Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Behav Sci (Basel) ; 14(5)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38785859

RESUMO

This study examined the associations between maternal involvement in education and bicultural acceptance and school adjustment during the first year of middle school among adolescents from Korean multicultural families as well as the reciprocal relationships between bicultural acceptance and school adjustment during the three years of middle school. The present study used three-wave longitudinal data of 1185 dyads of adolescents (50.8% girls; mean age = 12.96 ± 0.35 years at the first wave) and their immigrant mothers (mean age = 43.54 ± 5.19 years at the first wave), who participated in the Multicultural Adolescents Panel Study. An autoregressive cross-lagged modeling analysis revealed that maternal involvement in education was significantly and positively associated with adolescents' bicultural acceptance and school adjustment in the first year of middle school. Individual levels of bicultural acceptance and school adjustment among adolescents remained moderately stable over the three years. Whereas the positive effects of school adjustment on bicultural acceptance were significant over time, the effects of bicultural acceptance on school adjustment were not. Finally, this study highlights the roles of intervention programs (e.g., parent and multicultural education) in facilitating maternal involvement in education and school adjustment as well as in increasing bicultural acceptance among minority youths.

2.
Int Neurourol J ; 27(Suppl 2): S51-63, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38048819

RESUMO

In recent years, advancements in information and communication technologies, including artificial intelligence, big data, virtual reality, and augmented reality, have driven substantial growth in the field of digital medical diagnosis and treatment, thereby enhancing quality of life. Beginning in the mid-2010s with the advent of digital healthcare applications, and further accelerated by the impact of coronavirus disease 2019, digital therapeutic products have profoundly influenced society. Nevertheless, the expansion of digital therapeutics has encountered challenges associated with regulatory hurdles, differentiation from general digital healthcare, and the necessity for trustworthiness, which have contributed to a slower rate of progress. This study proposes a 3P content model-encompassing pre-education, prediction/diagnosis/treatment, and postmanagement-to increase the trustworthiness of digital therapeutics. The design of the 3P content model includes a fundamental structure that establishes networks with healthcare institutions, aiming to increase the reliability of data utilization and to facilitate integration with medical decision support systems. For case development, the study introduces a prototype of a mobile application that utilizes chronic disease urinary dysfunction data, demonstrating the cyclical structure inherent in the 3P content model.

3.
J Immunother Cancer ; 11(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37848261

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a type of leukemia in adults with a high mortality rate and poor prognosis. Although targeted therapeutics, chemotherapy, and hematopoietic stem cell transplantation can improve the prognosis, the recurrence rate is still high, with a 5-year survival rate of approximately 40%. This study aimed to develop an IgG-based asymmetric bispecific antibody that targets CLL-1 and CD3 for treating AML. METHODS: ABL602 candidates were compared in terms of binding activity, T-cell activation, and tumor-killing activities. ABL602-mediated T-cell activation and tumor-killing activities were determined by measuring the expression of activation markers, cytokines, cytolytic proteins, and the proportion of dead cells. We evaluated in vivo tumor growth inhibitory activity in two mouse models bearing subcutaneously and orthotopically engrafted human AML. Direct tumor-killing activity and T-cell activation in patient-derived AML blasts were also evaluated. RESULTS: ABL602 2+1 showed a limited CD3 binding in the absence of CLL-1, suggesting that steric hindrance on the CD3 binding arm could reduce CLL-1 expression-independent CD3 binding. Although the CD3 binding activity was attenuated compared with that of 1+1, ABL602 2+1 exhibited much stronger T-cell activation and potent tumor-killing activities in AML cell lines. ABL602 2+1 efficiently inhibited tumor progression in subcutaneously and orthotopically engrafted AML mouse models. In the orthotopic mouse model, tumor growth inhibition was observed by gross measurement of luciferase activity, as well as a reduced proportion of AML blasts in the bone marrow, as determined by flow cytometry and immunohistochemistry (IHC) staining. ABL602 2+1 efficiently activated T cells and induced the lysis of AML blasts, even at very low effector:target (E:T) ratios (eg, 1:50). Compared with the reference 1+1 antibody, ABL602 did not induce the release of cytokines including interleukin-6 and tumor necrosis factor-α in the healthy donor-derived peripheral blood mononuclear cell. CONCLUSIONS: With its potent tumor-killing activity and reduced cytokine release, ABL602 2+1 is a promising candidate for treating patients with AML and warrants further study.


Assuntos
Anticorpos Biespecíficos , Leucemia Linfocítica Crônica de Células B , Leucemia Mieloide Aguda , Camundongos , Adulto , Animais , Humanos , Citocinas/metabolismo , Leucócitos Mononucleares , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico
4.
Children (Basel) ; 10(6)2023 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-37371241

RESUMO

In Korea, marriages between Korean men and foreign women have surged since the late 1990s, resulting in public interest in the psychosocial adjustment of their children. This study examined the mediating effects of immigrant mothers' school involvement, adolescents' school adjustment, and bicultural acceptance on the relationship between the mothers' acculturative stress and adolescents' depression, as well as whether the structural relationships differed by the sex of adolescents. Data were collected from 1238 dyads of first-grade students (605 boys, 633 girls; age = 12.97 ± 0.35 years) in Korean middle schools and their immigrant mothers (age = 43.52 ± 5.13 years) who participated in the Multicultural Adolescents Panel Study. The structural equation modeling analysis revealed that mothers' acculturative stress was indirectly and positively related to adolescents' depression through (1) the serial mediations of mothers' school involvement and adolescents' school adjustment and (2) the individual mediation of adolescents' school adjustment. Furthermore, the multigroup analysis indicated that the relationships between adolescents' school adjustment and depression and between adolescents' bicultural acceptance and depression significantly differed between male and female adolescents. The study provides directions for schools and communities to increase immigrant mothers' school involvement and to facilitate their children's school adjustment and bicultural acceptance.

5.
J Immunother Cancer ; 9(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34230109

RESUMO

BACKGROUND: Stimulation of 4-1BB with agonistic antibodies is a promising strategy for improving the therapeutic efficacy of immune checkpoint inhibitors (ICIs) or for overcoming resistance to ICIs. However, dose-dependent hepatotoxicity was observed in clinical trials with monoclonal anti-4-1BB agonistic antibodies due to the activation of 4-1BB signaling in liver resident Kupffer cells. METHODS: To avoid this on-target liver toxicity, we developed a novel bispecific antibody (4-1BB×PD-L1 bispecific antibody, termed "ABL503") uniquely designed to activate 4-1BB signaling only in the context of PD-L1, while also blocking PD-1/PD-L1 signaling. RESULTS: Functional evaluation using effector cells expressing both 4-1BB and PD-1 revealed superior biological activity of ABL503 compared with the combination of each monoclonal antibody. ABL503 also augmented T-cell activation in in vitro assays and further enhanced the anti-PD-L1-mediated reinvigoration of tumor-infiltrating CD8+ T cells from patients with cancer. Furthermore, in humanized PD-L1/4-1BB transgenic mice challenged with huPD-L1-expressing tumor cells, ABL503 induced superior anti-tumor activity and maintained an anti-tumor response against tumor rechallenge. ABL503 was well tolerated, with normal liver function in monkeys. CONCLUSION: The novel anti-4-1BB×PD-L1 bispecific antibody may exert a strong anti-tumor therapeutic efficacy with a low risk of liver toxicity through the restriction of 4-1BB stimulation in tumors.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Masculino , Camundongos
6.
Cancers (Basel) ; 12(11)2020 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-33142709

RESUMO

We aimed to evaluate the preclinical efficacy of GC1118, a novel anti-epidermal growth factor receptor (EGFR) monoclonal antibody (mAb), against glioblastoma (GBM) tumors using patient-derived xenograft (PDX) models. A total of 15 distinct GBM PDX models were used to evaluate the therapeutic efficacy of GC1118. Genomic data derived from PDX models were analyzed to identify potential biomarkers associated with the anti-tumor efficacy of GC1118. A patient-derived cell-based high-throughput drug screening assay was performed to further validate the efficacy of GC1118. Compared to cetuximab, GC1118 exerted comparable growth inhibitory effects on the GBM tumors in the PDX models. We confirmed that GC1118 accumulated within the tumor by crossing the blood-brain barrier in in vivo specimens and observed the survival benefit in GC1118-treated intracranial models. Genomic analysis revealed high EGFR amplification as a potent biomarker for predicting the therapeutic efficacy of GC1118 in GBM tumors. In summary, GC1118 exerted a potent anti-tumor effect on GBM tumors in PDX models, and its therapeutic efficacy was especially pronounced in the tumors with high EGFR amplification. Our study supports the importance of patient stratification based on EGFR copy number variation in clinical trials for GBM. The superiority of GC1118 over other EGFR mAbs in GBM tumors should be assessed in future studies.

7.
Biomolecules ; 10(3)2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32143496

RESUMO

As mesothelin is overexpressed in various types of cancer, it is an attractive target for therapeutic antibodies. T-cell bispecific antibodies bind to target cells and engage T cells via binding to CD3, resulting in target cell killing by T-cell activation. However, the affinity of the CD3-binding arm may influence CD3-mediated plasma clearance or antibody trapping in T-cell-containing tissues. This may then affect the biodistribution of bispecific antibodies. In this study, we used scFab and knob-into-hole technologies to construct novel IgG-based 1 + 1 MG1122-A and 2 + 1 MG1122-B bispecific antibodies against mesothelin and CD3ε. MG1122-B was designed to be bivalent to mesothelin and monovalent to CD3ε, using a 2 + 1 head-to-tail format. Activities of the two antibodies were evaluated in mesothelin-positive tumor cells in vitro and xenograft models in vivo. Although both antibodies exhibited target cell killing efficacy and produced regression of xenograft tumors with CD8+ T-cell infiltration, the antitumor efficacy of MG1122-B was significantly higher. MG1122-B may improve tumor targeting because of its bivalency for tumor antigen. It may also reduce systemic toxicity by limiting the activation of circulating T cells. Thus, MG1122-B may be useful for treating mesothelin-positive solid tumors.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos Imunológicos , Proteínas Ligadas por GPI/imunologia , Imunoglobulina G , Proteínas de Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacologia , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Células Jurkat , Mesotelina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Mol Sci ; 20(23)2019 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-31771279

RESUMO

Epidermal growth factor receptor (EGFR)-targeted monoclonal antibodies, including cetuximab and panitumumab, are used to treat metastatic colorectal cancer (mCRC). However, this treatment is only effective for a small subset of mCRC patients positive for the wild-type KRAS GTPase. GC1118 is a novel, fully humanized anti-EGFR IgG1 antibody that displays potent inhibitory effects on high-affinity EGFR ligand-induced signaling and enhanced antibody-mediated cytotoxicity. In this study, using 51 CRC patient-derived xenografts (PDXs), we showed that KRAS mutants expressed remarkably elevated autocrine levels of high-affinity EGFR ligands compared with wild-type KRAS. In three KRAS-mutant CRCPDXs, GC1118 was more effective than cetuximab, whereas the two agents demonstrated comparable efficacy against three wild-type KRAS PDXs. Persistent phosphatidylinositol-3-kinase (PI3K)/AKT signaling was thought to underlie resistance to GC1118. In support of these findings, a preliminary improved anti-cancer response was observed in a CRC PDX harboring mutated KRAS with intrinsically high AKT activity using GC1118 combined with the dual PI3K/mammalian target of rapamycin (mTOR)/AKT inhibitor BEZ-235, without observed toxicity. Taken together, the superior antitumor efficacy of GC1118 alone or in combination with PI3K/mTOR/AKT inhibitors shows great therapeutic potential for the treatment of KRAS-mutant mCRC with elevated ratios of high- to low-affinity EGFR ligands and PI3K-AKT pathway activation.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Receptores ErbB/imunologia , Feminino , Humanos , Camundongos , Camundongos Nus , Mutação , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Basic Clin Pharmacol Toxicol ; 120(3): 243-249, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27637171

RESUMO

GC1118 is an anti-epidermal growth factor receptor (EGFR) monoclonal antibody that is currently under clinical development. In this study, the pharmacokinetics (PK) of GC1118 were modelled in monkeys to predict human PK and receptor occupancy (RO) profiles. The serum concentrations of GC1118 and its comparator (cetuximab) were assessed in monkeys with a non-compartmental analysis and a target-mediated drug disposition (TMDD) model after intravenous infusion (3-25 mg/kg) of these drugs. The scaling exponent of the EGFR synthesis rate was determined using a sensitivity analysis. The human cetuximab exposures were simulated by applying different exponents (0.7-1.0) for the EGFR synthesis rate in the allometric monkey PK model. Simulated Cmax and area under the curve values therein were compared with those previously reported in the literature to find the best exponent for the EGFR synthesis rate in human beings. The TMDD model appropriately described the monkey PK profile, which showed a decrease in clearance (CL; 1.2-0.4 ml/hr/kg) as the dose increased. The exponents for CL (0.75) and volume of distribution (Vd; 1.0) were used for the allometric scaling to predict human PK. The allometric coefficient for the EGFR synthesis rate chosen by the sensitivity analysis was 0.85, and the RO profiles that could not be measured experimentally were estimated based on the predicted concentrations of the total target and the drug-target complex. Our monkey TMDD model successfully predicts human PK and RO profiles of GC1118 and can be used to determine the appropriate dose for a first-in-human study investigating this drug.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Modelos Biológicos , Animais , Anticorpos Monoclonais Humanizados/sangue , Antineoplásicos/sangue , Cetuximab/sangue , Cetuximab/farmacologia , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos , Macaca fascicularis
10.
Mol Cancer Ther ; 15(2): 251-63, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26586721

RESUMO

The EGFR-targeted monoclonal antibodies are a valid therapeutic strategy for patients with metastatic colorectal cancer (mCRC). However, only a small subset of mCRC patients has therapeutic benefits and there are high demands for EGFR therapeutics with a broader patient pool and more potent efficacy. In this study, we report GC1118 exhibiting a different character in terms of binding epitope, affinity, mode of action, and efficacy from other anti-EGFR antibodies. Structural analysis of the EGFR-GC1118 crystal complex revealed that GC1118 recognizes linear, discrete N-terminal epitopes of domain III of EGFR, critical for EGF binding but not overlapping with those of other EGFR-targeted antibodies. GC1118 exhibited superior inhibitory activity against high-affinity EGFR ligands in terms of EGFR binding, triggering EGFR signaling, and proliferation compared with cetuximab and panitumumab. EGFR signaling driven by low-affinity ligands, on the contrary, was well inhibited by all the antibodies tested. GC1118 demonstrated robust antitumor activity in tumor xenografts with elevated expression of high-affinity ligands in vivo, whereas cetuximab did not. Considering the significant role of high-affinity EGFR ligands in modulating tumor microenvironment and inducing resistance to various cancer therapeutics, our study suggests a potential therapeutic advantage of GC1118 in terms of efficacy and a range of benefited patient pool. Mol Cancer Ther; 15(2); 251-63. ©2015 AACR.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Epitopos/metabolismo , Receptores ErbB/química , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Feminino , Humanos , Ligantes , Camundongos , Modelos Moleculares , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Biol Chem ; 290(27): 16943-53, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25979339

