Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Parasit Vectors ; 16(1): 371, 2023 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-37858158

RESUMO

BACKGROUND: Toxoplasmosis is a zoonosis with a worldwide presence that is caused by the intracellular parasite Toxoplasma gondii. Active regulation of apoptosis is an important immune mechanism by which host cells resist the growth of T. gondii or avoid excessive pathological damage induced by this parasite. Previous studies found that upregulated expression of microRNA-185 (miR-185) during T. gondii infection has a potential role in regulating the expression of the ARAF gene, which is reported to be associated with cell proliferation and apoptosis. METHODS: The expression levels of miR-185 and the ARAF gene were evaluated by qPCR and Western blot, respectively, in mice tissues, porcine kidney epithelial cells (PK-15) and porcine alveolar macrophages (3D4/21) following infection with the T. gondii ToxoDB#9 and RH strains. The dual luciferase reporter assay was then used to verify the relationship between miR-185 and ARAF targets in PK-15 cells. PK-15 and 3D4/21 cell lines with stable knockout of the ARAF gene were established by CRISPR, and then the apoptosis rates of the cells following T. gondii infection were detected using cell flow cytometry assays. Simultaneously, the activities of cleaved caspase-3, as a key apoptosis executive protein, were detected by Western blot to evaluate the apoptosis levels of cells. RESULTS: Infection with both the T. gondii ToxoDB#9 and RH strains induced an increased expression of miR-185 and a decreased expression of ARAF in mice tissues, PK-15 and 3D4/21 cells. MiR-185 mimic transfections showed a significantly negative correlation in expression levels between miR-185 and the ARAF gene. The dual luciferase reporter assay confirmed that ARAF was a target of miR-185. Functional investigation revealed that T. gondii infection induced the apoptosis of PK-15 and 3D4/21 cells, which could be inhibited by ARAF knockout or overexpression of miR-185. The expression levels of cleaved caspase-3 protein were significantly lower in cells with ARAF knockout than in normal cells, which were consistent with the results of the cell flow cytometry assays. CONCLUSIONS: Toxoplasma gondii infection could lead to the upregulation of miR-185 and the downregulation of ARAF, which was not related to the strain of T. gondii and the host cells. Toxoplasma gondii infection could regulate the apoptosis of host cells via the miR-185/ARAF axis, which represents an additional strategy used by T. gondii to counteract host-cell apoptosis in order to maintain survival and reproduce in the host cells.


Assuntos
MicroRNAs , Proteínas Proto-Oncogênicas A-raf , Doenças dos Suínos , Toxoplasma , Toxoplasmose , Animais , Camundongos , Apoptose/genética , Apoptose/imunologia , Caspase 3 , Células Cultivadas , Luciferases , MicroRNAs/genética , MicroRNAs/metabolismo , Suínos/genética , Suínos/metabolismo , Suínos/parasitologia , Doenças dos Suínos/genética , Doenças dos Suínos/metabolismo , Doenças dos Suínos/parasitologia , Toxoplasmose/genética , Toxoplasmose/metabolismo , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas A-raf/metabolismo
2.
Mol Cell ; 82(13): 2443-2457.e7, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35613620

RESUMO

RAF protein kinases are effectors of the GTP-bound form of small guanosine triphosphatase RAS and function by phosphorylating MEK. We showed here that the expression of ARAF activated RAS in a kinase-independent manner. Binding of ARAF to RAS displaced the GTPase-activating protein NF1 and antagonized NF1-mediated inhibition of RAS. This reduced ERK-dependent inhibition of RAS and increased RAS-GTP. By this mechanism, ARAF regulated the duration and consequences of RTK-induced RAS activation and supported the RAS output of RTK-dependent tumor cells. In human lung cancers with EGFR mutation, amplification of ARAF was associated with acquired resistance to EGFR inhibitors, which was overcome by combining EGFR inhibitors with an inhibitor of the protein tyrosine phosphatase SHP2 to enhance inhibition of nucleotide exchange and RAS activation.


Assuntos
Neurofibromina 1 , Proteínas Proto-Oncogênicas A-raf , Proteínas Ativadoras de ras GTPase , Receptores ErbB/genética , Receptores ErbB/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Neurofibromina 1/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas A-raf/metabolismo , Transdução de Sinais , Proteínas Ativadoras de ras GTPase/metabolismo
3.
Cancer Lett ; 494: 107-120, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32814086

RESUMO

Pancreatic cancer (PC) is a malignant cancer with high mortality and poor prognosis. In this study, we found that Linc01232 was significantly upregulated in PC tissues and cells and higher Linc01232 expression was associated with poorer prognosis. Linc01232 overexpression promoted and Linc01232 knockdown inhibited the migration and invasion of PC cells. The results of RNA pull-down, RNA Binding Protein Immunoprecipitation (RIP) assays revealed that Linc01232 physically interacted with Heterogeneous Nuclear Ribonucleoprotein A2/B1 (HNRNPA2B1) (680-890 nt fragment with the RNA recognition motif 2 domain) to inhibit its ubiquitin-mediated degradation in PC cells. RNA sequencing was performed to obtain the transcriptional profiles regulated by Linc01232 and we further demonstrated that Linc01232 participated in the alternative splicing of A-Raf by stabilizing HNRNPA2B1 and subsequently regulated the MAPK/ERK signaling pathway. Collected, our study showed that Linc01232/HNRNPA2B1/A-Raf/MAPK axis participated in the progression of PC and provided a potential therapeutic target for PC.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas A-raf/metabolismo , RNA Longo não Codificante/genética , Ubiquitina/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Prognóstico , Proteólise , Proteínas Proto-Oncogênicas A-raf/genética , Análise de Sequência de RNA , Regulação para Cima
4.
Anticancer Agents Med Chem ; 19(2): 236-247, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30324893

RESUMO

BACKGROUND: Identification of events leading to hepatocellular carcinoma (HCC) progression is essential for understanding its pathophysiology. The aims of this study are to identify and characterize differentially expressed proteins in serum of HCC-bearing rats and the corresponding controls during cancer initiation, progression and tumorigenesis. METHODS: Chemical carcinogens, N-Nitrosodiethylamine and 2-aminoacetylfluorine are administered to induce HCC to male Wistar rats. The 2D-Electrophoresis and PD-Quest analyses are performed to identify several differentially expressed proteins in serum of HCC-bearing animals. These proteins are further characterized by MALDI-TOF-MS/MS analyses. Using pathwaylinker a HCC-specific network is analyzed among the MALDITOF- MS/MS characterized proteins and their interactors. RESULTS: Carcinogen administration caused inflammation leading to liver injury and HCC development. Liver inflammation was confirmed by increase in the levels of TNF-α and IL-6 in carcinogen treated rats. We report significant increase in expression of two differentially expressed proteins, namely, A-Raf and Fatty Acid 2- Hydroxylase (FA2H), at early stage of HCC initiation, during its progression and at tumor stage. Real-time PCR analysis of mRNA for these proteins confirmed up-regulation of their transcripts. Further, we validated our experimental data with sera of clinically confirmed liver cancer patients. CONCLUSION: The study suggests that FA2H and A-Raf play a major role in the progression of HCC.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas A-raf/genética , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/metabolismo , Dietilnitrosamina , Humanos , Metabolismo dos Lipídeos/genética , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Masculino , Oxigenases de Função Mista/sangue , Oxigenases de Função Mista/metabolismo , Proteômica , Proteínas Proto-Oncogênicas A-raf/sangue , Proteínas Proto-Oncogênicas A-raf/metabolismo , RNA Mensageiro/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
5.
Biochem Soc Trans ; 46(6): 1393-1406, 2018 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30381334

RESUMO

RAF (rapidly accelerated fibrosarcoma) Ser/Thr kinases (ARAF, BRAF, and CRAF) link the RAS (rat sarcoma) protein family with the MAPK (mitogen-activated protein kinase) pathway and control cell growth, differentiation, development, aging, and tumorigenesis. Their activity is specifically modulated by protein-protein interactions, post-translational modifications, and conformational changes in specific spatiotemporal patterns via various upstream regulators, including the kinases, phosphatase, GTPases, and scaffold and modulator proteins. Dephosphorylation of Ser-259 (CRAF numbering) and dissociation of 14-3-3 release the RAF regulatory domains RAS-binding domain and cysteine-rich domain for interaction with RAS-GTP and membrane lipids. This, in turn, results in RAF phosphorylation at Ser-621 and 14-3-3 reassociation, followed by its dimerization and ultimately substrate binding and phosphorylation. This review focuses on structural understanding of how distinct binding partners trigger a cascade of molecular events that induces RAF kinase activation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animais , Humanos , Proteínas Quinases Ativadas por Mitógeno/genética , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-raf/genética
6.
Cell Death Dis ; 9(9): 926, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206208

RESUMO

Trophoblast dysfunction is one mechanism implicated in the etiology of recurrent miscarriage (RM). Regulation of trophoblast function, however, is complex and the mechanisms contributing to dysregulation remain to be elucidated. Herein, we found EIF5A1 expression levels to be significantly decreased in cytotrophoblasts in RM villous tissues compared with healthy controls. Using the HTR-8/SVneo cell line as a model system, we found that overexpression of EIF5A1 promotes trophoblast proliferation, migration and invasion in vitro. Knockdown of EIF5A1 or inhibiting its hypusination with N1-guanyl-1,7-diaminoheptane (GC7) suppresses these activities. Similarly, mutating EIF5A1 to EIF5A1K50A to prevent hypusination abolishes its effects on proliferation, migration and invasion. Furthermore, upregulation of EIF5A1 increases the outgrowth of trophoblasts in a villous explant culture model, whereas knockdown has the opposite effect. Suppression of EIF5A1 hypusination also inhibits the outgrowth of trophoblasts in explants. Mechanistically, ARAF mediates the regulation of trophoblast migration and invasion by EIF5A1. Hypusinated EIF5A1 regulates the integrin/ERK signaling pathway via controlling the translation of ARAF. ARAF level is also downregulated in trophoblasts of RM villous tissues and expression of ARAF is positively correlated with EIF5A1. Together, our results suggest that EIF5A1 may be a regulator of trophoblast function at the maternal-fetal interface and low levels of EIF5A1 and ARAF may be associated with RM.


Assuntos
Movimento Celular/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Integrinas/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas de Ligação a RNA/metabolismo , Trofoblastos/metabolismo , Aborto Habitual/patologia , Linhagem Celular , Proliferação de Células , Feminino , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Fatores de Iniciação de Peptídeos/genética , Gravidez , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Fator de Iniciação de Tradução Eucariótico 5A
7.
Biochim Biophys Acta Proteins Proteom ; 1866(8): 849-856, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29777862

RESUMO

A-Raf is a member of the Raf kinase family. Unlike B-Raf and C-Raf, the functions of A-Raf remain obscure. To gain more insight into the biological functions of A-Raf, we investigated the A-Raf interactome using proteomics. We found 132 proteins that interact with A-Raf and confirmed the interaction of 12 of these proteins with A-Raf by western blotting. Our data suggested that A-Raf regulates apoptosis, RNA catabolism, GTPase activity, and cell adhesion by interacting with proteins located in different cellular compartments. We identified all ten hallmarks of cancer in these interacting proteins, suggesting that A-Raf is involved in carcinogenesis. Our results also indicated that A-Raf may play a role in different diseases and signaling pathways. These findings have identified potential regulators of A-Raf and provide a systemic insight into its biological functions.


Assuntos
Proteômica , Proteínas Proto-Oncogênicas A-raf/metabolismo , Apoptose , Western Blotting , Carcinogênese/genética , Adesão Celular , GTP Fosfo-Hidrolases/metabolismo , Células HEK293 , Humanos , Imunoprecipitação , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas A-raf/genética , RNA/metabolismo , Transdução de Sinais
8.
Anticancer Agents Med Chem ; 18(8): 1163-1176, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29732980

RESUMO

BACKGROUND: The network interactions link human disease proteins to regulatory cellular pathways leading to better understanding of protein functions and cellular processes. Revealing the network of signaling pathways in cancer through protein-protein interactions at molecular level enhances our understanding of Hepatocellular Carcinoma (HCC). OBJECTIVE: A rodent model for study of HCC was developed to identify differentially expressed proteins at very early stage of cancer initiation and throughout its progression. METHODOLOGY: HCC was induced by administrating N-Nitrosodiethylamine (DEN) and 2-aminoacetylfluorine (2-AAF) to male Wistar rats. Proteomic approaches such as 2D-Electrophoresis, PD-Quest, MALDI-TOF-MS and Western blot analyses have been used to identify, characterize and validate the differentially expressed proteins in HCC-bearing animals vis-a-vis controls. RESULTS: The step-wise analysis of morphological and histological parameters revealed HCC induction and tumorigenesis at 1 and 4 months after carcinogen treatment, respectively. We report a novel protein network of 735 different proteins out of which eight proteins are characterized by MALDI-TOF-MS analysis soon after HCC was chemically induced in rats. We have analyzed four different novel routes representing the association of experimentally identified proteins with HCC progression. CONCLUSION: The study suggests that A-Raf, transthyretin and epidermal growth factor receptor play major role in HCC progression by regulating MAPK signaling pathway and lipid metabolism leading to continuous proliferation, neoplastic transformation and tumorigenesis.


Assuntos
Carcinoma Hepatocelular/metabolismo , Biologia Computacional , Receptores ErbB/metabolismo , Neoplasias Hepáticas/metabolismo , Pré-Albumina/metabolismo , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas A-raf/metabolismo , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/patologia , Dietilnitrosamina , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/análise , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Masculino , Estrutura Molecular , Pré-Albumina/análise , Proteínas Proto-Oncogênicas A-raf/análise , Ratos Wistar , Relação Estrutura-Atividade
9.
Cell Signal ; 38: 212-222, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28743549

RESUMO

Apoptotic death of pancreatic ß cells is a major cause of type 2 diabetes mellitus (T2D) progression. Two isoforms of pyruvate kinase, PKM1 and PKM2, have been reported to participate in cell death in several cell types; however, little is known about their causal pathways in pancreatic ß-cell death. We examined whether the suppression of PKM1 or PKM2 affects endoplasmic reticulum (ER) stress-induced apoptosis in a pancreatic ß-cell line, MIN6, and Beta-TC-6 and found that knockdown of PKM1, but not of PKM2, leads to the induction of ER stress-induced apoptosis in these cells. We also investigated the mechanism by which PKM1 inhibits ER stress-induced apoptosis. We confirmed that PKM1 interacts with A-Raf, an upstream regulator of the MEK/ERK pathway, and that this interaction contributes to MEK1 phosphorylation by A-Raf. PKM1 knockdown suppresses the phosphorylation of MEK, ERK, and caspase-9 (Thr125), which is phosphorylated by the MEK/ERK pathway, thereby inhibiting the cleavage and activation of caspase-9. Thus, PKM1 knockdown activates the caspase-9/caspase-3 pathway under ER stress conditions and leads to apoptosis.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Estresse do Retículo Endoplasmático , Insulinoma/enzimologia , Insulinoma/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Hormônios Tireóideos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Ácido Pirúvico/metabolismo , eIF-2 Quinase/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
10.
J Biol Chem ; 292(8): 3164-3171, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28073913

RESUMO

Hypophosphatemia causes rickets by impairing hypertrophic chondrocyte apoptosis. Phosphate induction of MEK1/2-ERK1/2 phosphorylation in hypertrophic chondrocytes is required for phosphate-mediated apoptosis and growth plate maturation. MEK1/2 can be activated by numerous molecules including Raf isoforms. A- and B-Raf ablation in chondrocytes does not alter skeletal development, whereas ablation of C-Raf decreases hypertrophic chondrocyte apoptosis and impairs vascularization of the growth plate. However, ablation of C-Raf does not impair phosphate-induced ERK1/2 phosphorylation in vitro, but leads to rickets by decreasing VEGF protein stability. To determine whether Raf isoforms are required for phosphate-induced hypertrophic chondrocyte apoptosis, mice lacking all three Raf isoforms in chondrocytes were generated. Raf deletion caused neonatal death and a significant expansion of the hypertrophic chondrocyte layer of the growth plate, accompanied by decreased cleaved caspase-9. This was associated with decreased phospho-ERK1/2 immunoreactivity in the hypertrophic chondrocyte layer and impaired vascular invasion. These data further demonstrated that Raf kinases are required for phosphate-induced ERK1/2 phosphorylation in cultured hypertrophic chondrocytes and perform essential, but partially redundant roles in growth plate maturation.


Assuntos
Condrócitos/metabolismo , Condrogênese , Lâmina de Crescimento/crescimento & desenvolvimento , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , Apoptose , Desenvolvimento Ósseo , Células Cultivadas , Condrócitos/citologia , Condrócitos/patologia , Lâmina de Crescimento/metabolismo , Camundongos Endogâmicos C57BL , Fosfatos/metabolismo , Fosforilação , Isoformas de Proteínas/metabolismo , Quinases raf/metabolismo
11.
Oncotarget ; 8(6): 9251-9266, 2017 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-27999210

RESUMO

Activating mutations in the KRAS and BRAF genes, leading to hyperactivation of the RAS/RAF/MAPK oncogenic signaling cascade, are common in patients with colorectal cancer (CRC). While selective BRAF inhibitors are efficacious in BRAFmut melanoma, they have limited efficacy in BRAFmut CRC patients. In a RASmut background, selective BRAF inhibitors are contraindicated due to paradoxical activation of the MAPK pathway through potentiation of CRAF kinase activity. A way to overcome such paradoxical activation is through concurrent inhibition of the kinase activity of both RAF isoforms. Here, we further examined the effects of LY3009120, a panRAF and RAF dimer inhibitor, in human models of CRC with various mutational backgrounds. We demonstrate that LY3009120 induced anti-proliferative effects in BRAFmut and KRASmut CRC cell lines through G1-cell cycle arrest. The anti-proliferative effects of LY3009120 in KRASmut CRC cell lines phenocopied molecular inhibition of RAF isoforms by simultaneous siRNA-mediated knockdown of ARAF, BRAF and CRAF. Additionally, LY3009120 displayed significant activity in in vivo BRAFmut and KRASmut CRC xenograft models. Examination of potential resistance to LY3009120 demonstrated RAF-independent ERK and AKT activation in the KRASmut CRC cell line HCT 116. These findings describe the preclinical activity of a panRAF inhibitor in a BRAFmut and KRASmut CRC setting.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Mutação , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirimidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Predisposição Genética para Doença , Células HCT116 , Células HT29 , Humanos , Fenótipo , Proteínas Proto-Oncogênicas A-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Interferência de RNA , Ratos Nus , Fatores de Tempo , Transfecção , Carga Tumoral/efeitos dos fármacos
12.
Biochem Biophys Res Commun ; 476(4): 438-444, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27240957

RESUMO

Regulation of osteoblast activity by mechanical stress is important for bone remodeling. However, the precise mechanotransduction mechanism that triggers the anabolic reaction of osteoblasts is largely unknown. In this study, we performed RNA interference (RNAi) screening to identify the signaling molecules upstream of ERK, which was responsible for osteogenesis. Of twenty-two mitogen-activated protein kinase (MAPK) kinase kinases (MAP3Ks), we identified A-Raf and C-Raf as upstream MAP3Ks of the mechanical stretch-activated ERK pathway. Subsequently we screened the mechanosensitive cation channel, and identified P2X7 as an upstream molecule of the ERK pathway. Intriguingly, P2X7 functioned as an upstream activator of A-Raf but not of C-Raf. Furthermore, A-Raf contributed to mechanical stretch-induced osteoblast differentiation. In contrast, C-Raf but not A-Raf protected osteoblasts from mechanical stretch-induced apoptosis. These results suggested that A-Raf and C-Raf were involved in mechanobiological osteogenesis in a distinct way: A-Raf was responsible for osteogenesis while C-Raf for anti-apoptotic protection and promotion.


Assuntos
Osteoblastos/citologia , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Sobrevivência Celular , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoblastos/fisiologia , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas c-raf/genética , Interferência de RNA , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Estresse Mecânico
13.
Cell Death Differ ; 23(8): 1283-95, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26891695

RESUMO

A-Raf belongs to the family of oncogenic Raf kinases that are involved in mitogenic signaling by activating the MEK-ERK pathway. Low kinase activity of A-Raf toward MEK suggested that A-Raf might have alternative functions. We recently identified A-Raf as a potent inhibitor of the proapoptotic mammalian sterile 20-like kinase (MST2) tumor suppressor pathway in several cancer entities including head and neck, colon, and breast. Independent of kinase activity, A-Raf binds to MST2 thereby efficiently inhibiting apoptosis. Here, we show that the interaction of A-Raf with the MST2 pathway is regulated by subcellular compartmentalization. Although in proliferating normal cells and tumor cells A-Raf localizes to the mitochondria, differentiated non-carcinogenic cells of head and neck epithelia, which express A-Raf at the plasma membrane. The constitutive or induced re-localization of A-Raf to the plasma membrane compromises its ability to efficiently sequester and inactivate MST2, thus rendering cells susceptible to apoptosis. Physiologically, A-Raf re-localizes to the plasma membrane upon epithelial differentiation in vivo. This re-distribution is regulated by the scaffold protein kinase suppressor of Ras 2 (KSR2). Downregulation of KSR2 during mammary epithelial cell differentiation or siRNA-mediated knockdown re-localizes A-Raf to the plasma membrane causing the release of MST2. By using the MCF7 cell differentiation system, we could demonstrate that overexpression of A-Raf in MCF7 cells, which induces differentiation. Our findings offer a new paradigm to understand how differential localization of Raf complexes affects diverse signaling functions in normal cells and carcinomas.


Assuntos
Apoptose , Diferenciação Celular , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Caspase 8/metabolismo , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuregulina-1/farmacologia , Proteínas Proto-Oncogênicas A-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas A-raf/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Serina-Treonina Quinase 3
14.
Crit Rev Biochem Mol Biol ; 50(6): 520-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26508523

RESUMO

The Ras-Raf-MEK-MAPK (mitogen-activated protein kinase)-signaling pathway plays a key role in the regulation of many cellular functions, including cell proliferation, differentiation and transformation, by transmitting signals from membrane receptors to various cytoplasmic and nuclear targets. One of the key components of this pathway is the serine/threonine protein kinase, Raf. The Raf family kinases (A-Raf, B-Raf and C-Raf) have been intensively studied since being identified in the early 1980s as retroviral oncogenes, especially with respect to the discovery of activating mutations of B-Raf in a large number of tumors which led to intensified efforts to develop drugs targeting Raf kinases. This also resulted in a rapid increase in our knowledge of the biological functions of the B-Raf and C-Raf isoforms, which may in turn be contrasted with the little that is known about A-Raf. The biological functions of A-Raf remain mysterious, although it appears to share some of the basic properties of the other two isoforms. Recently, emerging evidence has begun to reveal the functions of A-Raf, of which some are kinase-independent. These include the inhibition of apoptosis by binding to MST2, acting as safeguard against oncogenic transformation by suppressing extracellular signal-regulated kinases (ERK) activation and playing a role in resistance to Raf inhibitors. In this review, we discuss the regulation of A-Raf protein expression, and the roles of A-Raf in apoptosis and cancer, with a special focus on its role in resistance to Raf inhibitors. We also describe the scaffold functions of A-Raf and summarize the unexpected complexity of Raf signaling.


Assuntos
Neoplasias/genética , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas A-raf/metabolismo , Animais , Resistencia a Medicamentos Antineoplásicos , Endocitose , Regulação Neoplásica da Expressão Gênica , Humanos , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Mapas de Interação de Proteínas , Isoformas de Proteínas/análise , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas A-raf/análise , Proteínas Proto-Oncogênicas A-raf/antagonistas & inibidores , Transdução de Sinais
15.
Mol Biol Cell ; 26(20): 3615-27, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26310448

RESUMO

We examine the dynamics and function of the apical scaffolding protein E3KARP/NHERF2, which consists of two PDZ domains and a tail containing an ezrin-binding domain. The exchange rate of E3KARP is greatly enhanced during mitosis due to phosphorylation at Ser-303 in its tail region. Whereas E3KARP can substitute for the function of the closely related scaffolding protein EBP50/NHERF1 in the formation of interphase microvilli, E3KARP S303D cannot. Moreover, the S303D mutation enhances the in vivo dynamics of the E3KARP tail alone, whereas in vitro the interaction of E3KARP with active ezrin is unaffected by S303D, implicating another factor regulating dynamics in vivo. A-Raf is found to be required for S303 phosphorylation in mitotic cells. Regulation of the dynamics of EBP50 is known to be dependent on its tail region but modulated by PDZ domain occupancy, which is not the case for E3KARP. Of interest, in both cases, the mechanisms regulating dynamics involve the tails, which are the most diverged region of the paralogues and probably evolved independently after a gene duplication event that occurred early in vertebrate evolution.


Assuntos
Ciclo Celular/fisiologia , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sítios de Ligação , Células CACO-2 , Técnicas de Cultura de Células , Ciclo Celular/genética , Proteínas do Citoesqueleto/metabolismo , Humanos , Espectrometria de Massas , Mitose/fisiologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Domínios PDZ/genética , Fosfoproteínas/genética , Fosforilação , Filogenia , Ligação Proteica , Proteínas Proto-Oncogênicas A-raf/metabolismo , Trocadores de Sódio-Hidrogênio/genética
16.
PLoS One ; 10(5): e0127888, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996975

RESUMO

Myofibroblasts play critical roles in the development of idiopathic pulmonary fibrosis by depositing components of extracellular matrix. One source of lung myofibroblasts is thought to be alveolar epithelial type 2 cells that undergo epithelial-mesenchymal transition (EMT). Rat RLE-6TN alveolar epithelial type 2 cells treated with transforming growth factor-ß1 (TGF-ß1) are converted into myofibroblasts through EMT. TGF-ß induces both canonical Smad signaling and non-canonical signaling, including the Ras-induced ERK pathway (Raf-MEK-ERK). However, the signaling mechanisms regulating TGF-ß1-induced EMT are not fully understood. Here, we show that the Ras-ERK pathway negatively regulates TGF-ß1-induced EMT in RLE-6TN cells and that DA-Raf1 (DA-Raf), a splicing isoform of A-Raf and a dominant-negative antagonist of the Ras-ERK pathway, plays an essential role in EMT. Stimulation of the cells with fibroblast growth factor 2 (FGF2), which activated the ERK pathway, prominently suppressed TGF-ß1-induced EMT. An inhibitor of MEK, but not an inhibitor of phosphatidylinositol 3-kinase, rescued the TGF-ß1-treated cells from the suppression of EMT by FGF2. Overexpression of a constitutively active mutant of a component of the Ras-ERK pathway, i.e., H-Ras, B-Raf, or MEK1, interfered with EMT. Knockdown of DA-Raf expression with siRNAs facilitated the activity of MEK and ERK, which were only weakly and transiently activated by TGF-ß1. Although DA-Raf knockdown abrogated TGF-ß1-induced EMT, the abrogation of EMT was reversed by the addition of the MEK inhibitor. Furthermore, DA-Raf knockdown impaired the TGF-ß1-induced nuclear translocation of Smad2, which mediates the transcription required for EMT. These results imply that intrinsic DA-Raf exerts essential functions for EMT by antagonizing the TGF-ß1-induced Ras-ERK pathway in RLE-6TN cells.


Assuntos
Células Epiteliais Alveolares/metabolismo , Transição Epitelial-Mesenquimal , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Proteínas ras/metabolismo , Células Epiteliais Alveolares/efeitos dos fármacos , Animais , Linhagem Celular , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
17.
Sci Signal ; 7(337): ra73, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25097033

RESUMO

The RAF family of kinases mediates RAS signaling, and RAF inhibitors can be effective for treating tumors with BRAF(V600E) mutant protein. However, RAF inhibitors paradoxically accelerate metastasis in RAS-mutant tumors and become ineffective in BRAF(V600E) tumors because of reactivation of downstream mitogen-activated protein kinase (MAPK) signaling. We found that the RAF isoform ARAF has an obligatory role in promoting MAPK activity and cell migration in a cell type-dependent manner. Knocking down ARAF prevented the activation of MAPK kinase 1 (MEK1) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and decreased the number of protrusions from tumor cell spheroids in three-dimensional culture that were induced by BRAF(V600E)-specific or BRAF/CRAF inhibitors (GDC-0879 and sorafenib, respectively). RAF inhibitors induced the homodimerization of ARAF and the heterodimerization of BRAF with CRAF and the scaffolding protein KSR1. In a purified protein solution, recombinant proteins of the three RAF isoforms competed for binding to MEK1. In cells in culture, overexpressing mutants of ARAF that could not homodimerize impaired the interaction between ARAF and endogenous MEK1 and thus prevented the subsequent activation of MEK1 and ERK1/2. Our findings reveal a new role for ARAF in directly activating the MAPK cascade and promoting tumor cell invasion and suggest a new therapeutic target for RAS- and RAF-mediated cancers.


Assuntos
Movimento Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Modelos Moleculares , Proteínas Proto-Oncogênicas A-raf/metabolismo , Análise de Variância , Ligação Competitiva , Western Blotting , Dimerização , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Indenos/farmacologia , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Invasividade Neoplásica , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Compostos de Fenilureia/farmacologia , Proteínas Proto-Oncogênicas A-raf/química , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-raf/antagonistas & inibidores , Pirazóis/farmacologia , RNA Interferente Pequeno/genética , Sorafenibe , Imagem com Lapso de Tempo , Células Tumorais Cultivadas
18.
Proc Natl Acad Sci U S A ; 111(22): E2291-300, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24843139

RESUMO

Alveolar formation is coupled to the spatiotemporally regulated differentiation of alveolar myofibroblasts (AMYFs), which contribute to the morphological changes of interalveolar walls. Although the Ras-ERK signaling pathway is one of the key regulators for alveolar formation in developing lungs, the intrinsic molecular and cellular mechanisms underlying its role remain largely unknown. By analyzing the Ras-ERK signaling pathway during postnatal development of lungs, we have identified a critical role of DA-Raf1 (DA-Raf)-a dominant-negative antagonist for the Ras-ERK signaling pathway-in alveolar formation. DA-Raf-deficient mice displayed alveolar dysgenesis as a result of the blockade of AMYF differentiation. DA-Raf is predominantly expressed in type 2 alveolar epithelial cells (AEC2s) in developing lungs, and DA-Raf-dependent MEK1/2 inhibition in AEC2s suppresses expression of tissue inhibitor of matalloprotienase 4 (TIMP4), which prevents a subsequent proteolytic cascade matrix metalloproteinase (MMP)14-MMP2. Furthermore, MMP14-MMP2 proteolytic cascade regulates AMYF differentiation and alveolar formation. Therefore, DA-Raf-dependent inhibition of the Ras-ERK signaling pathway in AEC2s is required for alveolar formation via triggering MMP2 activation followed by AMYF differentiation. These findings reveal a pivotal role of the Ras-ERK signaling pathway in the dynamic regulation of alveolar development.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas A-raf/metabolismo , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Animais , Diferenciação Celular/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Inibidor Tecidual 4 de Metaloproteinase
19.
Nature ; 510(7504): 283-7, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24847881

RESUMO

Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.


Assuntos
Transformação Celular Neoplásica/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Lisina/metabolismo , MAP Quinase Quinase Quinase 2/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , MAP Quinase Quinase Quinase 2/química , MAP Quinase Quinase Quinases/química , Metilação , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Oncogênica p21(ras)/genética , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Transdução de Sinais
20.
Arch Pharm Res ; 37(9): 1177-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24497038

RESUMO

It was reported that mast cell degranulation is inversely related to the enzymatic activity of M2-type pyruvate kinase (M2PK). This study shows that activation of high-affinity IgE receptor (FcεRI) evokes a sequential dual regulation of M2PK, i.e., an immediate decrement followed by slow phase increment of enzymatic activities. Changes in the activities of M2PK and mast cell degranulation showed similar time course after antigenic stimulation of FcεRI. The immediate inhibition of M2PK involved tyrosine phosphorylation, and subsequently led to a cellular accumulation of glycolytic intermediates, including fructose 1,6-biphosphate (FBP), a feedforward activator of M2PK. As the cellular levels of FBP were increased, both the enzymatic acitivity of M2PK and mast cell degranulation slowly returned to near basal levels. A-Raf, when exogenously introduced into RBL-2H3 cells, phosphorylated M2PK on the serine residues, elevated enzyme activities of M2PK, and resulted in the inhibition of degranulation. These results suggest that dual regulation of M2PK which involves the phosphorylation of M2PK and accumulation of a feedforward activator of M2PK plays important roles in the control of mast cell degranulation.


Assuntos
Degranulação Celular , Imunoglobulina E/metabolismo , Mastócitos/fisiologia , Piruvato Quinase/metabolismo , Receptores de IgE/metabolismo , Transdução de Sinais , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Fosfo-Específicos , Linhagem Celular Tumoral , Ativação Enzimática , Frutosedifosfatos/metabolismo , Células HEK293 , Humanos , Mastócitos/enzimologia , Mastócitos/imunologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas A-raf/genética , Proteínas Proto-Oncogênicas A-raf/metabolismo , Piruvato Quinase/antagonistas & inibidores , Piruvato Quinase/química , Piruvato Quinase/genética , Ratos , Receptores de IgE/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA