Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(13): 1643-56, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26165839

RESUMO

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


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Estrogênios/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Supressoras de Tumor/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células MCF-7 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo
2.
Oncogene ; 33(4): 411-20, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23318423

RESUMO

Overexpression of the NEDD9/HEF1/Cas-L scaffolding protein is frequent, and drives invasion and metastasis in breast, head and neck, colorectal, melanoma, lung and other types of cancer. We have examined the consequences of genetic ablation of Nedd9 in the MMTV-HER2/ERBB2/neu mouse mammary tumor model. Unexpectedly, we found that only a limited effect on metastasis in MMTV-neu;Nedd9(-/-) mice compared with MMTV-neu;Nedd9(+/+) mice, but instead a dramatic reduction in tumor incidence (18 versus 80%), and a significantly increased latency until tumor appearance. Orthotopic reinjection and tail-vein injection of cells arising from tumors, coupled with in vivo analysis, indicated tumors arising in MMTV-neu;Nedd9(-/-) mice had undergone mutational selection that overcame the initial requirement for Nedd9. To better understand the defects in early tumor growth, we compared mammary progenitor cell pools from MMTV-neu;Nedd9(-/-) versus MMTV-neu;Nedd9(+/+) mice. The MMTV-neu;Nedd9(-/-) genotype selectively reduced both the number and colony-forming potential of mammary luminal epithelial progenitor cells, while not affecting basal epithelial progenitors. MMTV-neu;Nedd9(-/-) mammospheres had striking defects in morphology and cell polarity. All of these defects were seen predominantly in the context of the HER2/neu oncogene, and were not associated with randomization of the plane of mitotic division, but rather with depressed expression the cell attachment protein FAK, accompanied by increased sensitivity to small molecule inhibitors of FAK and SRC. Surprisingly, in spite of these significant differences, only minimal changes were observed in the gene expression profile of Nedd9(-/-) mice, indicating critical Nedd9-dependent differences in cell growth properties were mediated via post-transcriptional regulation of cell signaling. Coupled with emerging data indicating a role for NEDD9 in progenitor cell populations during the morphogenesis of other tissues, these results indicate a functional requirement for NEDD9 in the growth of mammary cancer progenitor cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinogênese/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Invasividade Neoplásica/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinogênese/genética , Feminino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo , Camundongos , Camundongos Knockout , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia
3.
Oncogene ; 33(5): 539-49, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23334327

RESUMO

Aurora kinase A (AURKA) localizes to centrosomes and mitotic spindles where it mediates mitotic progression and chromosomal stability. Overexpression of AURKA is common in cancer, resulting in acquisition of alternate non-mitotic functions. In the current study, we identified a novel role for AURKA in regulating ovarian cancer cell dissemination and evaluated the efficacy of an AURKA-selective small molecule inhibitor, alisertib (MLN8237), as a single agent and combined with paclitaxel using an orthotopic xenograft model of epithelial ovarian cancer (EOC). Ovarian carcinoma cell lines were used to evaluate the effects of AURKA inhibition and overexpression on migration and adhesion. Pharmacological or RNA interference-mediated inhibition of AURKA significantly reduced ovarian carcinoma cell migration and adhesion and the activation-associated phosphorylation of the cytoskeletal regulatory protein SRC at tyrosine 416 (pSRC(Y416)). Conversely, enforced expression of AURKA resulted in increased migration, adhesion and activation of SRC in cultured cells. In vivo tumor growth and dissemination were inhibited by alisertib treatment as a single agent. Moreover, combination of alisertib with paclitaxel, an agent commonly used in treatment of EOC, resulted in more potent inhibition of tumor growth and dissemination compared with either drug alone. Taken together, these findings support a role for AURKA in EOC dissemination by regulating migration and adhesion. They also point to the potential utility of combining AURKA inhibitors with taxanes as a therapeutic strategy for the treatment of EOC patients.


Assuntos
Aurora Quinase A/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Azepinas/farmacologia , Carcinoma Epitelial do Ovário , Adesão Celular , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Camundongos , Mitose/efeitos dos fármacos , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias Epiteliais e Glandulares/enzimologia , Neoplasias Ovarianas/enzimologia , Paclitaxel/farmacologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/metabolismo
4.
Br J Cancer ; 109(4): 1063-71, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23839489

RESUMO

BACKGROUND: Dual specificity phosphatase 6 (DUSP6) is a member of a family of mitogen-activated protein kinase phosphatases that dephosphorylates and inhibits activated ERK1/2. Dual specificity phosphatase 6 is dynamically regulated in developmental and pathological conditions such as cancer. METHODS: Cancer cell lines were made deficient in DUSP6 by siRNA and shRNA silencing. Sensitivity to anti-EGFR and chemotherapeutic agents was determined in viability and apoptosis assays, and in xenografts established in SCID mice. Cellular effects of DUSP6 inactivation were analysed by proteomic methods, followed by analysis of markers of DNA damage response (DDR) and cell cycle. RESULTS: We determined that depletion of DUSP6 reduced the viability of cancer cell lines and increased the cytotoxicity of EGFR and other targeted inhibitors, and cytotoxic agents, in vitro and in vivo. Subsequent phosphoproteomic analysis indicated DUSP6 depletion significantly activated CHEK2 and p38, which function in the DDR pathway, and elevated levels of phosphorylated H2AX, ATM, and CHEK2, for the first time identifying a role for DUSP6 in regulating DDR. CONCLUSION: Our results provide a novel insight into the DUSP6 function in regulating genomic integrity and sensitivity to chemotherapy in cancer.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Fosfatase 6 de Especificidade Dupla/fisiologia , Receptores ErbB/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quinase do Ponto de Checagem 2 , Células HEK293 , Humanos , Marcação In Situ das Extremidades Cortadas , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos SCID , Fosforilação , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteômica , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Br J Cancer ; 106(4): 748-55, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22240781

RESUMO

BACKGROUND: Increased Aurora kinase A gene copy number (AURKA-CN) has been reported in metastatic colorectal cancer (mCRC), with unknown relationship to clinical outcome. We correlated increased AURKA-CN in mCRC tumours with KRAS mutation status, overall and progression-free survival (OS, PFS). METHODS: Sixty-one mCRC tumours were analysed for AURKA-CN using q-PCR, and KRAS mutation status by direct sequencing. Expression of AURKA protein was analysed by immunohistochemistry. Cox-proportional hazard method, Kaplan-Meier curves and log-rank statistics were used to estimate and compare the hazard ratios and median survival between the groups. RESULTS: In all, 68% of tumour exhibited high AURKA-CN, and 29% had a KRAS mutation, without correlation between the two. Patients with high AURKA-CN tumours had longer median OS (48.6 vs 18.8 months, P=0.01), with stronger trend among KRAS wild-type tumours (median OS not reached vs 18.8 months, P=0.003). Progression-free survival was longer on first-line or second-line chemotherapy among patients with KRAS wild-type and high vs low AURKA-CN (first: 17.6 vs 5.13 months, P=0.04; second: 10.4 vs 5.1 months, P=0.01). AURKA-CN level did not affect outcomes among patients with KRAS mutant tumours. CONCLUSION: Increased AURKA-CN is common in mCRC tumours and is associated with longer OS and longer PFS during chemotherapy, particularly in KRAS wild-type tumours.


Assuntos
Neoplasias Colorretais/genética , Dosagem de Genes , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Aurora Quinase A , Aurora Quinases , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Metástase Neoplásica , Prognóstico , Proteínas Proto-Oncogênicas p21(ras)
6.
Oncogene ; 31(10): 1217-27, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21785464

RESUMO

Increased activity of SRC family kinases promotes tumor invasion and metastasis, and overexpression of the mitotic regulator Aurora kinase A (AURKA) drives tumor aneuploidy and chromosomal instability. These functions nominate SRC and AURKA as valuable therapeutic targets for cancer, and inhibitors for SRC and Aurora kinases are now being used in the clinic. In this study, we demonstrate potent synergy between multiple inhibitors of Aurora and SRC kinases in ovarian and colorectal cancer cell lines, but not in normal ovarian epithelial cell lines. Combination of Aurora and SRC inhibitors selectively killed cells that have undergone a preceding aberrant mitosis, and was associated with a postmitotic reattachment defect, and selective removal of aneuploid cell populations. Combined inhibition of Aurora kinase and SRC potentiated dasatinib-dependent loss of activated (Y(416)-phosphorylated) SRC. SRC and AURKA share a common interaction partner, NEDD9, which serves as a scaffolding protein with activities in cell attachment and mitotic control, suggesting SRC and AURKA might interact directly. In vitro, we observed physical interaction and mutual cross-phosphorylation between SRC and AURKA that enhanced SRC kinase activity. Together, these findings suggest that combination of SRC and Aurora-targeting inhibitors in the clinic may be a productive strategy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinases da Família src/antagonistas & inibidores , Aurora Quinase A , Aurora Quinases , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dasatinibe , Feminino , Humanos , Mitose/efeitos dos fármacos , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Tiazóis/farmacologia , Quinases da Família src/fisiologia
7.
Oncogene ; 30(23): 2633-43, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21317929

RESUMO

Misregulation of the canonical Wnt/ß-catenin pathway and aberrant activation of Wnt signaling target genes are common in colorectal cancer (CRC) and contribute to cancer progression. Altered expression of human enhancer of filamentation 1 (HEF1; also known as NEDD9 or Cas-L) has been implicated in progression of melanoma, breast, and CRC. However, the regulation of HEF1 and the role of HEF1 in CRC tumorigenesis are not fully understood. We here identify HEF1 as a novel Wnt signaling target. The expression of HEF1 was upregulated by Wnt-3a, ß-catenin, and Dvl2 in a dose-dependent manner, and was suppressed following ß-catenin downregulation by shRNA. In addition, elevated HEF1 mRNA and protein levels were observed in CRC cell lines and primary tumor tissues, as well as in the colon and adenoma polyps of Apc(Min/+) mice. Moreover, HEF1 levels in human colorectal tumor tissues increased with the tumor grade. Chromatin immunoprecipitation (ChIP) assays and promoter analyses revealed three functional T-cell factor (TCF)-binding sites in the promoter of HEF1 responsible for HEF1 induction by Wnt signaling. Ectopic expression of HEF1 increased cell proliferation and colony formation, while downregulation of HEF1 in SW480 cells by shRNA had the opposite effects and inhibited the xenograft tumor growth. Furthermore, overexpression of HEF1 in SW480 cells promoted cell migration and invasion. Together, our results determined a novel role of HEF1 as a mediator of the canonical Wnt/ß-catenin signaling pathway for cell proliferation, migration, and tumorigenesis, as well as an important player in colorectal tumorigenesis and progression. HEF1 may represent an attractive candidate for drug targeting in CRC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Colo/metabolismo , Neoplasias Colorretais/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Colo/citologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Nus , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fosfoproteínas/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição TCF/metabolismo , Transplante Heterólogo , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
8.
Neuroscience ; 162(4): 1106-19, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19464348

RESUMO

Cell migration is essential for the development of numerous structures derived from embryonic neural crest cells (NCCs), however the underlying molecular mechanisms are incompletely understood. NCCs migrate long distances in the embryo and contribute to many different cell types, including peripheral neurons, glia and pigment cells. In the present work we report expression of Nedd9, a scaffolding protein within the integrin signaling pathway, in non-lineage-restricted neural crest progenitor cells. In particular, Nedd9 was found to be expressed in the dorsal neural tube at the time of neural crest delamination and in early migrating NCCs. To analyze the role of Nedd9 in neural crest development we performed loss- and gain-of-function experiments and examined the subsequent effects on delamination and migration in vitro and in vivo. Our results demonstrate that loss of Nedd9 activity in chick NCCs perturbs cell spreading and the density of focal complexes and actin filaments, properties known to depend on integrins. Moreover, a siRNA dose-dependent decrease in Nedd9 activity results in a graded reduction of NCC's migratory distance while forced overexpression increases it. Retinoic acid (RA) was found to regulate Nedd9 expression in NCCs. Our results demonstrate in vivo that Nedd9 promotes the migration of NCCs in a graded manner and suggest a role for RA in the control of Nedd9 expression levels.


Assuntos
Actinas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Movimento Celular , Crista Neural/citologia , Células-Tronco/fisiologia , Tretinoína/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Adesão Celular , Embrião de Galinha , Feminino , Regulação da Expressão Gênica , Camundongos , Crista Neural/metabolismo , Gravidez , RNA Interferente Pequeno/genética , Tretinoína/farmacologia
9.
Oncogene ; 26(33): 4825-32, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17297447

RESUMO

Human enhancer of invasion, clone 10 (HEI10) (CCNB1IP1) was first described as a RING-finger family ubiquitin ligase that regulates cell cycle by interacting with cyclin B and promoting its degradation. Subsequently, other studies suggested specific upregulation of HEI10 in metastatic melanoma and demonstrated direct interaction between HEI10 and the tumor suppressor Merlin, encoded by the neurofibromatosis 2 gene. These and other results led us to hypothesize that HEI10 also influences the processes of cell migration and metastasis. We here show that cells with depleted HEI10 both migrate more rapidly and invade more effectively than control cells. HEI10 depletion post-transcriptionally increases the expression of a group of promotility regulatory proteins including p130Cas, paxillin, Cdk1 and cyclin B2, but excluding Merlin. Among these, only inhibition of Cdk1/cyclin B activity specifically reversed the motility and invasion of HEI10-depleted cells. Finally, HEI10 is abundantly transcribed in many human tissues, and particularly abundant in some tumor cell lines, suggesting that it may be commonly involved in coordinating cell cycle with cell migration and invasion.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimento Celular/fisiologia , Ciclina B/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Proteína Quinase CDC2/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Cisplatino/farmacologia , Proteína Substrato Associada a Crk/metabolismo , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Paxilina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Interferência de RNA , Processamento Pós-Transcricional do RNA , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Roscovitina , Fatores de Tempo , Ubiquitina-Proteína Ligases , Proteína cdc42 de Ligação ao GTP/metabolismo
10.
Oncogene ; 25(32): 4389-98, 2006 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-16532029

RESUMO

Merlin and ezrin are homologous proteins with opposite effects on neoplastic growth. Merlin is a tumor suppressor inactivated in the neurofibromatosis 2 disease, whereas upregulated ezrin expression is associated with increased malignancy. Merlin's tumor suppressor mechanism is not known, although participation in cell cycle regulation has been suggested. To characterize merlin's biological activities, we screened for molecules that would interact with merlin but not ezrin. We identified the cyclin B-binding protein and cell cycle regulator HEI10 as a novel merlin-binding partner. The interaction is mediated by the alpha-helical domain in merlin and the coiled-coil domain in HEI10 and requires conformational opening of merlin. The two proteins show partial subcellular colocalization, which depends on cell cycle stage and cell adhesion. Comparison of Schwann cells and schwannoma cultures demonstrated that the distribution of HEI10 depends on merlin expression. In transfected cells, a constitutively open merlin construct affected HEI10 protein integrity. These results link merlin to the cell cycle control machinery and may help to understand its tumor suppressor function.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclo Celular/fisiologia , Neurofibromina 2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Neurofibromina 2/biossíntese , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Ratos , Ubiquitina-Proteína Ligases
11.
Oncogene ; 25(12): 1721-32, 2006 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-16288224

RESUMO

The highly invasive behavior of glioblastoma cells contributes to the morbidity and mortality associated with these tumors. The integrin-mediated adhesion and migration of glioblastoma cells on brain matrix proteins is enhanced by stimulation with growth factors, including platelet-derived growth factor (PDGF). As focal adhesion kinase (FAK), a nonreceptor cytoplasmic tyrosine kinase, has been shown to promote cell migration in various other cell types, we analysed its role in glioblastoma cell migration. Forced overexpression of FAK in serum-starved glioblastoma cells plated on recombinant (rec)-osteopontin resulted in a twofold enhancement of basal migration and a ninefold enhancement of PDGF-BB-stimulated migration. Both expression of mutant FAK(397F) and the downregulation of FAK with small interfering (si) RNA inhibited basal and PDGF-stimulated migration. FAK overexpression and PDGF stimulation was found to increase the phosphorylation of the Crk-associated substrate (CAS) family member human enhancer of filamentation 1 (HEF1), but not p130CAS or Src-interacting protein (Sin)/Efs, although the levels of expression of these proteins was similar. Moreover downregulation of HEF1 with siRNA, but not p130CAS, inhibited basal and PDGF-stimulated migration. The phosphorylated HEF1 colocalized with vinculin and was associated almost exclusively with 0.1% Triton X-100 insoluble material, consistent with its signaling at focal adhesions. FAK overexpression promoted invasion through normal brain homogenate and siHEF1 inhibited this invasion. Results presented here suggest that HEF1 acts as a necessary and specific downstream effector of FAK in the invasive behavior of glioblastoma cells and may be an effective target for treatment of these tumors.


Assuntos
Neoplasias Encefálicas/patologia , Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Glioblastoma/patologia , Invasividade Neoplásica/patologia , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Humanos , Immunoblotting , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , RNA Interferente Pequeno
13.
J Biol Chem ; 276(44): 40606-13, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11514549

RESUMO

p21-activated kinase 1 (Pak1) is an effector for the small GTPases Cdc42 and Rac. Because Pak1 binds to and is activated by both these GTPases, it has been difficult to precisely delineate the signaling pathways that link extracellular stimuli to Pak1 activation. To separate activation of Pak1 by Cdc42 versus activation by Rac, we devised a genetic screen in yeast that enabled us to create and identify Pak1 mutants that selectively couple to Cdc42 but not Rac1. We recovered several such Pak1 mutants and found that the residues most often affected lie within the p21 binding domain, a region previously known to mediate Pak1 binding to GTPases, but that several mutations also map outside the borders of the p21 binding domain. Pak1 mutants that associate with Cdc42 but not Rac1 were also activated by Cdc42 but not Rac1. In rat 3Y1 cells expressing oncogenic Ha-Ras, the Pak1 mutants defective in Rac1 binding are not activated, suggesting that Ras signals through a GTPase other than Cdc42 to activate Pakl. Similar results were obtained when epidermal growth factor was used to activate Pak1. However, Pak1 mutants that are unable to bind Rac are nonetheless well activated by calf serum, implying that this stimulus may induce Pak activation independent of Rac.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Mutagênese , Testes de Precipitina , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Quinases Ativadas por p21 , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
Eur J Immunol ; 31(5): 1417-27, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11465098

RESUMO

Although beta1 integrin-dependent T cell migration is required for immune function, little is known of the signaling pathways regulating this migration. We now show that the cytoplasmic tyrosine kinase, focal adhesion kinase (FAK) plays an essential role in the beta1 integrin-stimulated migration of T cells through regulation of the unique Crk-associated substrate (Cas) family docking protein, human enhancer of filamentation 1 (HEF1) and effects on "outside-in" beta1 integrin signaling. Overexpression of wild-type FAK promoted beta1 integrin-dependent Jurkat T cell migration, whereas FAK mutated in either its autophosphorylation site or proline rich region 1 (PR1)/HEF1 SH3 domain-binding site had a dominant negative effect on migration. In contrast, neither wild-type nor mutant FAK affected Jurkat cell adhesion to fibronectin, a beta1 integrin ligand. The migration of FAK-overexpressing cells directly correlated with the beta1 integrin-inducible tyrosine phosphorylation of endogenous plus wild-type exogenous FAK, and not with phosphorylation of the FAK-related kinase, Pyk2. FAK was also found to regulate both HEF1-promoted migration, and HEF1 tyrosine phosphorylation in beta1 integrin-stimulated cells, in a manner dependent upon the FAK autophosphorylation and PR1 sites, and HEF1 SH3 domain. Together, our results indicate that beta1 integrin-stimulated T cell migration requires a linear beta1 integrin-FAK-HEF1 effector pathway.


Assuntos
Movimento Celular , Integrina beta1/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Substituição de Aminoácidos/genética , Adesão Celular , Células Clonais/enzimologia , Células Clonais/metabolismo , Fibronectinas/metabolismo , Citometria de Fluxo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Células HeLa , Humanos , Células Jurkat , Modelos Biológicos , Mutação/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/genética , Linfócitos T/enzimologia , Transfecção
15.
Mol Cell Biol ; 21(15): 5094-108, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11438665

RESUMO

The dynamic regulation of focal adhesions is implicated in cellular processes of proliferation, differentiation, migration, and apoptosis. The focal adhesion-associated docking protein HEF1 is cleaved by caspases during both mitosis and apoptosis. Common to both of these cellular processes is the loss of focal adhesions, transiently during mitosis and permanently during apoptosis. The proteolytic processing of HEF1 during both mitosis and apoptosis therefore posits a general role for HEF1 as a sensor of altered adhesion states. In this study, we find that HEF1 undergoes proteolytic processing specifically in response to cellular detachment, while HEF1 proteolysis is prevented by specific integrin receptor ligation and focal adhesion formation. We show that overexpression of a C-terminal caspase-derived 28-kDa HEF1 peptide causes cellular rounding that is demonstrably separable from apoptosis. Mutation of the divergent helix-loop-helix motif found in 28-kDa HEF1 significantly reduces the induction of apoptosis by this peptide, while deletion of the amino-terminal 28 amino acids of 28-kDa HEF1 completely abrogates the induction of apoptosis. Conversely, these mutations have no effect on the rounding induced by 28-kDa HEF1. Finally, we detect a novel focal adhesion targeting domain located in the C terminus of HEF1 and show that this activity is necessary for HEF1-induced cell spreading. Together, these data suggest that proteolytic and other posttranslational modifications of HEF1 in response to loss of adhesion serve to modulate the disassembly of focal adhesions.


Assuntos
Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Clorometilcetonas de Aminoácidos/metabolismo , Sequência de Aminoácidos , Apoptose , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Adesão Celular , Técnicas de Cultura de Células , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Adesões Focais/metabolismo , Deleção de Genes , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Mitose , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Peptídeos/química , Fenótipo , Fosfoproteínas/química , Fosfoproteínas/genética , Testes de Precipitina , Ligação Proteica , Isoformas de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
17.
Methods ; 24(3): 201-17, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11403570

RESUMO

The detection of physical interaction between two or more molecules of interest can be facilitated if the act of association between the interactive partners leads to the production of a readily observed biological or physical readout. Many interacting molecule pairs (X, Y) can be made to induce such a readout if X and Y are each fused to defined protein elements with desired properties. For example, in the yeast forward two-hybrid system, X is synthesized as a translational fusion to a DNA-binding domain (DBD), Y is synthesized as a fusion to a transcriptional activation domain (AD), and coexpression of DBD-X and AD-Y induces transcription of easily scored responsive reporters. Other approaches use paradigms based on the artificial production of two, hybrid, molecules, but substitute a variety of readouts including the repression of transcription, activation of signal transduction pathways, or reconstitution of a disrupted enzymatic activity. In this article, we summarize a number of two-hybrid-based approaches, and detail the use of the forward yeast two-hybrid system in a screen to identify novel interacting partners for a protein of interest.


Assuntos
Proteínas/metabolismo , Saccharomyces cerevisiae , Técnicas do Sistema de Duplo-Híbrido , DNA Recombinante/química , DNA Recombinante/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Biblioteca Gênica , Vetores Genéticos/metabolismo , Técnicas In Vitro , Biblioteca de Peptídeos , Plasmídeos/metabolismo , Proteínas/química , Proteínas/genética , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/genética , Transcrição Gênica/fisiologia , Transformação Genética/fisiologia
20.
Biotechniques ; 30(3): 634-6, 638, 640 passim, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11252799

RESUMO

Examination of the pattern of reagent creation and application in the two-hybrid system since 1989 reveals the expansion of a simple core technology to address increasingly sophisticated problems in protein interaction. As the technology has matured, its clear suitability for large-scale proteomic projects has made a major focus of its application the generation of global organismal protein interaction networks. In an inversion of emphasis, the increasing availability of such information now provides a master plan with the potential to specify the most promising directions for biological investigations (i.e., by directing the physiological validation of predicted critical protein-protein interactions). Recent derivatives of the two-hybrid system enable the targeting of such key interactions by facilitating the identification of essential amino acids conferring protein interaction specificity and of small molecules that selectively disrupt defined interaction pairs. Finally, the creation of mammalian expression systems based on two-hybrid principles became a new tool to create and probe novel biological systems. Taken in sum, this trajectory emphasizes the point that the creation of tools and the evolution of the idea of what is an interesting biological problem are in intimate dialogue.


Assuntos
Saccharomyces cerevisiae/genética , Técnicas do Sistema de Duplo-Híbrido , Farmacogenética , Proteoma , RNA/metabolismo , Proteínas Recombinantes de Fusão/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...