Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Mol Cancer Ther ; 22(3): 381-392, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36805730

RESUMO

FDA-approved enzalutamide is commonly prescribed to reduce the growth of advanced prostate cancer by blocking androgen receptor function. However, enzalutamide-resistant prostate cancer (ERPC) invariably develops and progresses to metastatic, lethal disease. Management of ERPC poses a special problem not only because available therapeutic regimens cannot effectively kill ERPC cells but also due to their propensity to invade large bones. Moreover, molecular mechanism(s) behind enzalutamide resistance is not properly understood, which is delaying development of newer agents. We found that the pseudokinase, Tribbles 2 (TRIB2), is overexpressed in ERPC cells and plays a critical role in their survival. Forced overexpression of TRIB2 enhances prostate cancer cell growth and confers resistance to physiologic doses of enzalutamide, suggesting that TRIB2 plays an important role in the development and progression of ERPC. Though TRIB2 has emerged as an excellent molecular target for ERPC, suitable inhibitors are not commercially available for effective targeting. By designing a luciferase-tagged TRIB2 fusion protein-based assay system, we screened a library of about 1,600 compounds and found that daclatasvir (DCV), an antiviral drug, effectively inhibits TRIB2-luciferase. We also found that DCV degrades TRIB2 proteins by direct binding and resensitizes ERPC cells to enzalutamide treatment. Moreover, DCV at lower, sublethal doses synergizes with enzalutamide to decrease the viability and induce apoptosis in prostate cancer cells. Because DCV is already approved by the FDA and well tolerated in humans, based on our findings, it appears that DCV is a promising new agent for development of an effective therapy for advanced, enzalutamide-resistant, lethal prostate cancer.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Masculino , Humanos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Antivirais/farmacologia , Antivirais/uso terapêutico , Transdução de Sinais , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/metabolismo , Nitrilas/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/uso terapêutico
2.
Endocr J ; 69(4): 385-397, 2022 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-34719526

RESUMO

Promoting white adipose tissue (WAT) "browning" and brown adipose tissue (BAT) activation could contribute to increasing energy expenditure. We explored the mechanisms by which the natural compound rutin induced adipose tissue differentiation and ameliorated obesity in vivo and in vitro. 3T3-L1 preadipocytes were cultured in adipogenic differentiation media with/out rutin. Male C57BL/6 mice (n = 6) were fed a high-fat diet (HFD) for 12 weeks with/out rutin. In HFD-fed mice, rutin treatment significantly inhibited weight gain, improved the metabolic profile of plasma samples, decreased the weights of epididymal WAT (eWAT), inguina WAT (iWAT), and liver, and adipocyte size. Furthermore, rutin also increased the expression of uncoupling protein 1 (Ucp-1) and other thermogenic markers in the WAT and BAT. In 3T3-L1 cells, rutin effectively reduced the formation of lipid droplets, stimulated the expression of thermogenic markers, and reduced the expression of adipogenic genes. Additionally, rutin markedly upregulated the AMP-activated protein kinase (AMPK) pathway, and these effects were diminished by treatment with the AMPK inhibitor compound C (CC). Pretreatment with the calmodulin-dependent protein kinase kinase ß (CaMKKß) inhibitor STO-609 blocked the induction of thermogenic markers in 3T3-L1 cells by rutin. Our results indicated that rutin increased energy consumption, induced WAT "browning" and BAT activation, and thus was a promising target for the development of new therapeutic approaches to improve adipose tissue energy metabolism.


Assuntos
Proteínas Quinases Ativadas por AMP , Tecido Adiposo Marrom , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Dieta Hiperlipídica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rutina/metabolismo , Rutina/farmacologia , Termogênese/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
3.
J Periodontol ; 85(3): 498-508, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23805815

RESUMO

BACKGROUND: Previous studies have shown that periodontal ligament exists in a hypoxic microenvironment, especially under the condition of periodontitis or physical stress. The present study is designed to investigate the effects and mechanisms of hypoxia on regulating the proliferation and osteogenic differentiation of human periodontal ligament cells (hPDLCs) under cyclic tensile stress (CTS). METHODS: hPDLCs were cultured in 2% O2 (hypoxia) or 20% O2 (normoxia) and then subjected to a cyclic in-plane tensile deformation of 10% at 0.5 Hz. The following parameters were measured: 1) cell proliferation by flow cytometry; 2) cell ultrastructure by transmission electron microscopy; 3) expression of hypoxia-inducible factor-1α (HIF-1α) and osteogenic relative factors (i.e., secreted phosphoprotein 1 [SPP1; also known as bone sialoprotein I/osteopontin], runt-related transcription factor 2 [RUNX2], and transcription factor Sp7 [SP7]) by real-time polymerase chain reaction and Western blot; and 4) involvement of mitogen-activated protein kinase (MAPK) signaling pathways by Western blot with specific inhibitor. RESULTS: Proliferation index in the hypoxia with CTS group was significantly higher than in other groups. Significant increases in HIF-1α, SPP1, RUNX2, and SP7 occurred in the presence of hypoxia for 24 hours. In addition, MAPK inhibitor (PD 98,059) significantly attenuated hypoxia and CTS-induced phosphor-ERK1/2 (extracellular regulated kinase 1/2), phosphor-JNK (c-jun N-terminal kinase), and phosphor-P38 expression. CONCLUSIONS: Hypoxia regulates CTS-responsive changes in proliferation and osteogenic differentiation of hPDLCs via MAPK pathways. Hypoxia-treated hPDLCs may serve as an in vitro model to explore the molecular mechanisms of periodontitis.


Assuntos
Hipóxia Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Osteogênese/fisiologia , Ligamento Periodontal/citologia , Adolescente , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Microambiente Celular/fisiologia , Criança , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Feminino , Flavonoides , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Osteopontina/análise , Ligamento Periodontal/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição Sp7 , Resistência à Tração , Fatores de Tempo , Fatores de Transcrição/análise , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
4.
Reproduction ; 144(4): 411-22, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22843772

RESUMO

The mechanisms leading to capacitation in stallion sperm are poorly understood. The objective of our study was to define factors associated with regulation of protein tyrosine phosphorylation in stallion sperm. Stallion sperm were incubated for 4 h in modified Whitten's media with or without bicarbonate, calcium, or BSA. When sperm were incubated in air at 30×106/ml at initial pH 7.25, protein tyrosine phosphorylation was detected only in medium containing 25 mM bicarbonate alone; calcium and BSA inhibited phosphorylation. Surprisingly, this inhibition did not occur when sperm were incubated at 10×106/ml. The final pH values after incubation at 30×106 and 10×106 sperm/ml were 7.43 ± 0.04 and 7.83 ± 0.07 (mean ± s.e.m.) respectively. Sperm were then incubated at initial pH values of 7.25, 7.90, or 8.50 in either air or 5% CO2. Protein tyrosine phosphorylation increased with increasing final medium pH, regardless of the addition of bicarbonate or BSA. An increase in environmental pH was observed when raw semen was instilled into the uteri of estrous mares and retrieved after 30 min (from 7.47 ± 0.10 to 7.85 ± 0.08), demonstrating a potential physiological role for pH regulation of capacitation. Sperm incubated in the presence of the calmodulin (CaM) inhibitor W-7 exhibited a dose-dependent increase in protein tyrosine phosphorylation, suggesting that the inhibitory effect of calcium was CaM mediated. These results show for the first time a major regulatory role of external pH, calcium, and CaM in stallion sperm protein tyrosine phosphorylation.


Assuntos
Sinalização do Cálcio , Calmodulina/metabolismo , Cavalos/fisiologia , Fosfoproteínas/metabolismo , Capacitação Espermática , Espermatozoides/metabolismo , Tirosina/metabolismo , Animais , Cálcio/análise , Sinalização do Cálcio/efeitos dos fármacos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Calmodulina/antagonistas & inibidores , Quelantes/farmacologia , Ácido Egtázico/farmacologia , Inibidores Enzimáticos , Concentração de Íons de Hidrogênio , Masculino , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Sêmen/química , Capacitação Espermática/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Sulfonamidas/farmacologia
5.
Cardiology ; 117(3): 207-15, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21150201

RESUMO

AIM: Inflammation plays an important role in coronary microembolization (CME)-induced myocardial injury. The present study was designed to investigate the role of extracellular signal-regulated kinases 1/2 (ERK1/2) signaling pathway in regulating myocardial inflammation and cardiac function in a rat model of CME. METHODS: Sprague-Dawley rats were randomly divided into three groups: sham-operated group (sham group), CME group and PD98059 group (15 animals per group). CME was produced by injection of 42-µm microspheres into the left ventricle with occlusion of the ascending aorta. Rats in the PD98059 group were injected with PD98059, a specific ERK1/2 inhibitor, 30 min before the CME operation. Western blotting and immunohistochemistry analysis were used to determine the activation of ERK1/2. Echocardiography was employed to evaluate cardiac function. Hematoxylin-eosin staining was performed to assay myocardial inflammation. Expression of TNF-α mRNA was determined by RT-PCR analysis, and activity of NF-κB was assessed by electrophoretic mobility shift assay. RESULTS: CME dramatically induced cardiac dysfunction (left ventricular ejection fraction, LVEF, was 72.97 ± 3.20% in the CME vs. 82.69 ± 3.50% in the sham group, p < 0.05) and local myocardial inflammatory response, both of which were ameliorated significantly by PD98059 (LVEF was 76.46 ± 4.46 and p < 0.05 vs. CME group). When compared to the CME group, PD98059 markedly attenuated the increased phosphorylation of ERK1/2 (0.48 ± 0.11 vs. 0.92 ± 0.10, p < 0.05), expression of TNF-α mRNA (0.42 ± 0.06 vs. 0.94 ± 0.04, p < 0.05) and activity of NF-κB (104.83 ± 13.65 vs. 540.79 ± 24.95, p < 0.05) in CME rat myocardium. CONCLUSIONS: The present study demonstrates a novel role of the ERK1/2 signaling pathway in promoting myocardium inflammation and dysfunction in CME, and suggests that ERK1/2 is a novel potential therapeutic target for CME.


Assuntos
Oclusão Coronária/complicações , Oclusão Coronária/fisiopatologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Miocárdio/patologia , Transdução de Sinais/fisiologia , Animais , Far-Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Placa Aterosclerótica/complicações , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
6.
Mol Carcinog ; 49(8): 771-82, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20572161

RESUMO

Fucoidan, a sulfated polysaccharide in brown seaweed, has various biological activities including anti-tumor activity. We investigated the effects of fucoidan on the apoptosis of human promyeloid leukemic cells and fucoidan-mediated signaling pathways. Fucoidan induced apoptosis of HL-60, NB4, and THP-1 cells, but not K562 cells. Fucoidan treatment of HL-60 cells induced activation of caspases-8, -9, and -3, the cleavage of Bid, and changed mitochondrial membrane permeability. Fucoidan-induced apoptosis, cleavage of procaspases, and changes in the mitochondrial membrane permeability were efficiently blocked by depletion of mitogen-activated protein kinase (MAPK) kinase kinase 1 (MEKK1), and inhibitors of MAPK kinase 1 (MEK1) and c Jun NH2-terminal kinase (JNK). The phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and JNK was increased in fucoidan-treated HL-60, NB4, and THP-1 cells, but not K562 cells. ERK1/2 activation occurred at earlier times than JNK activation and JNK activation was blocked by MEK1 inhibitor. In addition, fucoidan-induced apoptosis was inhibited by addition of glutathione and/or L-NAME, and fucoidan decreased intracellular glutathione concentrations and stimulated nitric oxide (NO) production. Buthionine-[R,S]-sulfoximine rendered HL-60 cells more sensitive to fucoidan. Depletion of MEKK1 and inhibition of MEK1 restored the intracellular glutathione content and abrogated NO production, whereas inhibition of JNK activation by SP600125 restored intracellular glutathione content but failed to inhibit NO production in fucoidan-treated HL-60 cells. These results suggest that activation of MEKK1, MEK1, ERK1/2, and JNK, depletion of glutathione, and production of NO are important mediators in fucoidan-induced apoptosis of human leukemic cells.


Assuntos
Apoptose/efeitos dos fármacos , Glutationa/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Antracenos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Caspase 8/metabolismo , Glutationa/farmacologia , Células HL-60 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células K562 , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 1/farmacologia , MAP Quinase Quinase Quinase 1/metabolismo , MAP Quinase Quinase Quinase 1/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/farmacologia , NG-Nitroarginina Metil Éster/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/farmacologia , Fosforilação/efeitos dos fármacos , Polissacarídeos/metabolismo , Polissacarídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
7.
Cancer Genet Cytogenet ; 199(2): 81-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20471510

RESUMO

MG132 (carbobenzoxy-Leu-Leu-leucinal) as a proteasome inhibitor has been shown to induce apoptotic cell death through formation of reactive oxygen species (ROS). In this study, we investigated the effects of MEK (mitogen-activated protein [MAP] kinase or extracellular signal-regulated kinase [ERK] kinase) or p38 inhibitor on MG132-treated Calu-6 lung cancer cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. Treatment with 10 mumol/L MG132 inhibited the growth of Calu-6 cells at 24 hours. MG132 induced apoptosis in Calu-6 cells, which was accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)). ROS were increased in MG132-treated Calu-6 cells. MG132 also induced GSH depletion in Calu-6 cells. Treatment with MEK inhibitor did not significantly affect cell growth, cell death, ROS, and GSH levels in MG132-treated Calu-6 cells. Furthermore, MG132 increased the phosphorylation of p38 in Calu-6 cells at 1 and 24 hours. Treatment with p38 inhibitor significantly prevented cell growth inhibition, MMP (DeltaPsi(m)) loss and apoptosis in MG132-treated Calu-6 cells. This inhibitor increased ROS level and decreased GSH depletion in these cells. In conclusion, p38 inhibitor partially prevented Calu-6 cell death by MG132, which might be affected by GSH level changes.


Assuntos
Adenocarcinoma/tratamento farmacológico , Inibidores de Cisteína Proteinase/toxicidade , Imidazóis/uso terapêutico , Leupeptinas/toxicidade , Neoplasias Pulmonares/tratamento farmacológico , Piridinas/uso terapêutico , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Apoptose/efeitos dos fármacos , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Flavonoides/farmacologia , Glutationa/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
8.
Invest Ophthalmol Vis Sci ; 49(1): 113-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18172082

RESUMO

PURPOSE: To investigate the effects of corneal fibroblasts on the differentiation of corneal epithelial cells in a coculture system based on a collagen vitrigel membrane. METHODS: Simian virus 40-transformed human corneal epithelial (HCE) cells and human corneal fibroblasts were cultured on opposite sides of a collagen vitrigel membrane. The distribution of HCE cells and corneal fibroblasts on the collagen membrane was determined by immunofluorescence staining and immunoblot analysis of marker proteins. Expression of the tight-junctional proteins ZO-1, occludin, and claudin and of the adherens-junctional proteins E- and N-cadherin in HCE cells was determined at the mRNA and protein levels by reverse transcription-polymerase chain reaction analysis and immunoblot analysis, respectively. RESULTS: The abundance of ZO-1, occludin, and claudin mRNA and proteins in HCE cells was markedly increased by coculture with corneal fibroblasts. The expression of E- or N-cadherin did not differ between HCE cells cultured with corneal fibroblasts and those cultured without them. PD98059, a specific inhibitor of signaling by extracellular signal regulated kinase (ERK), prevented the upregulation of tight-junctional proteins in HCE cells by corneal fibroblasts. CONCLUSIONS: Human corneal fibroblasts regulated the expression of tight-junctional proteins in HCE cells, suggesting that corneal fibroblasts may play an important role in the differentiation of corneal epithelial cells.


Assuntos
Epitélio Corneano/metabolismo , Fibroblastos/fisiologia , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Regulação para Cima/fisiologia , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Diferenciação Celular , Linhagem Celular Transformada , Claudina-1 , Técnicas de Cocultura , Colágeno , Córnea/citologia , Epitélio Corneano/efeitos dos fármacos , Flavonoides/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Immunoblotting , Proteínas de Membrana/genética , Ocludina , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus 40 dos Símios , Proteína da Zônula de Oclusão-1
9.
J Mol Cell Cardiol ; 43(3): 281-91, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17643448

RESUMO

We aimed to define the relative contribution of both PKA and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) cascades to the phosphorylation of RyR2 and the activity of the channel during beta-adrenergic receptor (betaAR) stimulation. Rat hearts were perfused with increasing concentrations of the beta-agonist isoproterenol in the absence and the presence of CaMKII inhibition. CaMKII was inhibited either by preventing the Ca(2+) influx to the cell by low [Ca](o) plus nifedipine or by the specific inhibitor KN-93. We immunodetected RyR2 phosphorylated at Ser2809 (PKA and putative CaMKII site) and at Ser2815 (CaMKII site) and measured [(3)H]-ryanodine binding and fast Ca(2+) release kinetics in sarcoplasmic reticulum (SR) vesicles. SR vesicles were isolated in conditions that preserved the phosphorylation levels achieved in the intact heart and were actively and equally loaded with Ca(2+). Our results demonstrated that Ser2809 and Ser2815 of RyR2 were dose-dependently phosphorylated under betaAR stimulation by PKA and CaMKII, respectively. The isoproterenol-induced increase in the phosphorylation of Ser2815 site was prevented by the PKA inhibitor H-89 and mimicked by forskolin. CaMKII-dependent phosphorylation of RyR2 (but not PKA-dependent phosphorylation) was responsible for the beta-induced increase in the channel activity as indicated by the enhancement of the [(3)H]-ryanodine binding and the velocity of fast SR Ca(2+) release. The present results show for the first time a dose-dependent increase in the phosphorylation of Ser2815 of RyR2 through the PKA-dependent activation of CaMKII and a predominant role of CaMKII-dependent phosphorylation of RyR2, over that of PKA-dependent phosphorylation, on SR-Ca(2+) release during betaAR stimulation.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Benzilaminas/farmacologia , Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico , Relação Dose-Resposta a Droga , Isoproterenol/farmacologia , Isoquinolinas/farmacologia , Cinética , Masculino , Nifedipino/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/farmacologia , Ratos , Ratos Wistar , Sulfonamidas/farmacologia
10.
Arch Otolaryngol Head Neck Surg ; 133(3): 281-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17372087

RESUMO

OBJECTIVE: To investigate the downstream molecular targets of hyaluronan (HA)-CD44 and phospholipase C (PLC)-mediated calcium ion (Ca(2+)) signaling in head and neck squamous cell carcinoma (HNSCC). Hyaluronan is a ligand for the CD44 receptor, which interacts with multiple signaling pathways to influence cellular behavior. We recently determined that HA-CD44 interaction promotes PLC-mediated Ca(2+) signaling and cisplatin resistance in HNSCC. DESIGN: Proliferation of HNSCC tumor cells and topoisomerase (Topo) II enzymatic activity, including DNA-cleavable complex formation and DNA decatenation, were analyzed in the presence or absence of HA, the Topo II poison etoposide (VP-16), and various inhibitors of PLC and Ca(2+)-calmodulin kinase II (CaMKII) signaling. RESULTS: Treatment with HA promoted Topo II phosphorylation, suggesting that HA can modulate Topo II activity. Topoisomerase II-mediated DNA cleavable complex formation was increased by VP-16, and this increase was significantly enhanced by noncytotoxic doses of the PLC inhibitor U73122 and the CaMKII inhibitor KN-62, implicating PLC and CaMKII in Topo II regulation. However, the drug- and inhibitor-mediated increase in DNA cleavable complex formation was reduced with HA pretreatment. Inhibitors of PLC and CaMKII also enhanced VP-16 inhibition of Topo II-mediated DNA decatenation. Treatment with HA reduced VP-16 cytotoxic activity. On the other hand, U73122 and KN-62 enhanced VP-16 cytotoxic activity and reduced the ability of HA to promote VP-16 resistance. CONCLUSION: Our results suggest that HA, PLC, and CaMKII are upstream regulators of Topo II-mediated DNA metabolism in HNSCC and that this signaling pathway could be a promising target for the development of novel therapies against HNSCC.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Carcinoma de Células Escamosas/metabolismo , DNA Topoisomerases Tipo II/efeitos dos fármacos , Etoposídeo/efeitos adversos , Neoplasias de Cabeça e Pescoço/metabolismo , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Antineoplásicos Fitogênicos/uso terapêutico , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , DNA Topoisomerases Tipo II/metabolismo , Etoposídeo/uso terapêutico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Ácido Hialurônico/farmacologia , Masculino , Fosforilação , Transdução de Sinais , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/farmacologia
11.
Urology ; 69(3): 596-602, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17382186

RESUMO

OBJECTIVES: To test the hypothesis that blockage of epidermal growth factor receptor (EGFR) or methylethylketone (MEK)1/2 kinase activities impairs the growth of kidney cancer cells and magnifies the growth inhibitory effect of the mammalian target of rapamycin (mTOR) inhibitor rapamycin. METHODS: The kidney cancer cells from eight cell lines (including a pair in which the VHL gene or an empty vector was transfected in a VHL(mut) cell line) were tested for the effect of treatment with an EGFR inhibitor or an MEK1/2 inhibitor on the phosphorylation status of the phosphatidyl inositol 3-OH kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways after stimulation with EGF. In vitro growth assays were performed with EGFR inhibitors (gefitinib or erlotinib) or with MEK1/2 inhibitors (UO126 or PD184352) alone or in combination with rapamycin. The effects of PD184352, rapamycin, and their combination in the cell cycle were evaluated by flow cytometry. RESULTS: The growth suppressive effect of combined gefitinib (or erlotinib) and rapamycin was greater than the effect of each drug alone and was not dependent on VHL status. By affecting downstream signaling, the MEK1/2 inhibitors U0126 and PD184352 blocked growth more effectively than did the EGFR inhibitors in selected renal cell carcinoma lines; this effect was enhanced by the addition of rapamycin. At the cell cycle level, the combination resulted in enhanced G1 arrest. Although eIF4E overexpression has been suggested to make cells resistant to rapamycin, we observed marked growth inhibition with rapamycin as a single agent in SKRC39, which has marked overexpression of eIF4E. CONCLUSIONS: The results of our study have shown that combined mTOR and other upstream inhibitors have strong potential in the treatment of renal cell carcinoma.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Renais/tratamento farmacológico , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Sirolimo/farmacologia , Antibióticos Antineoplásicos/farmacologia , Benzamidas/farmacologia , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Carcinoma de Células Renais/fisiopatologia , Ciclo Celular/efeitos dos fármacos , Cloridrato de Erlotinib , Citometria de Fluxo , Gefitinibe , Neoplasias Renais/fisiopatologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases , Quinazolinas/farmacologia , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas
12.
J Neurochem ; 100(4): 1032-46, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17156128

RESUMO

Synapse Associated Protein 97 (SAP97), a member of membrane-associated guanylate kinase (MAGUK) protein family, has been involved in the correct targeting and clustering of ionotropic glutamate receptors (iGluRs) at postsynaptic sites. Calcium/calmodulin kinase II (CaMKII) phosphorylates SAP97 on two major sites in vivo; one located in the N-terminal domain (Ser39) and the other in the first postsynaptic density disc large ZO1 (PDZ) domain (Ser232). CaMKII-mediated phosphorylation of SAP97-Ser39 is necessary and sufficient to drive SAP97 to the postsynaptic compartment in cultured hippocampal neurons. CaMKII-dependent phosphorylation of Ser232 disrupts SAP97 interaction with NR2A subunit, thereby regulating synaptic targeting of this NMDA receptor subunit. Here we show by means of phospho-specific antibodies that SAP97-Ser39 phosphorylation represents the driving force to release SAP97/NR2A complex from the endoplasmic reticulum. Ser39 phosphorylation does not interfere with SAP97 capability to bind NR2A. On the contrary, SAP97-Ser232 phosphorylation occurs within the postsynaptic compartment and is responsible for both the disruption of NR2A/SAP97 complex and, consequently, for NR2A insertion in the postsynaptic membrane. Thus, CaMKII-dependent phosphorylation of SAP97 in different time frames and locations within the neurons controls both NR2A trafficking and insertion.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Proteínas de Membrana/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Cafeína/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Células Cultivadas , Cricetinae , Cricetulus , Interações Medicamentosas , Embrião de Mamíferos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/citologia , Imunoprecipitação/métodos , Técnicas In Vitro , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurônios/ultraestrutura , Inibidores de Fosfodiesterase/farmacologia , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Serina/metabolismo , Transfecção/métodos
13.
Neuron ; 51(4): 417-29, 2006 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-16908408

RESUMO

Efflux of dopamine through the dopamine transporter (DAT) is critical for the psychostimulatory properties of amphetamines, but the underlying mechanism is unclear. Here we show that Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) plays a key role in this efflux. CaMKIIalpha bound to the distal C terminus of DAT and colocalized with DAT in dopaminergic neurons. CaMKIIalpha stimulated dopamine efflux via DAT in response to amphetamine in heterologous cells and in dopaminergic neurons. CaMKIIalpha phosphorylated serines in the distal N terminus of DAT in vitro, and mutation of these serines eliminated the stimulatory effects of CaMKIIalpha. A mutation of the DAT C terminus impairing CaMKIIalpha binding also impaired amphetamine-induced dopamine efflux. An in vivo role for CaMKII was supported by chronoamperometry measurements showing reduced amphetamine-induced dopamine efflux in response to the CaMKII inhibitor KN93. Our data suggest that CaMKIIalpha binding to the DAT C terminus facilitates phosphorylation of the DAT N terminus and mediates amphetamine-induced dopamine efflux.


Assuntos
Anfetaminas/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Dopamina/metabolismo , Neurônios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Benzilaminas/farmacologia , Transporte Biológico/efeitos dos fármacos , Western Blotting/métodos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Proteínas da Membrana Plasmática de Transporte de Dopamina/química , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Inibidores Enzimáticos/farmacologia , Humanos , Imuno-Histoquímica/métodos , Imunoprecipitação/métodos , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mesencéfalo/citologia , Neurônios/fisiologia , Técnicas de Patch-Clamp/métodos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Ratos , Sulfonamidas/farmacologia , Transfecção/métodos
14.
J Neurochem ; 98(3): 713-22, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16893417

RESUMO

We have shown that the splicing isoform of Dp71 (Dp71d) localizes to the nucleus of PC12 cells, an established cell line derived from a rat pheochromocytoma; however, the mechanisms governing its nuclear localization are unknown. As protein phosphorylation modulates the nuclear import of proteins, and as Dp71d presents several potential sites for phosphorylation, we analyzed whether Dp71d is phosphorylated in PC12 cells and the role of phosphorylation on its nuclear localization. We demonstrated that Dp71d is phosphorylated under basal conditions at serine and threonine residues by endogenous protein kinases. Dp71d phosphorylation was activated by 2-O-tetradecanoyl phorbol 13-acetate (TPA), but this effect was blocked by EGTA. Supporting the role of intracellular calcium on Dp71d phosphorylation, we observed that the stimulation of calcium influx by cell depolarization increased Dp71d phosphorylation, and that the calcium-calmodulin inhibitor N-(6-aminohexyl)-1-naphthalenesulfonamide (W-7) blocked such induction. The blocking action of bisindolylmaleimide I (Bis I), a specific inhibitor for Ca2+/diacylglicerol-dependent protein kinase (PKC), on Dp71d phosphorylation suggested the participation of PKC in this event. In addition, transfection experiments with Ca2+/calmodulin-dependent protein kinase II (CaMKII) expression vectors as well as the use of KN-62, a CaMKII-specific inhibitor, demonstrated that CaMKII is also involved in Dp71d phosphorylation. Stimulation of Dp71d phosphorylation by cell depolarization and/or the overexpression of CaMKII favored the Dp71d nuclear accumulation. Overall, our results indicate that CAMKII-mediated Dp71d phosphorylation modulates its nuclear localization.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Distrofina/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Distrofina/genética , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Células PC12 , Fosforilação , Ratos
15.
J Virol ; 79(20): 12752-62, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16188978

RESUMO

Members of the mitogen-activated protein kinase (MAPK) family, including Jun amino-terminal kinase (JNK) and extracellular signal-related kinase (ERK), play an important role in the proliferation of erythroid cells in response to erythropoietin (Epo). Erythroid cells infected with the Friend spleen focus-forming virus (SFFV) proliferate in the absence of Epo and show constitutive activation of Epo signal transduction pathways. We previously demonstrated that the ERK pathway was constitutively activated in Friend SFFV-infected erythroid cells, and in this study JNK is also shown to be constitutively activated. Pharmacological inhibitors of both the ERK and JNK pathways stopped the proliferation of primary erythroleukemic cells from Friend SFFV-infected mice, with little induction of apoptosis, and furthermore blocked their ability to form Epo-independent colonies. However, only the JNK inhibitor blocked the proliferation of erythroleukemia cell lines derived from these mice. The JNK inhibitor caused significant apoptosis in these cell lines as well as an increase in the fraction of cells in G(2)/M and undergoing endoreduplication. In contrast, the growth of erythroleukemia cell lines derived from Friend murine leukemia virus (MuLV)-infected mice was inhibited by both the MEK and JNK inhibitors. JNK is important for AP1 activity, and we found that JNK inhibitor treatment reduced AP1 DNA-binding activity in primary erythroleukemic splenocytes from Friend SFFV-infected mice and in erythroleukemia cell lines from Friend MuLV-infected mice but did not alter AP1 DNA binding in erythroleukemia cell lines from Friend SFFV-infected mice. These data suggest that JNK plays an important role in cell proliferation and/or the survival of erythroleukemia cells.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leucemia Experimental/metabolismo , Infecções por Retroviridae/metabolismo , Transdução de Sinais , Vírus Formadores de Foco no Baço/fisiologia , Infecções Tumorais por Vírus/metabolismo , Animais , Antracenos/farmacologia , Apoptose , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Linhagem Celular Transformada/virologia , Proliferação de Células , Transformação Celular Viral , Células Cultivadas , Eritropoetina , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas/fisiologia
16.
Virus Res ; 113(2): 153-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15993506

RESUMO

MAP kinase signaling has been implicated in coxsackievirus B3 (CVB3) pathogenesis and as necessary in the virus lifecycle. We studied the correlation with extracellular signal-regulated kinase 1/2 (ERK1/2) signaling and virus replication in the presence of coxsackievirus and adenovirus receptor (CAR). In CHO cells that do not expressed CAR, specific ERK1/2 phosphorylation (pERK1/2) was not detected, and progeny virus was not produced after infection. By contrast, in HeLa and CHO-CAR cells, which expressed CAR, the specific early and late pERK1/2 at 0.5 and 8 h were induced, and progeny viruses were produced progressively through 24 h after infection. However, when CHO-CAR cells were infected with replication-defective CVB3, specific pERK1/2 was not detected. In addition, when late pERK1/2 is inhibited by the MEK1 inhibitor PD98059, at 4 h after infection, virus replication significantly decreased. Therefore, our findings suggest that early pERK1/2 is a response to virus binding to CAR, whereas late pERK1/2 is related to the viral replication.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Enterovirus Humano B/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Células CHO , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Cricetinae , Enterovirus Humano B/genética , Flavonoides/farmacologia , Deleção de Genes , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Transdução de Sinais , Replicação Viral
17.
Nat Neurosci ; 8(7): 853-4, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15924137

RESUMO

GluR1 and GluR2 subunits compose AMPA receptors (AMPARs) in the mature hippocampus, and both the GluR1 subunit and Ca2+/calmodulin-dependent protein kinase II (CaMKII) are required for synaptic plasticity, memory and learning. Although GluR1 phosphorylationby CaMKII is preserved, the functional regulation of AMPARs by phosphorylation is lost in the presence of the GIuR2 subunit. Our findings define a previously unknown, dominant role of the GluR2 subunit in signaling mediated by CaMKII at AMPARs.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Receptores de AMPA/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Linhagem Celular , Condutividade Elétrica , Humanos , Receptores de AMPA/fisiologia , Transdução de Sinais
18.
Cancer Res ; 65(5): 1994-2000, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15753399

RESUMO

Activation of mitogen-activated protein kinase (MAPK) occurs in response to various growth stimulating signals and as a result of activating mutations of the upstream regulators, KRAS and BRAF, which can be found in many types of human cancer. To investigate the roles of MAPK activation in tumors harboring KRAS or BRAF mutations, we inactivated MAPK in ovarian tumor cells using CI-1040, a compound that selectively inhibits MAPK kinase, an upstream regulator of MAPK and thus prevents MAPK activation. Profound growth inhibition and apoptosis were observed in CI-1040-treated tumor cells with mutations in either KRAS or BRAF in comparison with the ovarian cancer cells containing wild-type sequences. Long serial analysis of gene expression identified several differentially expressed genes in CI-1040-treated MPSC1 cells harboring an activating mutation in BRAF (V599L). The most striking changes were down-regulation of cyclin D1, COBRA1, and transglutaminase-2 and up-regulation of tumor necrosis factor-related apoptosis-induced ligand, thrombospondin-1, optineurin, and palladin. These patterns of gene expression were validated in other CI-1040-treated tumor cells based on quantitative PCR. Constitutive expression of cyclin D1 partially reversed the growth inhibitory effect of CI-1040 in MPSC1 cells. Our findings indicate that an activated MAPK pathway is critical in tumor growth and survival of ovarian tumors with KRAS or BRAF mutations and suggest that the CI-1040 induced phenotypes depend on the mutational status of KRAS and BRAF in ovarian tumors.


Assuntos
Cistadenocarcinoma Seroso , Genes ras/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação/genética , Neoplasias Ovarianas , Proteínas Proto-Oncogênicas B-raf/genética , Transdução de Sinais , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Células Tumorais Cultivadas
19.
Am J Physiol Cell Physiol ; 288(2): C304-13, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15456698

RESUMO

The voltage-dependent K+ channel 4.3 (Kv4.3) is one of the major molecular correlates encoding a class of rapidly inactivating K+ currents, including the transient outward current in the heart (Ito) and A currents (IA) in neuronal and smooth muscle preparations. Recent studies have shown that Ito in human atrial myocytes and IA in murine colonic myocytes are modulated by Ca2+/calmodulin-dependent protein kinase II (CaMKII); however, the molecular target of CaMKII in these studies has not been elucidated. We performed experiments to investigate whether CaMKII could regulate Kv4.3 currents directly. Inclusion of the autothiophosphorylated form of CaMKII in the patch pipette (10 nM) prolonged Kv4.3 currents such that the time required to reach 50% inactivation from peak more than doubled, with positive shifts in voltage dependence of both activation and inactivation. In contrast, the rate of recovery from inactivation was accelerated under these conditions. CaMKII-inhibitory peptide or KN-93 produced effects opposite to that above; thus the rate of inactivation was increased, and recovery from inactivation decreased. A number of mutagenesis experiments were conducted on the three candidate CaMKII consensus sequence sites on the channel. Mutations at S550A, located at the COOH-terminal region of the channel, resulted in currents that inactivated more rapidly but recovered from inactivation at a slower rate than that of wild-type controls. In addition, these currents were unaffected by dialysis with either autothiophosphorylated CaMKII or the specific inhibitory peptide of CaMKII, suggesting that CaMKII slows the inactivation and accelerates the rate of recovery from inactivation of Kv4.3 currents by a direct effect at S550A, located at the COOH-terminal region of the channel.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Potenciais da Membrana/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Linhagem Celular , Humanos , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Canais de Potássio Shal , Sulfonamidas/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...