RESUMO

Syndecans, a family of transmembrane heparansulfate proteoglycans, are known to interact through their transmembrane domains to form non-covalently linked homodimers, a process essential for their individual functions. Because all syndecan transmembrane domains are highly conserved and thus might mediate interactions between different members of the syndecan family, we investigated syndecan interactions in detail. All recombinant syndecan-2 and -4 protein variants containing the transmembrane domain formed not only sodium dodecyl sulfate (SDS)-resistant homodimers but also SDS-resistant heterodimers. Biochemical and structural data revealed that recombinant syndecan-2 and -4 formed intermolecular interactions in vitro, and the GXXXG motif in transmembrane domain mediated this interaction. When exogenously expressed in rat embryonic fibroblasts, syndecan-2 interacted with syndecan-4 and vice versa. Furthermore, bimolecular fluorescence complementation-based assay demonstrated specific hetero-molecular interactions between syndecan-2 and -4, supporting hetero-oligomer formation of syndecans in vivo. Interestingly, hetero-oligomerization significantly reduced syndecan-4-mediated cellular processes such as protein kinase Cα activation and protein kinase Cα-mediated cell adhesion as well as syndecan-2-mediated tumorigenic activities in colon cancer cells such as migration and anchorage-independent growth. Taken together, these data provide evidence that hetero-oligomerization produces distinct syndecan functions and offer insights into the underlying signaling mechanisms of syndecans.


Assuntos
Sindecana-2/química , Sindecana-2/metabolismo , Sindecana-4/química , Sindecana-4/metabolismo , Motivos de Aminoácidos , Animais , Dimerização , Fibroblastos/química , Fibroblastos/metabolismo , Ligação Proteica , Ratos , Sindecana-2/genética , Sindecana-4/genética
12.
Diabetes ; 61(6): 1599-608, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22427380

RESUMO

Retinal neovascularization is observed in progression of diabetic retinopathy. New vessels grow into the vitreous cavity in proliferative diabetic retinopathy, resulting in traction retinal detachment and vitreous hemorrhage. To overcome the catastrophic visual loss due to these complications, efforts have been focused on the treatment of retinal neovascularization. In this study, we demonstrated the inhibitory effect of recombinant human apolipoprotein(a) kringle V (rhLK8) in an animal model of ischemia-induced retinal neovascularization. rhLK8 induced no definite toxicity on endothelial cells and retinal tissues at the therapeutic dosage. Interestingly, rhLK8 showed antiangiogenic effect, particularly on fibronectin-mediated migration of endothelial cells. Further experiments demonstrated high binding affinity of rhLK8 to α3ß1 integrin, and suppression of it might be the mechanism of antiangiogenic effect of rhLK8. Furthermore, rhLK8 inhibited phosphorylation of focal adhesion kinase, resulting in suppression of activation of consequent p130CAS-Jun NH(2)-terminal kinase. Taken together, our data suggested the possible application of rhLK8 in the treatment of retinal neovascularization by suppression of fibronectin-mediated angiogenesis.


Assuntos
Apolipoproteínas A/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Fibronectinas/metabolismo , Retina/efeitos dos fármacos , Neovascularização Retiniana/tratamento farmacológico , Animais , Apolipoproteínas A/farmacologia , Movimento Celular/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Fosforilação/efeitos dos fármacos , Retina/metabolismo , Neovascularização Retiniana/metabolismo
13.
J Biol Chem ; 285(3): 1743-53, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19880522

RESUMO

Pyk2 is a cytoplasmic tyrosine kinase related to focal adhesion kinase (FAK). Compensatory Pyk2 expression occurs upon FAK loss in mice. However, the impact of Pyk2 up-regulation remains unclear. Previous studies showed that nuclear-localized FAK promotes cell proliferation and survival through FAK FERM domain-enhanced p53 tumor suppressor degradation (Lim, S. T., Chen, X. L., Lim, Y., Hanson, D. A., Vo, T. T., Howerton, K., Larocque, N., Fisher, S. J., Schlaepfer, D. D., and Ilic, D. (2008) Mol. Cell 29, 9-22). Here, we show that FAK knockdown triggered p53 activation and G(1) cell cycle arrest in human umbilical vein endothelial cells after 4 days. However, by 7 days elevated Pyk2 expression occurred with a reduction in p53 levels and the release of the G(1) block under conditions of continued FAK knockdown. To determine whether Pyk2 regulates p53, experiments were performed in FAK(-/-)p21(-/-) mouse embryo fibroblasts expressing endogenous Pyk2 and in ID8 ovarian carcinoma cells expressing both Pyk2 and FAK. In both cell lines, Pyk2 knockdown increased p53 levels and inhibited cell proliferation associated with G(1) cell cycle arrest. Pyk2 FERM domain re-expression was sufficient to reduce p53 levels and promote increased BrdUrd incorporation. Pyk2 FERM promoted Mdm2-dependent p53 ubiquitination. Pyk2 FERM effects on p53 were blocked by proteasomal inhibition or mutational-inactivation of Pyk2 FERM nuclear localization. Staurosporine stress of ID8 cells promoted endogenous Pyk2 nuclear accumulation and enhanced Pyk2 binding to p53. Pyk2 knockdown potentiated ID8 cell death upon staurosporine addition. Moreover, Pyk2 FERM expression in human fibroblasts upon FAK knockdown prevented cisplatin-mediated apoptosis. Our studies demonstrate that nuclear Pyk2 functions to limit p53 levels, thus facilitating cell growth and survival in a kinase-independent manner.


Assuntos
Adaptação Fisiológica , Quinase 2 de Adesão Focal/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinase 2 de Adesão Focal/química , Quinase 2 de Adesão Focal/deficiência , Quinase 2 de Adesão Focal/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Mutação , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Estaurosporina/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
14.
FEBS Lett ; 583(14): 2395-400, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19560461

RESUMO

Although phosphatidylinositol 4,5-bisphosphate (PIP(2)) regulates syndecan-4 function, the potential influence of syndecan-4 on PIP(2) remains unknown. GFP containing PIP(2)-binding-PH domain of phospholipase Cdelta (GFP-PHdelta) was used to monitor PIP(2). Syndecan-4 overexpression in COS-7 cells enhanced membrane translocation of GFP-PHdelta, while the opposite was observed when syndecan-4 was knocked-down. PIP(2) levels were higher in total phospholipids extracted from rat embryo fibroblasts expressing syndecan-4. Syndecan-4-induced membrane targeting of GFP-PHdelta was further enhanced by phosphoinositide-3-kinase inhibitor, but not by phospholipase C (PLC) inhibitor. Besides, both ionomycin and epidermal growth factor caused dissociation of GFP-PHdelta from plasma membrane, an effect that was significantly delayed by syndecan-4 over-expression. Collectively, these data suggest that syndecan-4 promotes plasma membrane retention of PIP(2) by negatively regulating PLC-dependent PIP(2) degradation.


Assuntos
Membrana Celular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Sindecana-4/metabolismo , Animais , Células COS , Chlorocebus aethiops , Fator de Crescimento Epidérmico/metabolismo , Fosfolipídeos/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sindecana-4/genética , Fosfolipases Tipo C/metabolismo
15.
J Cell Sci ; 122(Pt 11): 1852-62, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19435801

RESUMO

Directional motility is a complex process requiring the spatiotemporal integration of signals that regulate cytoskeletal changes, and the establishment of an anteroposterior or polarized cell axis. Focal adhesion kinase (FAK) promotes cell migration, but a molecular role for FAK in promoting cell polarity remains undefined. Here, using wound healing and Golgi-reorientation analyses, we show that fibroblast, endothelial and carcinoma polarity during cell migration requires FAK and is associated with a complex between FAK, p120RasGAP and p190RhoGAP (p190A), leading to p190A tyrosine phosphorylation. Fibronectin-integrin-mediated FAK activation and phosphorylation promote SH2-mediated binding of p120RasGAP to FAK and FAK-mediated p190A tyrosine phosphorylation. The association of p120RasGAP with FAK facilitates the formation of a FAK-p120RasGAP-p190A complex targeted to leading-edge focal adhesions by FAK. Knockdown of p120RasGAP, mutation of FAK Y397 or inhibition of FAK activity prevent the association of FAK with p190A and subsequent tyrosine phosphorylation of p190A, and result in the loss of cell polarity. Because reconstitution of FAK-null fibroblasts with FAK or a Pyk2-FAK chimera restore the normal decrease in RhoA GTP binding upon cell spreading on fibronectin, our studies support a model whereby FAK activity facilitates the recruitment and stabilization of a p120RasGAP-p190A complex at leading-edge focal adhesions connected to the transient inhibition of RhoA activity and the regulation of cell polarity.


Assuntos
Movimento Celular/fisiologia , Polaridade Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Repressoras/metabolismo , Proteína p120 Ativadora de GTPase/metabolismo , Animais , Carcinoma/metabolismo , Carcinoma/patologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Quinase 2 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Adesões Focais/metabolismo , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Tirosina/metabolismo , Proteína p120 Ativadora de GTPase/genética
16.
Mol Cell ; 29(1): 9-22, 2008 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-18206965

RESUMO

FAK is known as an integrin- and growth factor-associated tyrosine kinase promoting cell motility. Here we show that, during mouse development, FAK inactivation results in p53- and p21-dependent mesodermal cell growth arrest. Reconstitution of primary FAK-/-p21-/- fibroblasts revealed that FAK, in a kinase-independent manner, facilitates p53 turnover via enhanced Mdm2-dependent p53 ubiquitination. p53 inactivation by FAK required FAK FERM F1 lobe binding to p53, FERM F2 lobe-mediated nuclear localization, and FERM F3 lobe for connections to Mdm2 and proteasomal degradation. Staurosporine or loss of cell adhesion enhanced FERM-dependent FAK nuclear accumulation. In primary human cells, FAK knockdown raised p53-p21 levels and slowed cell proliferation but did not cause apoptosis. Notably, FAK knockdown plus cisplatin triggered p53-dependent cell apoptosis, which was rescued by either full-length FAK or FAK FERM re-expression. These studies define a scaffolding role for nuclear FAK in facilitating cell survival through enhanced p53 degradation under conditions of cellular stress.


Assuntos
Quinase 1 de Adesão Focal/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/fisiologia , Núcleo Celular/metabolismo , Sobrevivência Celular/fisiologia , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/deficiência , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Quinase 1 de Adesão Focal/química , Quinase 1 de Adesão Focal/deficiência , Quinase 1 de Adesão Focal/genética , Mesoderma/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Estaurosporina/farmacologia , Ubiquitina/metabolismo , Ubiquitinação
17.
J Cell Biol ; 180(1): 187-203, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18195107

RESUMO

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK(-/-) or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK-p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


Assuntos
Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/fisiologia , ras-GRF1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/genética , Regulação da Expressão Gênica , Camundongos , Paxilina/metabolismo , Fosforilação , Tirosina/metabolismo , ras-GRF1/genética
18.
J Biol Chem ; 282(24): 17450-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17438336

RESUMO

Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase that promotes cell migration, survival, and gene expression. Here we show that FAK signaling is important for tumor necrosis factor-alpha (TNFalpha)-induced interleukin 6 (IL-6) mRNA and protein expression in breast (4T1), lung (A549), prostate (PC-3), and neural (NB-8) tumor cells by FAK short hairpin RNA knockdown and by comparisons of FAK-null (FAK(-/-)) and FAK(+/+) mouse embryo fibroblasts. FAK promoted TNFalpha-stimulated MAPK activation needed for maximal IL-6 production. FAK was not required for TNFalpha-mediated nuclear factor-kappaB or c-Jun N-terminal kinase activation. TNFalpha-stimulated FAK catalytic activation and IL-6 production were inhibited by FAK N-terminal but not FAK C-terminal domain overexpression. Analysis of FAK(-/-) fibroblasts stably reconstituted with wild type or various FAK point mutants showed that FAK catalytic activity, Tyr-397 phosphorylation, and the Pro-712/713 proline-rich region of FAK were required for TNFalpha-stimulated MAPK activation and IL-6 production. Constitutively activated MAPK kinase-1 (MEK1) expression in FAK(-/-) and A549 FAK short hairpin RNA-expressing cells rescued TNFalpha-stimulated IL-6 production. Inhibition of Src protein-tyrosine kinase activity or mutation of Src phosphorylation sites on FAK (Tyr-861 or Tyr-925) did not affect TNFalpha-stimulated IL-6 expression. Moreover, analyses of Src(-/-), Yes(-/-), and Fyn(-/-) fibroblasts showed that Src expression was inhibitory to TNFalpha-stimulated IL-6 production. These studies provide evidence for a novel Src-independent FAK to MAPK signaling pathway regulating IL-6 expression with potential importance to inflammation and tumor progression.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Interleucina-6/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Interleucina-6/genética , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/genética , NF-kappa B/metabolismo , Prolina/metabolismo , Interferência de RNA , Tirosina/metabolismo , Quinases da Família src/metabolismo
19.
J Biol Chem ; 282(14): 10398-404, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17303567

RESUMO

Focal adhesion kinase (FAK) mediates signal transduction in response to multiple extracellular inputs via tyrosine phosphorylation at specific residues. Although several tyrosine phosphorylation events have been linked to FAK activation and downstream signal transduction, the function of FAK phosphorylation at Tyr(407) was previously unknown. Here, we show for the first time that phosphorylation of FAK Tyr(407) increases during serum starvation, contact inhibition, and cell cycle arrest, all conditions under which activating FAK Tyr(397) phosphorylation decreases. Transfection of NIH3T3 cells with a phosphorylation-mimicking FAK 407E mutant decreased autophosphorylation at Tyr(397) and inhibited both FAK kinase activity in vitro and FAK-mediated functions such as cell adhesion, spreading, proliferation, and migration. The opposite effects were observed in cells transfected with nonphosphorylatable mutant FAK 407F. Taken together, these data suggest the novel concept that FAK Tyr(407) phosphorylation negatively regulates the enzymatic and biological activities of FAK.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células , Quinase 1 de Adesão Focal/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Transdução de Sinais/fisiologia , Animais , Adesão Celular/fisiologia , Ativação Enzimática/genética , Quinase 1 de Adesão Focal/genética , Camundongos , Mutação , Células NIH 3T3 , Fosforilação , Tirosina/genética , Tirosina/metabolismo
20.
Arch Biochem Biophys ; 454(1): 1-6, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16962988

RESUMO

Protein kinase Calpha (PKCalpha) activation is known to be dependent on the metabolic product of phosphatidylinositol 4,5-bisphosphate (PIP2) by phospholipase C (PLC). Here we report that fibroblasts may have an additional PIP2-dependent mechanism for membrane localization of PKCalpha. We observed PKCalpha membrane localization in both wild type and PLCgamma1 -/- mouse embryonic fibroblasts. Treatment of cells with a specific PLC inhibitor U73122 resulted in increased PIP2 levels and enhanced membrane localization of PKCalpha. PKCalpha levels in the membrane fraction decreased following incubation with PLCgamma, but increased following treatment with U73122 or addition of exogenous PIP2 in vitro. In addition, PKCalpha interacted with PIP2-conjugate bead and mixed micelles containing PIP2. Finally, we found that PIP2 is involved in syndecan-4-mediated membrane localization of PKCalpha. Taken together, these data suggest that PIP2 might contribute to directly regulating the membrane localization of PKCalpha.


Assuntos
Membrana Celular/metabolismo , Fibroblastos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C-alfa/metabolismo , Transdução de Sinais/fisiologia , Frações Subcelulares/metabolismo , Animais , Células COS , Chlorocebus aethiops , Camundongos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA