant interfered with bacterial-induced actin foci formation. Crk was shown to directly interact with tyrosine-phosphorylated cortactin and to condition cortactin-dependent actin polymerization required for Shigella uptake. These results point at a major role for a Crk-cortactin complex in actin polymerization downstream of tyrosine kinase signaling.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Células Epiteliais/microbiologia , Proteínas dos Microfilamentos/fisiologia , Shigella/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Extensões da Superfície Celular/microbiologia , Cortactina , Endocitose , Células Epiteliais/ultraestrutura , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-crk , RNA Interferente Pequeno/farmacologia
8.
J Cell Biol ; 162(2): 281-91, 2003 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-12876277

RESUMO

Activation of the Ras-MAPK signal transduction pathway is necessary for biological responses both to growth factors and ECM. Here, we provide evidence that phosphorylation of S298 of MAPK kinase 1 (MEK1) by p21-activated kinase (PAK) is a site of convergence for integrin and growth factor signaling. We find that adhesion to fibronectin induces PAK1-dependent phosphorylation of MEK1 on S298 and that this phosphorylation is necessary for efficient activation of MEK1 and subsequent MAPK activation. The rapid and efficient activation of MEK and phosphorylation on S298 induced by cell adhesion to fibronectin is influenced by FAK and Src signaling and is paralleled by localization of phospho-S298 MEK1 and phospho-MAPK staining in peripheral membrane-proximal adhesion structures. We propose that FAK/Src-dependent, PAK1-mediated phosphorylation of MEK1 on S298 is central to the organization and localization of active Raf-MEK1-MAPK signaling complexes, and that formation of such complexes contributes to the adhesion dependence of growth factor signaling to MAPK.


Assuntos
Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células COS , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Chlorocebus aethiops , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Adesões Focais/metabolismo , Regulação da Expressão Gênica , Fator de Crescimento Insulin-Like I/farmacologia , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Mutação , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , Pirimidinas/farmacologia , Ratos , Proteínas Recombinantes/metabolismo , Quinases Ativadas por p21 , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 28(12): 2115-22, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18787183

RESUMO

OBJECTIVE: Smooth muscle cell (SMC) differentiation is a dynamic process that must be tightly regulated for proper vascular development and to control the onset of vascular disease. Our laboratory previously reported that a specific focal adhesion kinase (FAK) inhibitor termed FRNK (FAK Related Non-Kinase) is selectively expressed in large arterioles when SMCs are transitioning from a synthetic to contractile phenotype and that FRNK inhibits FAK-dependent SMC proliferation and migration. Herein, we sought to determine whether FRNK expression modulates SMC phenotypes in vivo. METHODS AND RESULTS: We present evidence that FRNK(-/-) mice exhibit attenuated SM marker gene expression during postnatal vessel growth and after vascular injury. We also show that FRNK expression is regulated by transforming growth factor (TGF)-beta and that forced expression of FRNK in cultured cells induces serum- and TGF-beta-stimulated SM marker gene expression, whereas FRNK deletion or expression of a constitutively activated FAK variant attenuated SM gene transcription. CONCLUSIONS: These data highlight the possibility that extrinsic signals regulate the SMC gene profile, at least in part, by modulating the expression of FRNK and that tight regulation of FAK activity by FRNK is important for proper SMC differentiation during development and after vascular injury.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/lesões , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Tirosina Quinases/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia
10.
Clin Cancer Res ; 14(3): 627-32, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18245520

RESUMO

The tumor microenvironment plays a central role in cancer progression and metastasis. Within this environment, cancer cells respond to a host of signals including growth factors and chemotactic factors, as well as signals from adjacent cells, cells in the surrounding stroma, and signals from the extracellular matrix. Targeting the pathways that mediate many of these signals has been a major goal in the effort to develop therapeutics.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Neoplasias/enzimologia , Neoplasias/fisiopatologia , Transdução de Sinais , Antineoplásicos/uso terapêutico , Progressão da Doença , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/genética , Humanos , Integrinas/fisiologia , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Oligonucleotídeos Antissenso/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , RNA Interferente Pequeno/genética
11.
Lab Chip ; 19(7): 1193-1204, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30839006

RESUMO

The development of drugs to treat cancer is hampered by the inefficiency of translating pre-clinical in vitro monoculture and mouse studies into clinical benefit. There is a critical need to improve the accuracy of evaluating pre-clinical drug efficacy through the development of more physiologically relevant models. In this study, a human triculture 3D in vitro tumor microenvironment system (TMES) was engineered to accurately mimic the tumor microenvironment. The TMES recapitulates tumor hemodynamics and biological transport with co-cultured human microvascular endothelial cells, pancreatic ductal adenocarcinoma, and pancreatic stellate cells. We demonstrate that significant tumor cell transcriptomic changes occur in the TMES that correlate with the in vivo xenograft and patient transcriptome. Treatment with therapeutically relevant doses of chemotherapeutics yields responses paralleling the patients' clinical responses. Thus, this model provides a unique platform to rigorously evaluate novel therapies and is amenable to using patient tumor material directly, with applicability for patient avatars.


Assuntos
Biomimética/métodos , Carcinoma Ductal Pancreático/patologia , Microambiente Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Microambiente Tumoral/efeitos dos fármacos
12.
Oncotarget ; 9(10): 8972-8984, 2018 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-29507667

RESUMO

Successful therapeutic options remain elusive for pancreatic cancer. The exquisite sensitivity and specificity of humoral and cellular immunity may provide therapeutic approaches if antigens specific for pancreatic cancer cells can be identified. Here we characterize SAS1B (ovastacin, ASTL, astacin-like), a cancer-oocyte antigen, as an attractive immunotoxin target expressed at the surface of human pancreatic cancer cells, with limited expression among normal tissues. Immunohistochemistry shows that most pancreatic cancers are SAS1Bpos (68%), while normal pancreatic ductal epithelium is SAS1Bneg. Pancreatic cancer cell lines developed from patient-derived xenograft models display SAS1B cell surface localization, in addition to cytoplasmic expression, suggesting utility for SAS1B in multiple immunotherapeutic approaches. When pancreatic cancer cells were treated with an anti-SAS1B antibody-drug conjugate, significant cell death was observed at 0.01-0.1 µg/mL, while SAS1Bneg human keratinocytes were resistant. Cytotoxicity was correlated with SAS1B cell surface expression; substantial killing was observed for tumors with low steady state SAS1B expression, suggesting a substantial proportion of SAS1Bpos tumors can be targeted in this manner. These results demonstrate SAS1B is a surface target in pancreatic cancer cells capable of binding monoclonal antibodies, internalization, and delivering cytotoxic drug payloads, supporting further development of SAS1B as a novel target for pancreatic cancer.

13.
Clin Cancer Res ; 24(6): 1415-1425, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29288236

RESUMO

Purpose: Patients with pancreatic ductal adenocarcinoma (PDAC) who undergo surgical resection and adjuvant chemotherapy have an expected survival of only 2 years due to disease recurrence, frequently in the liver. We investigated the role of liver macrophages in progression of PDAC micrometastases to identify adjuvant treatment strategies that could prolong survival.Experimental Design: A murine splenic injection model of hepatic micrometastatic PDAC was used with five patient-derived PDAC tumors. The impact of liver macrophages on tumor growth was assessed by (i) depleting mouse macrophages in nude mice with liposomal clodronate injection, and (ii) injecting tumor cells into nude versus NOD-scid-gamma mice. Immunohistochemistry and flow cytometry were used to measure CD47 ("don't eat me signal") expression on tumor cells and characterize macrophages in the tumor microenvironment. In vitro engulfment assays and mouse experiments were performed with CD47-blocking antibodies to assess macrophage engulfment of tumor cells, progression of micrometastases in the liver and mouse survival.Results:In vivo clodronate depletion experiments and NOD-scid-gamma mouse experiments demonstrated that liver macrophages suppress the progression of PDAC micrometastases. Five patient-derived PDAC cell lines expressed variable levels of CD47. In in vitro engulfment assays, CD47-blocking antibodies increased the efficiency of PDAC cell clearance by macrophages in a manner which correlated with CD47 receptor surface density. Treatment of mice with CD47-blocking antibodies resulted in increased time-to-progression of metastatic tumors and prolonged survival.Conclusions: These findings suggest that following surgical resection of PDAC, adjuvant immunotherapy with anti-CD47 antibody could lead to substantially improved outcomes for patients. Clin Cancer Res; 24(6); 1415-25. ©2017 AACR.


Assuntos
Antígeno CD47/antagonistas & inibidores , Imunomodulação , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Animais , Antígeno CD47/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Humanos , Imuno-Histoquímica , Imunoterapia/métodos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Metástase Neoplásica , Estadiamento de Neoplasias , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Commun ; 9(1): 4275, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30323222

RESUMO

Predicting the response and identifying additional targets that will improve the efficacy of chemotherapy is a major goal in cancer research. Through large-scale in vivo and in vitro CRISPR knockout screens in pancreatic ductal adenocarcinoma cells, we identified genes whose genetic deletion or pharmacologic inhibition synergistically increase the cytotoxicity of MEK signaling inhibitors. Furthermore, we show that CRISPR viability scores combined with basal gene expression levels could model global cellular responses to the drug treatment. We develop drug response evaluation by in vivo CRISPR screening (DREBIC) method and validated its efficacy using large-scale experimental data from independent experiments. Comparative analyses demonstrate that DREBIC predicts drug response in cancer cells from a wide range of tissues with high accuracy and identifies therapeutic vulnerabilities of cancer-causing mutations to MEK inhibitors in various cancer types.


Assuntos
Antineoplásicos/farmacologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Técnicas de Química Combinatória , Sistemas de Liberação de Medicamentos , Técnicas de Inativação de Genes , Testes Genéticos , Modelos Biológicos , Neoplasias Pancreáticas/genética , Animais , Pontos de Checagem do Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Reprodutibilidade dos Testes
15.
Curr Biol ; 12(21): 1852-7, 2002 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-12419186

RESUMO

The GTPase dynamin is required for endocytic vesicle formation. Dynamin has also been implicated in regulating the actin cytoskeleton, but the mechanism by which it does so is unclear. Through interactions via its proline-rich domain (PRD), dynamin binds several proteins, including cortactin, profilin, syndapin, and murine Abp1, that regulate the actin cytoskeleton. We investigated the interaction of dynamin2 and cortactin in regulating actin assembly in vivo and in vitro. When expressed in cultured cells, a dynamin2 mutant with decreased affinity for GTP decreased actin dynamics within the cortical actin network. Expressed mutants of cortactin that have decreased binding of Arp2/3 complex or dynamin2 also decreased actin dynamics. Dynamin2 influenced actin nucleation by purified Arp2/3 complex and cortactin in vitro in a biphasic manner. Low concentrations of dynamin2 enhanced actin nucleation by Arp2/3 complex and cortactin, and high concentrations were inhibitory. Dynamin2 promoted the association of actin filaments nucleated by Arp2/3 complex and cortactin with phosphatidylinositol 4,5-bisphosphate (PIP2)-containing lipid vesicles. GTP hydrolysis altered the organization of the filaments and the lipid vesicles. We conclude that dynamin2, through an interaction with cortactin, regulates actin assembly and actin filament organization at membranes.


Assuntos
Actinas/metabolismo , Dinamina II/metabolismo , Proteínas dos Microfilamentos/metabolismo , Cortactina , GTP Fosfo-Hidrolases/metabolismo
16.
Curr Biol ; 12(15): 1270-8, 2002 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-12176354

RESUMO

BACKGROUND: Dynamic actin assembly is required for diverse cellular processes and often involves activation of Arp2/3 complex. Cortactin and N-WASp activate Arp2/3 complex, alone or in concert. Both cortactin and N-WASp contain an acidic (A) domain that is required for Arp2/3 complex binding. RESULTS: We investigated how cortactin and the constitutively active VCA domain of N-WASp interact with Arp2/3 complex. Structural studies showed that cortactin is a thin, elongated monomer. Chemical crosslinking studies demonstrated selective interaction of the Arp2/3 binding NTA domain of cortactin (cortactin NTA) with the Arp3 subunit and VCA with Arp3, Arp2, and ARPC1/p40. Cortactin NTA and VCA crosslinking to the Arp3 subunit were mutually exclusive; however, cortactin NTA did not inhibit VCA crosslinking to Arp2 or ARPC1/p40, nor did it inhibit activation of Arp2/3 complex by VCA. We conducted an experiment in which a saturating concentration of cortactin NTA modestly lowered the binding affinity of VCA for Arp2/3; the results of this experiment provided further evidence for ternary complex formation. Consistent with a common binding site on Arp3, a saturating concentration of VCA abolished binding of cortactin to Arp2/3 complex. CONCLUSIONS: Under certain circumstances, cortactin and N-WASp can bind simultaneously to Arp2/3 complex, accounting for their synergy in activation of actin assembly. The interaction of cortactin NTA with Arp2/3 complex does not inhibit Arp2/3 activation by N-WASp, despite competition for a common binding site located on the Arp3 subunit. These results suggest a model in which cortactin may bridge Arp2/3 complex to actin filaments via Arp3 and N-WASp activates Arp2/3 complex by binding Arp2 and/or ARPC1/p40.


Assuntos
Actinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteína 2 Relacionada a Actina , Proteína 3 Relacionada a Actina , Sequência de Aminoácidos , Animais , Sítios de Ligação , Encéfalo/metabolismo , Bovinos , Galinhas , Cortactina , Proteínas do Citoesqueleto/química , Cinética , Proteínas dos Microfilamentos/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Timo/metabolismo , Triptofano , Síndrome de Wiskott-Aldrich , Proteína Neuronal da Síndrome de Wiskott-Aldrich
17.
Curr Biol ; 13(5): 384-93, 2003 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-12620186

RESUMO

BACKGROUND: Modulation of actin cytoskeleton assembly is an integral step in many cellular events. A key regulator of actin polymerization is Arp2/3 complex. Cortactin, an F-actin binding protein that localizes to membrane ruffles, is an activator of Arp2/3 complex. RESULTS: A yeast two-hybrid screen revealed the interaction of the cortactin Src homology 3 (SH3) domain with a peptide fragment derived from a cDNA encoding a region of WASp-Interacting Protein (WIP). GST-cortactin interacted with WIP in an SH3-dependent manner. The subcellular localization of cortactin and WIP coincided at the cell periphery. WIP increased the efficiency of cortactin-mediated Arp2/3 complex activation of actin polymerization in a concentration-dependent manner. Lastly, coexpression of cortactin and WIP stimulated membrane protrusions. CONCLUSIONS: WIP, a protein involved in filopodia formation, binds to both actin monomers and cortactin. Thus, recruitment of actin monomers to a cortactin-activated Arp2/3 complex likely leads to the observed increase in cortactin activation of Arp2/3 complex by WIP. These data suggest that a cortactin-WIP complex functions in regulating actin-based structures at the cell periphery.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteína 2 Relacionada a Actina , Proteína 3 Relacionada a Actina , Actinas/metabolismo , Animais , Células CHO , Cortactina , Cricetinae , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Estrutura Terciária de Proteína
18.
Mol Cell Biol ; 24(6): 2308-17, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993270

RESUMO

Cell adhesion and spreading depend on activation of mitogen-activated kinase, which in turn is regulated both by growth factor and integrin signaling. Growth factors, such as epidermal growth factor, are capable of activating Ras and Raf, but integrin signaling is required to couple Raf to MEK and MEK to extracellular signal-regulated protein kinase (ERK). It was previously shown that Rac-p21-activated kinase (PAK) signaling regulated the physical association of MEK1 with ERK2 through phosphorylation sites in the proline-rich sequence (PRS) of MEK1. It was also shown that activation of MEK1 and ERK by integrins depends on PAK phosphorylation of S298 in the PRS. Here we report a novel MEK1-specific regulatory feedback mechanism that provides a means by which activated ERK can terminate continued PAK phosphorylation of MEK1. Activated ERK can phosphorylate T292 in the PRS, and this blocks the ability of PAK to phosphorylate S298 and of Rac-PAK signaling to enhance MEK1-ERK complex formation. Preventing ERK feedback phosphorylation on T292 during cellular adhesion prolonged phosphorylation of S298 by PAK and phosphorylation of S218 and S222, the MEK1 activating sites. We propose that activation of ERK during adhesion creates a feedback system in which ERK phosphorylates MEK1 on T292, and this in turn blocks additional S298 phosphorylation in response to integrin signaling.


Assuntos
Adesão Celular/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Sítios de Ligação/genética , Células COS , Ativação Enzimática , Retroalimentação , MAP Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/química , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Quinases Ativadas por p21
19.
Mol Biol Cell ; 13(6): 2147-56, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12058076

RESUMO

ASAP1 (ADP ribosylation factor [ARF]- GTPase-activating protein [GAP] containing SH3, ANK repeats, and PH domain) is a phospholipid-dependent ARF-GAP that binds to and is phosphorylated by pp60(Src). Using affinity chromatography and yeast two-hybrid interaction screens, we identified ASAP1 as a major binding partner of protein tyrosine kinase focal adhesion kinase (FAK). Glutathione S-transferase pull-down and coimmunoprecipitation assays showed the binding of ASAP1 to FAK is mediated by an interaction between the C-terminal SH3 domain of ASAP1 with the second proline-rich motif in the C-terminal region of FAK. Transient overexpression of wild-type ASAP1 significantly retarded the spreading of REF52 cells plated on fibronectin. In contrast, overexpression of a truncated variant of ASAP1 that failed to bind FAK or a catalytically inactive variant of ASAP1 lacking GAP activity resulted in a less pronounced inhibition of cell spreading. Transient overexpression of wild-type ASAP1 prevented the efficient organization of paxillin and FAK in focal adhesions during cell spreading, while failing to significantly alter vinculin localization and organization. We conclude from these studies that modulation of ARF activity by ASAP1 is important for the regulation of focal adhesion assembly and/or organization by influencing the mechanisms responsible for the recruitment and organization of selected focal adhesion proteins such as paxillin and FAK.


Assuntos
Proteínas de Transporte/metabolismo , Adesões Focais/fisiologia , Proteínas Tirosina Quinases/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/genética , Cromatografia de Afinidade , Clonagem Molecular , Primers do DNA , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Cinética , Camundongos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Reação em Cadeia da Polimerase , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Dedos de Zinco
20.
Mol Biol Cell ; 14(8): 3216-29, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12925758

RESUMO

Cortactin is an F-actin binding protein that activates actin-related protein 2/3 complex and is localized within lamellipodia. Cortactin is a substrate for Src and other protein tyrosine kinases involved in cell motility, where its phosphorylation on tyrosines 421, 466, and 482 in the carboxy terminus is required for cell movement and metastasis. In spite of the importance of cortactin tyrosine phosphorylation in cell motility, little is known regarding the structural, spatial, or signaling requirements regulating cortactin tyrosine phosphorylation. Herein, we report that phosphorylation of cortactin tyrosine residues in the carboxy terminus requires the aminoterminal domain and Rac1-mediated localization to the cell periphery. Phosphorylation-specific antibodies directed against tyrosine 421 and 466 were produced to study the regulation and localization of tyrosine phosphorylated cortactin. Phosphorylation of cortactin tyrosine 421 and 466 was elevated in response to Src, epidermal growth factor receptor and Rac1 activation, and tyrosine 421 phosphorylated cortactin localized with F-actin in lamellipodia and podosomes. Cortactin tyrosine phosphorylation is progressive, with tyrosine 421 phosphorylation required for phosphorylation of tyrosine 466. These results indicate that cortactin tyrosine phosphorylation requires Rac1-induced cortactin targeting to cortical actin networks, where it is tyrosine phosphorylated in hierarchical manner that is closely coordinated with its ability to regulate actin dynamics.


Assuntos
Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Pseudópodes/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Cortactina , Camundongos , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Transdução de Sinais/fisiologia , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA