Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Neurosci ; 131(10): 975-983, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32378973

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common aggressive primary cancer occurring in the brain tissue. GBM accounts 16% of primary brain tumors and half of gliomas. Additionally, the incidence of GBM is increases with aging, and reaches the peak at the age of 75 to 84 years. The survival of patients with GBM remains at a low level, only less than 5% patients diagnosed with GBM survive for 5 years. Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM. TMZ combined with radiation therapy has been shown to prolong the overall survival (OS) to 14.6 months compared with 12.1 months for radiation therapy alone. NF-E2-related factor 2 (Nrf2) is a transcription factor that contains seven functional domains. The binding of Keap1 to Nrf2 is a central regulator of the cellular defense mechanism against environmental stresses. METHODS: First, Nrf2 overexpression and inhibition models were constructed in U251 cells using transfection. The percentage of viable cells was detected using the MTT assay. Then, the expression of the HO-1 regulator was detected using qPCR, and the concentrations of oxidative stress related factors were detected using ELISAs. The levels of proteins related to oxidative stress and the Ras/Raf/MEK signaling pathway was detected using western blotting analysis. RESULTS: We initially established Nrf2 inhibition and activation cell models in U251 cells and found that the inhibition of Nrf2 expression decreased the mRNA and protein levels of the anti-oxidative enzymes, as well as the secretion of these enzymes into the cellular microenvironment. These effects might be mediated by the inhibition of Ras/Raf/MEK signaling pathway, leading to the inhibition of cellular proliferation. CONCLUSIONS: Inhibition of Nrf2 expression might enhance the effect of TMZ on the treatment of GBM and might be a new therapeutic strategy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Glioma/tratamento farmacológico , MAP Quinase Quinase Quinases/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Temozolomida/farmacologia , Quinases raf/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Quinases raf/antagonistas & inibidores , Proteínas ras/antagonistas & inibidores
2.
Life Sci ; 239: 116916, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31626792

RESUMO

AIMS: To investigate the effects of sevoflurane on proliferation, cell cycle, apoptosis, autophagy, invasion and epithelial-mesenchymal transition of colon cancer cell line SW480, and to explore its possible mechanism. MATERIALS AND METHODS: SW480 and SW620 cells were treated with a mixture of 95% O2+5% CO2 containing different concentrations of sevoflurane (1.7% SAV, 3.4% SAV and 5.1% SAV) for 6 h. Meanwhile, we performed a rescue experiment by treating cells with the ERK pathway activator LM22B-10 prior to treatment of cells with 5.1% sevoflurane。 KEY FINDINGS: High concentration (5.1%) of sevoflurane significantly inhibited the proliferation and invasion of cells, causing G0/G1 phase arrest and promoted apoptosis and autophagy. 5.1% sevoflurane can participate in the regulation of EMT by regulating the expression of E-cadherin, Vimentin and N-cadherin proteins. LM22B-10 promoted proliferation and invasion of cancer cells and inhibited apoptosis and autophagy, while 5.1% sevoflurane could reverse the effect of LM22B-10 on the biological characteristics of cells. Sevoflurane can significantly inhibit tumor growth in SW480 cells transplanted nude mice. Moreover, 5.1% sevoflurane significantly increased the expression of p-Raf, p-MEK1/2, and p-ERK1/2 in SW480 cells and tumor tissues without affecting p-JNK and p-p38 proteins, meanwhile, 5.1% sevoflurane can inhibit the activation of ERK signaling pathway by LM22B-10 in vitro and in vivo. SIGNIFICANCE: Sevoflurane can inhibit the proliferation and invasion of colon cancer cells, induce apoptosis and autophagy, and participate in the regulation of epithelial-mesenchymal transition, which may be related to its inhibition of the ERK signaling pathway.


Assuntos
Neoplasias do Colo/metabolismo , Sevoflurano/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Caderinas/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , China , Neoplasias do Colo/fisiopatologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sevoflurano/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos
3.
Cardiovasc Toxicol ; 16(1): 46-53, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25701259

RESUMO

Sunitinib is used extensively in the treatment of metastatic renal cell carcinoma and imatinib-resistant gastrointestinal stromal tumors. However, the undesirable cardiotoxic effects of sunitinib, such as congestive heart failure and hypertension, limit its use in the clinical setting. As multiple receptor tyrosine kinases are inhibited by sunitinib, it raises a question as to which target mediates sunitinib-induced cardiotoxicity. Here, we reported that the injection of fibroblast growth factor 2 (FGF2) mRNA into one- to two-cell stage embryos protected against sunitinib-induced cardiotoxicity in zebrafish. In addition, FGF2 significantly prevented sunitinib-induced cardiotoxicity in cardiomyoblast H9c2 cells, possibly via activating the PLC-γ/c-Raf/CREB pathway. Importantly, FGF2 did not compromise the antitumor activity of sunitinib in Caki-1 and OS-RC-2 renal cell carcinoma cells. Molecular docking simulations further revealed an interaction between the tyrosine kinase domain of FGF receptor 1 (FGFR1) and sunitinib. Taken together, our results clearly demonstrated that FGF2 inhibition plays an important role in sunitinib-induced cardiotoxicity both in vitro and in vivo. This study also provided a basis for further research on sunitinib-induced cardiotoxicity and may allow rational design of new sunitinib derivatives with fewer or weak cardiotoxic effects.


Assuntos
Inibidores da Angiogênese/toxicidade , Cardiotoxicidade/prevenção & controle , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Cardiopatias/induzido quimicamente , Cardiopatias/prevenção & controle , Indóis/antagonistas & inibidores , Indóis/toxicidade , Mioblastos/efeitos dos fármacos , Pirróis/antagonistas & inibidores , Pirróis/toxicidade , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Fosfolipase C gama/efeitos dos fármacos , Fosfolipase C gama/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Sunitinibe , Peixe-Zebra , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
4.
Pharmazie ; 69(3): 229-33, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24716415

RESUMO

BACKGROUND: Curcumin is a multi-targeted anti-cancer agent. However, there are few studies on its anti-leukemia activity in human acute monocytic leukemia. Here, we study the effect and mechanisms of curcumin on acute monocytic leukemia. METHODS: The acute monocytic leukemia cell line THP-1 was used as in vitro cell model to explore the anti-leukemia effects and mechanisms of curcumin. Cell proliferation was measured by MTT assay, cell apoptosis bodies were observed using a light microscope, cell apoptosis rate was evaluated by flow cytometry, and the expression alterations of growth-sinaling proteins were detected by Western blotting. RESULTS: Curcumin inhibited cell proliferation and induced cell apoptosis in time- and dose- dependent manner in THP-1 cells. Curcumin significantly inhibited the activations of AKT/mTOR and RAF/MEK/ERK signaling pathways simultaneously. CONCLUSION: This study demonstrates that curcumin inhibits proliferation and induces apoptosis in THP-1 cells via inhibiting the activations of AKT/mTOR and RAF/MEK/ERK signaling pathways simultaneously. Our data suggest that curcumin is a promising anti-tumor agent in acute monocytic leukemia.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Curcumina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , MAP Quinase Quinase Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Oncogênica v-akt/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Anexina A5 , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Corantes , Ativação Enzimática/efeitos dos fármacos , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Humanos , Leucemia Mieloide Aguda/patologia , Sais de Tetrazólio , Tiazóis
5.
Cancer Treat Rev ; 40(6): 750-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24613036

RESUMO

The hedgehog (Hh) pathway is aberrantly activated in a number of tumors. In medulloblastoma, basal cell carcinoma, and rhabdomyosarcoma, mutations in Hh pathway genes lead to ligand-independent pathway activation. In many other tumor types, ligand-dependent activation of Hh signaling is potentiated through crosstalk with other critical molecular signaling pathways. Among such pathways, RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, EGFR, and Notch are of particular interest because agents that selectively inhibit these pathways are available and can be readily combined with agents such as vismodegib, sonidegib (LDE225), and BMS-833923, which target smoothened-a key Hh pathway regulator. Numerous preclinical studies have revealed the ways in which Hh intersects with each of these pathways, and combination therapies have resulted in improved antitumor efficacy and survival in animal models. Hh also plays an important role in hematopoiesis and in the maintenance of BCR-ABL-driven leukemic stem cells. Thus, combined inhibition of the Hh pathway and BCR-ABL has emerged as a promising potential therapeutic strategy in chronic myeloid leukemia (CML). A number of clinical trials evaluating combinations of Hh inhibitors with other targeted agents are now underway in CML and a variety of solid tumors. This review highlights these trials and summarizes preclinical evidence of crosstalk between Hh and four other actionable pathways-RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, EGFR, and Notch-as well as the role of Hh in the maintenance of BCR-ABL-driven leukemic stem cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Hedgehog/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptor Cross-Talk/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Proteínas de Fusão bcr-abl/efeitos dos fármacos , Proteínas de Fusão bcr-abl/metabolismo , Proteínas Hedgehog/efeitos dos fármacos , Humanos , Janus Quinase 2/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores Notch/efeitos dos fármacos , Receptores Notch/metabolismo , Serina-Treonina Quinases TOR/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
6.
Toxicol Appl Pharmacol ; 271(2): 229-38, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23707609

RESUMO

Resistance to therapy is the major obstacle to more effective cancer treatment. Heme oxygenase-1 (HO-1) is often highly up-regulated in tumor tissues, and its expression is further increased in response to therapies. It has been suggested that inhibition of HO-1 expression is a potential therapeutic approach to sensitize tumors to chemotherapy and radiotherapy. In this study, we tested the hypothesis that the anti-tumor effects of metformin are mediated by suppression of HO-1 expression in cancer cells. Our results indicate that metformin strongly suppresses HO-1 mRNA and protein expression in human hepatic carcinoma HepG2, cervical cancer HeLa, and non-small-cell lung cancer A549 cells. Metformin also markedly reduced Nrf2 mRNA and protein levels in whole cell lysates and suppressed tert-butylhydroquinone (tBHQ)-induced Nrf2 protein stability and antioxidant response element (ARE)-luciferase activity in HepG2 cells. We also found that metformin regulation of Nrf2 expression is mediated by a Keap1-independent mechanism and that metformin significantly attenuated Raf-ERK signaling to suppress Nrf2 expression in cancer cells. Inhibition of Raf-ERK signaling by PD98059 decreased Nrf2 mRNA expression in HepG2 cells, confirming that the inhibition of Nrf2 expression is mediated by an attenuation of Raf-ERK signaling in cancer cells. The inactivation of AMPK by siRNA, DN-AMPK or the pharmacological AMPK inhibitor compound C, revealed that metformin reduced HO-1 expression in an AMPK-independent manner. These results highlight the Raf-ERK-Nrf2 axis as a new molecular target in anticancer therapy in response to metformin treatment.


Assuntos
Heme Oxigenase-1/antagonistas & inibidores , Hipoglicemiantes/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metformina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/fisiologia , Neoplasias/enzimologia , Quinases raf/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Núcleo Celular/química , Sobrevivência Celular/efeitos dos fármacos , Citosol/química , Galactosidases/metabolismo , Humanos , Indicadores e Reagentes , Luciferases/metabolismo , Plasmídeos/genética , RNA/biossíntese , RNA/genética , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transfecção
7.
Food Chem Toxicol ; 46(12): 3800-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18951945

RESUMO

The flavonoid naringin has been shown to play a role in preventing the development of cardiovascular disease. However, the exact molecular mechanisms underlying the roles of integrated cell cycle regulation and MAPK signaling pathways in the regulation of naringin-induced inhibition of cell proliferation in vascular smooth muscle cells (VSMCs) remain to be identified. Naringin treatment resulted in significant growth inhibition and G(1)-phase cell cycle arrest mediated by induction of p53-independent p21WAF1 expression; expression of cyclins and CDKs in VSMCs was also down-regulated. In addition, among the pathways examined, blockade of ERK function inhibited naringin-dependent p21WAF1 expression, reversed naringin-mediated inhibition of cell proliferation and decreased cell cycle proteins. Moreover, naringin treatment increased both Ras and Raf activations. Transfection of cells with dominant negative Ras (RasN17) and Raf (RafS621A) mutant genes suppressed naringin-induced ERK activity and p21WAF1 expression. Finally, naringin-induced reduction in cell proliferation and cell cycle protein was abolished in the presence of RasN17 and RafS621A mutant genes. The Ras/Raf/ERK pathway participates in p21WAF1 induction, leading to a decrease in cyclin D1/CDK4 and cyclin E/CDK2 complexes and in naringin-dependent inhibition of cell growth. These novel and unexpected findings provide a theoretical basis for preventive use of flavonoids to the atherosclerosis disease.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Flavanonas/farmacologia , Fase G1/efeitos dos fármacos , Genes ras/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Quinases raf/fisiologia , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/fisiologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Flavonoides/farmacologia , Genes ras/efeitos dos fármacos , Imunoprecipitação , Mutação/fisiologia , Ratos , Ratos Sprague-Dawley , Timidina/metabolismo , Transfecção , Quinases raf/efeitos dos fármacos
8.
J Neurochem ; 102(6): 1905-1917, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17767704

RESUMO

Several G protein-coupled receptors (GPCRs) mediate neuronal cell migration and survival upon activation by their native peptide ligands but activate death-signaling pathways when activated by certain non-native ligands. In cultured neurons, we recently described expression of the unique seven-transmembrane (7TM) -G protein-coupled receptor, APJ, which is also strongly expressed in neurons in the brain and various cell types in other tissues. We now demonstrate that the endogenous APJ peptide ligand apelin activates signaling pathways in rat hippocampal neurons and modulates neuronal survival. We found that (i) both APJ and apelin are expressed in hippocampal neurons; (ii) apelin peptides induce phosphorylation of the cell survival kinases AKT and Raf/ERK-1/2 in hippocampal neurons; and (iii) apelin peptides protect hippocampal neurons against NMDA receptor-mediated excitotoxicity, including that induced by human immunodeficiency virus type 1. Thus, apelin/APJ signaling likely represents an endogenous hippocampal neuronal survival response, and therefore apelin should be further investigated as a potential neuroprotectant against hippocampal injury.


Assuntos
Proteínas de Transporte/farmacologia , Citoproteção/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/antagonistas & inibidores , Animais , Apelina , Receptores de Apelina , Proteínas de Transporte/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Citoproteção/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , HIV-1/fisiologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
9.
Kidney Int ; 72(2): 135-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17625579

RESUMO

Tubule-cell hyperproliferation precedes cyst development in autosomal dominant polycystic kidney disease (ADPKD). Parker et al. report that insulin-like growth factor-1 stimulates ADPKD cell proliferation by activating Ras and Raf; inhibition of Ras or Raf abolished this effect. Inhibiting tubule-cell proliferation could halt cyst formation and prolong survival of functional tubules, offering a new treatment for ADPKD patients.


Assuntos
Proliferação de Células/efeitos dos fármacos , Túbulos Renais/patologia , Rim Policístico Autossômico Dominante/tratamento farmacológico , Cistos/prevenção & controle , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Rim Policístico Autossômico Dominante/patologia , Transdução de Sinais , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo , Proteínas Ativadoras de ras GTPase/efeitos dos fármacos , Proteínas Ativadoras de ras GTPase/metabolismo
10.
Kidney Int ; 72(2): 157-65, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17396115

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) largely results from mutations in the PKD1 gene leading to hyperproliferation of renal tubular epithelial cells and consequent cyst formation. Rodent models of PKD suggest that the multifunctional hormone insulin-like growth factor-1 (IGF-1) could play a pathogenic role in renal cyst formation. In order to test this possibility, conditionally immortalized renal epithelial cells were prepared from normal individuals and from ADPKD patients with known germline mutations in PKD1. All patient cell lines had a decreased or absence of polycystin-1 but not polycystin-2. These cells had an increased sensitivity to IGF-1 and to cyclic AMP, which required phosphatidylinositol-3 (PI3)-kinase and the mitogen-activated protein kinase, extracellular signal-regulated protein kinase (ERK) for enhanced growth. Inhibition of Ras or Raf abolished the stimulated cell proliferation. Our results suggest that haploinsufficiency of polycystin-1 lowers the activation threshold of the Ras/Raf signalling system leading to growth factor-induced hyperproliferation. Inhibition of Ras or Raf activity may be a therapeutic option for decreasing tubular cell proliferation in ADPKD.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP , Quinases raf/efeitos dos fármacos , Proteínas Ativadoras de ras GTPase/efeitos dos fármacos , Linhagem Celular , Cistos/patologia , Mutação em Linhagem Germinativa , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Túbulos Renais/patologia , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases raf/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
11.
Semin Oncol ; 33(4): 392-406, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16890795

RESUMO

Improvements in our understanding of the molecular basis of cancer have led to the clinical development of protein kinase inhibitors, which target pivotal molecules involved in intracellular signaling pathways implicated in tumorigenesis and progression. These novel targeted agents have demonstrated activity against a wide range of solid tumors, are generally better tolerated than standard chemotherapeutics, and may revolutionize the management of advanced refractory cancer. The ubiquitous Raf serine/threonine kinases are pivotal molecules within the Raf/mitogen extracellular kinase (MEK)/extracellular signal-related kinase (ERK) signaling pathway, which regulates cellular proliferation and survival. Raf kinase isoforms (wild-type Raf-1 or the b-raf V600E oncogene) are overactivated in a variety of solid tumor types, including renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), non-small cell lung cancer (NSCLC), melanoma, and papillary thyroid carcinoma. In this review, the role of Raf in normal cells and in cancer is discussed, and an overview is given of Raf inhibitors currently in development, focusing on sorafenib tosylate (BAY 43-9006 or sorafenib). Sorafenib is the first oral multi-kinase inhibitor to be developed that targets Raf kinases (Raf-1, wild-type B-Raf, and b-raf V600E), in addition to receptor tyrosine kinases associated with angiogenesis (vascular endothelial growth factor receptor [VEGFR]-2/-3, platelet-derived growth factor receptor [PDGFR]-beta) or tumor progression (Flt-3, c-kit). Preclinical and clinical sorafenib data that led to its recent approval for the treatment of advanced RCC are summarized, along with current thinking on sorafenib's mechanism of effect on the tumor and tumor vasculature in melanoma and RCC.


Assuntos
Antineoplásicos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Neoplasias/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Benzenossulfonatos/farmacologia , Benzenossulfonatos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Transformação Celular Neoplásica/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/química , Melanoma/tratamento farmacológico , Camundongos , Neoplasias/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia , Isoformas de Proteínas , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Sorafenibe , Quinases raf/química
12.
Mol Cancer Ther ; 5(3): 665-75, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16546981

RESUMO

The cellular and molecular effects of the proteasome inhibitor bortezomib on breast cancer cells are as yet poorly characterized. Here, in a panel of six breast cancer cell lines, bortezomib reduced viability in a concentration-dependent, time-dependent, and cell line-dependent manner. Proteasome activity was relatively high in two of the three more resistant cell lines. No relationship was observed between bortezomib effects on cell viability and expression/phosphorylation of HER-2, epidermal growth factor receptor (EGFR), AKT, or extracellular signal-regulated kinase 1/2 (ERK1/2). Molecular effects of bortezomib were further studied in SK-BR-3 and BT-474 cells because they share expression of EGFR and overexpression of HER-2 while, in contrast, SK-BR-3 cells were 200-fold more sensitive to this agent. Proteasome activity was inhibited to a similar extent in the two cell lines, and known proteasome substrates accumulated similarly. In SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration of bortezomib. In contrast, phosphorylation of Raf/mitogen-activated protein kinase kinase 1/2 (MEK 1/2)/ERK1/2 increased by bortezomib. In BT-474 cells, the effects were much less pronounced. Treatment of SK-BR-3 cells with bortezomib combined with pharmacologic inhibitors of EGFR, phosphatidylinositol 3'-kinase, or MEK resulted in modest or no enhancement of the effects on cell viability. Collectively, these results show that bortezomib has differential cellular and molecular effects in human breast cancer cells. The bortezomib-observed effects on signaling transduction molecules might be relevant to help to design mechanistic-based combination treatments.


Assuntos
Ácidos Borônicos/farmacologia , Neoplasias da Mama/enzimologia , Inibidores de Cisteína Proteinase/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Bortezomib , Proteínas de Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Fosfatase 1 de Especificidade Dupla , Ativação Enzimática , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Proteínas Imediatamente Precoces/efeitos dos fármacos , MAP Quinase Quinase 4/efeitos dos fármacos , MAP Quinase Quinase Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Fosfoproteínas Fosfatases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1 , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Tirosina Fosfatases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores Proteína Tirosina Quinases , Receptor ErbB-2/antagonistas & inibidores , Quinases raf/efeitos dos fármacos
13.
Leukemia ; 19(1): 18-26, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15470486

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically associated with several cancers including Kaposi's sarcoma (KS), primary effusion lymphoma, and multicentric Castleman's disease. KSHV-mediated pathogenesis is dependent mainly on KSHV infection as well as on the microenvironment provided by the growth factors (GFs)/inflammatory cytokines (ICs). Recently, we determined that oncoprotein Raf enhances KSHV infection of target cells. Interestingly, Raf regulates the expression of a variety of GFs/ICs including those involved in angiogenesis such as vascular endothelial growth factor (VEGF). In this review, we discuss the effect of the Raf-GF/IC autocrine/paracrine loop on KSHV infection of both hematopoietic and nonhematopietic cells, and associated disease conditions.


Assuntos
Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Humano 8/patogenicidade , Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Humanos , Fator A de Crescimento do Endotélio Vascular/genética , Quinases raf/genética
14.
J Cell Sci ; 118(Pt 2): 313-22, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15615777

RESUMO

The Wnt family of proteins regulates development and cell growth. We identified Wnt3a-based regulatory mechanisms for cell proliferation in NIH3T3 fibroblast cells. The degree of Wnt3a-induced proliferation was reduced by beta-catenin small interfering RNA (siRNA) and extracellular signal-regulated kinase (ERK) siRNA, indicating that both the ERK and Wnt/beta-catenin pathways are involved in Wnt3a-induced proliferation. Wnt3a immediately and transiently activated the Raf-1-MEK-ERK cascade in a manner distinct from that of the beta-catenin increase seen in cells treated with Wnt3a. Wnt3a-induced ERK activation was maintained even though basal ERK activities were reduced by beta-catenin siRNA, indicating that Wnt3a may activate the ERK pathway independently of beta-catenin. The ERK pathway was however, activated by beta-catenin transfection, which was abolished by co-transfection with dominant-negative Tcf-4. Therefore, ERK pathway activation by Wnt signaling could occur at multiple levels, including beta-catenin-independent direct signaling resulting from a Wnt3a and beta-catenin/Tcf-4-dependent post gene transcriptional event. Wnt3a stimulated the G1 to S phase cell cycle progression. This stimulation was reduced by the ERK pathway inhibitor, indicating that Wnt3a promotes proliferation by stimulating the ERK pathway. Wnt3a therefore stimulates the proliferation of fibroblast cells, at least in part, via activation of the ERK and Wnt/beta-catenin pathways.


Assuntos
Proteínas do Citoesqueleto/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proteínas do Citoesqueleto/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fase G1/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células NIH 3T3 , Proteínas/farmacologia , RNA Interferente Pequeno/metabolismo , Fase S/efeitos dos fármacos , Transativadores/efeitos dos fármacos , Proteínas Wnt , Proteína Wnt3 , Proteína Wnt3A , beta Catenina , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
15.
J Cell Physiol ; 201(2): 244-58, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15334659

RESUMO

Epidermal growth factor (EGF) and Ras mitogenic signal transduction pathways are frequently activated in breast carcinoma and inhibit mammary differentiation and apoptosis. HC11 mouse mammary epithelial cells, which differentiate and synthesize beta-casein following growth to confluency and stimulation with lactogenic hormones, were used to study EGF-dependent signaling during differentiation. Blocking Mek-Erk or phosphotidylinositol-3-kinase (PI-3 kinase) signaling with specific chemical inhibitors enhanced beta-casein promotor-driven luciferase activity. Because EGF stimulation of HC11 cells resulted in the activation of Ras, the effect of activated Ras (RasV12) or dominant negative (DNRasN17) on lactogen induced differentiation was examined. HC11 cell lines expressing RasV12 or DNRasN17 under the control of a tetracycline (tet)-responsive promotor were constructed. Activated RasV12 expression resulted in reduced tyrosine phosphorylation of Stat5 and a delay in beta-casein expression in response to prolactin. However, the expression of tet-regulated DNRasN17 and adenovirus-encoded DNRasN17 enhanced Stat5 tyrosine phosphorylation, Stat5 DNA binding, and beta-casein transcription. The expression of DNRasN17 blocked the activation of the Mek-Erk pathway by EGF but did not prevent the phosphorylation of AKT, a measure of activation of the PI-3-kinase pathway. Moreover, the expression of DNRasN17 prevented the block to lactogenic differentiation induced by EGF. Stimulation of HC11 cells with prolactin resulted in the association of the SHP2 phosphatase with Stat5, and this association was prevented by DNRasN17 expression. These results demonstrate that in HC11 cells DNRas inhibits the Mek-Erk pathway and enhances lactogenic hormone-induced differentiation. This occurs, in part, by inhibiting the association of the SHP2 phosphatase with Stat5.


Assuntos
Diferenciação Celular/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Glândulas Mamárias Animais/citologia , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Animais , Northern Blotting , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Imunoprecipitação , Camundongos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Prolactina/metabolismo , Prolactina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo , Proteínas ras/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...