Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Kaohsiung J Med Sci ; 33(11): 543-549, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29050671

RESUMO

Gambogic acid (GA) has been shown to inhibit cancer cell proliferation, induce apoptosis, and enhance reactive oxygen species accumulation. However, whether GA could improve multidrug resistance through modulating autophagy has never been explored. We demonstrated that the combination of GA and cisplatin (CDDP) resulted in a stronger growth inhibition effect on A549 and NCI-H460 cells using the MTT assay. Furthermore, treatment with GA significantly increased autophagy in these cells. More importantly, GA-induced cell death could be largely abolished by 3-methyladenine (3-MA) or chloroquine (CQ) treatment, suggesting that GA-induced cell death was dependent on autophagy. Western blot analysis showed that GA treatment suppressed the activation of Akt, mTOR, and S6. In addition, using a GA and rapamycin combination induced more cell death compared to either GA or rapamycin alone. In summary, GA may have utility as an adjunct therapy for non-small cell lung cancer (NSCLC) patients through autophagy-dependent cell death, even when cancer cells have developed resistance to apoptosis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Autofagia/efeitos dos fármacos , Garcinia/química , Regulação Neoplásica da Expressão Gênica , Xantonas/farmacologia , Células A549 , Adenina/análogos & derivados , Adenina/farmacologia , Antineoplásicos Fitogênicos/isolamento & purificação , Autofagia/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cloroquina/farmacologia , Cisplatino/farmacologia , Combinação de Medicamentos , Sinergismo Farmacológico , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Extratos Vegetais/química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/genética , Proteína S6 Ribossômica/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Xantonas/isolamento & purificação
2.
Toxicol Lett ; 233(2): 102-13, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25597859

RESUMO

Postnatal isoflurane exposure leads to neurodegeneration and deficits of spatial learning and memory in the adulthood. However, the underlying mechanisms remain unclear. Ribosomal protein S6 is demonstrated to play a pivotal role in control of cell survival, protein synthesis and synaptogenesis for brain development. In this study, the possible role of S6 and its upstream signaling pathways in the developmental neurotoxicity of isoflurane was evaluated using models of primary cultured hippocampal neurons and postnatal day 7 rats. We found that isoflurane decreased IGF-1 level and suppressed activation of IGF-1 receptor, sequentially inhibiting S6 activity via IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways. S6 inhibition enhanced isoflurane-induced decreased Bcl-xL and increased cleaved caspase-3 and Bad, also reduced PSD95 expression and aggravated deficits of spatial learning and memory. S6 activation could reverse the damages above. These results indicate that S6 inhibition, led by suppression of upstream IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways, is involved in the neuroapoptosis, synaptogenesis impairment and spatial learning and memory decline caused by postnatal isoflurane exposure. S6 activation may exhibit protective potential against developmental neurotoxicity of isoflurane.


Assuntos
Anestésicos Inalatórios/toxicidade , Encéfalo/crescimento & desenvolvimento , Isoflurano/toxicidade , Síndromes Neurotóxicas/patologia , Proteína S6 Ribossômica/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Deficiências da Aprendizagem/induzido quimicamente , Deficiências da Aprendizagem/psicologia , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/psicologia , Atividade Motora/efeitos dos fármacos , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley
3.
Eur J Pharm Sci ; 63: 87-95, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25008115

RESUMO

The PI3K/Akt/mTOR/S6 ribosomal protein signalling pathway is a key potential target in breast cancer therapy, playing a central role in proliferation and cell survival. In this study, we found that the seleno-compound 2,4-dihydroselenoquinazoline (3a) generally inhibited this signalling axis in MCF-7 breast cancer cells and caused downregulation of S6 ribosomal protein phosphorylation in a dose- and time-dependent manner. Furthermore, 3a caused a dose- and time-dependent decrease in MCF-7 cell viability as well as cell cycle arrest in G2/M. Interestingly 3a also induced apoptosis, as evidenced by cleavage of PARP and caspase-7, and inhibited autophagy, as demonstrated by accumulation of LC3-II and p62/SQSTM1. Given that induction of autophagy has been previously described as a mechanism by which some breast cancer cells counteract proapoptotic signalling and develop resistance to anti-hormone therapy, this suggests that this derivative, which both triggers apoptosis and inhibits autophagy, may be beneficial in preventing and overcoming resistance in breast cancer cells. The data also show the complexity of this signalling axis which is far from being understood.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Quinazolinas/farmacologia , Proteína S6 Ribossômica/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Células MCF-7 , Estrutura Molecular , Compostos Organosselênicos/química , Quinazolinas/química , Proteína S6 Ribossômica/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
4.
Exp Cell Res ; 326(2): 201-9, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24768699

RESUMO

Everolimus is an orally administered mTOR inhibitor. The effect, and mechanism of action, of everolimus on lung cancers with an epidermal growth factor receptor (EGFR) mutation remain unclear. Four gefitinib-sensitive and -resistant cell lines were used in the present work. Growth inhibition was determined using the MTT assay. Transgenic mice carrying the EGFR L858R mutation were treated with everolimus (10 mg/kg/day), or vehicle alone, from 5 to 20 weeks of age, and were then sacrificed. To evaluate the efficacy of everolimus in prolonging survival, everolimus (10 mg/kg/day) or vehicle was administered from 5 weeks of age. The four cell lines were similarly sensitive to everolimus. Expression of phosphorylated (p) mTOR and pS6 were suppressed upon treatment with everolimus in vitro, whereas the pAKT level increased. The numbers of lung tumors with a long axis exceeding 1mm in the everolimus-treated and control groups were 1.9 ± 0.9 and 9.4 ± 3.2 (t-test, p<0.001), respectively. pS6 was suppressed during eve r olimus treatment. Although apoptosis and autophagy were not induced in everolimus-treated EGFR transgenic mice, angiogenesis was suppressed. The median survival time in the everolimus-treated group (58.0 weeks) was significantly longer than that in the control group (31.2 weeks) (logrank test, p<0.001). These findings suggest that everolimus had an indirect effect on tumor formation by inhibiting angiogenesis and might be effective to treat lung tumors induced by an activating EGFR gene mutation.


Assuntos
Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Mutação , Sirolimo/análogos & derivados , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Everolimo , Gefitinibe , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Fosforilação , Quinazolinas/farmacologia , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo
5.
Am J Respir Cell Mol Biol ; 45(5): 1028-35, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21622293

RESUMO

Inhibiting hypoxia-inducible factor (HIF)-1α activity has been proposed as a novel therapeutic target in LPS-induced sepsis syndrome. We have reported that tanshinone IIA (TIIA) can reduce LPS-induced lethality and lung injury in mice, but the precise mechanisms have not been fully described. Therefore, the present study investigated whether the protective effect of TIIA was related to the inhibition of LPS-induced HIF-1α expression and what mechanisms accounted for it. This study showed that TIIA pretreatment improved LPS-induced biochemical and cellular changes and reduced the production of inflammatory cytokines. Pretreatment with TIIA decreased LPS-induced HIF-1α expression in vivo and in vitro. TIIA did not affect the LPS-induced HIF-1α mRNA level but inhibited HIF-1α protein translation by the inhibition of the PI3K/AKT and MAPK pathways and related protein translational regulators, such as p70S6K1, S6 ribosomal protein, 4E-BP1, and eIF4E, and promoted HIF-1α protein degradation via the proteasomal pathway in LPS-stimulated macrophages. These observations partially explain the antiinflammatory effects of TIIA, which provides scientific basis for its application for the treatment of acute lung injury/acute respiratory distress syndrome or sepsis.


Assuntos
Abietanos/farmacologia , Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Endotoxemia/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Endotoxemia/induzido quimicamente , Fatores de Iniciação em Eucariotos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosfoproteínas/antagonistas & inibidores , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteína S6 Ribossômica/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores
6.
Amino Acids ; 39(5): 1487-92, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20464435

RESUMO

The ribosomal protein S6 is essential for the formation of the subunits of higher eukaryotic ribosomes, and S6 heterozygosity leads to early embryonal lethality in mice. S6 is phosphorylated at clustered residues S235/236 and S240/244 upon numerous physiological and pathological stimuli. So far, the S6Kinases, S6K1 and S6K2 are the only proven S6 S240/244 phosphorylating enzymes in mammalian cells. The activity of these S6Kinases is strictly regulated via the mammalian target of rapamycin (mTOR) enzyme complex with raptor, named mTORC1. In time course experiments with the mTORC1 inhibitor rapamycin we here demonstrate rapamycin-resistant phosphorylation of the ribosomal protein S6 at S240/244. Serum-restimulation experiments further demonstrated that this rapamycin-resistant S6 240/244 phosphorylation is induced via serum factors in a cell cycle-dependent manner. Our data allow new insights into the regulation of S6 phosphorylation and provide evidence for the existence of rapamycin-resistant S6 phosphorylating kinase activities.


Assuntos
Proteína S6 Ribossômica/antagonistas & inibidores , Sirolimo/farmacologia , Ciclo Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Fosforilação , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Proteína S6 Ribossômica/metabolismo , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR
7.
Cancer Biol Ther ; 8(20): 1902-3, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19783905
8.
Blood ; 113(26): 6669-80, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19401561

RESUMO

Detailed genomic studies have shown that cytogenetic abnormalities contribute to multiple myeloma (MM) pathogenesis and disease progression. Nevertheless, little is known about the characteristics of MM at the epigenetic level and specifically how microRNAs regulate MM progression in the context of the bone marrow milieu. Therefore, we performed microRNA expression profiling of bone marrow derived CD138(+) MM cells versus their normal cellular counterparts and validated data by qRT-PCR. We identified a MM-specific microRNA signature characterized by down-expression of microRNA-15a/-16 and overexpression of microRNA-222/-221/-382/-181a/-181b (P < .01). We investigated the functional role of microRNA-15a and -16 and showed that they regulate proliferation and growth of MM cells in vitro and in vivo by inhibiting AKT serine/threonine-protein-kinase (AKT3), ribosomal-protein-S6, MAP-kinases, and NF-kappaB-activator MAP3KIP3. Moreover, miRNA-15a and -16 exerted their anti-MM activity even in the context of the bone marrow milieu in vitro and in vivo. These data indicate that microRNAs play a pivotal role in the biology of MM and represent important targets for novel therapies in MM.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/fisiologia , Mieloma Múltiplo/patologia , RNA Neoplásico/fisiologia , Inibidores da Angiogênese/fisiologia , Animais , Adesão Celular , Divisão Celular/fisiologia , Ensaios Clínicos Fase II como Assunto/estatística & dados numéricos , Técnicas de Cocultura , Células Endoteliais/citologia , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos SCID , MicroRNAs/biossíntese , MicroRNAs/genética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/metabolismo , Células Estromais/citologia
9.
J Biol Chem ; 282(19): 14056-64, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17360704

RESUMO

Converging signals from the mammalian target of rapamycin (mTOR) and phosphoinositide 3-kinase (PI3K) pathways are well established to modulate translation initiation. Less is known regarding the molecular basis of protein synthesis regulated by other inputs, such as agonists of the Ras/extracellular signal-regulated kinase (ERK) signaling cascade. Ribosomal protein (rp) S6 is a component of the 40S ribosomal subunit that becomes phosphorylated at several serine residues upon mitogen stimulation, but the exact molecular mechanisms regulating its phosphorylation and the function of phosphorylated rpS6 is poorly understood. Here, we provide evidence that activation of the p90 ribosomal S6 kinases (RSKs) by serum, growth factors, tumor promoting phorbol esters, and oncogenic Ras is required for rpS6 phosphorylation downstream of the Ras/ERK signaling cascade. We demonstrate that while ribosomal S6 kinase 1 (S6K1) phosphorylates rpS6 at all sites, RSK exclusively phosphorylates rpS6 at Ser(235/236) in vitro and in vivo using an mTOR-independent mechanism. Mutation of rpS6 at Ser(235/236) reveals that phosphorylation of these sites promotes its recruitment to the 7-methylguanosine cap complex, suggesting that Ras/ERK signaling regulates assembly of the translation preinitiation complex. These data demonstrate that RSK provides an mTOR-independent pathway linking the Ras/ERK signaling cascade to the translational machinery.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína S6 Ribossômica/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Proteínas ras/metabolismo , Células Cultivadas , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Rim/metabolismo , Luciferases/metabolismo , MAP Quinase Quinase Quinases , Mutação , Fosforilação , Polirribossomos/metabolismo , Biossíntese de Proteínas , RNA Interferente Pequeno/farmacologia , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Ribossomos/metabolismo , Transdução de Sinais , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
10.
Cancer Res ; 65(16): 7462-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16103100

RESUMO

Activation of protein kinase Cbeta (PKCbeta) has been repeatedly implicated in tumor-induced angiogenesis. The PKCbeta-selective inhibitor, Enzastaurin (LY317615.HCl), suppresses angiogenesis and was advanced for clinical development based upon this antiangiogenic activity. Activation of PKCbeta has now also been implicated in tumor cell proliferation, apoptosis, and tumor invasiveness. Herein, we show that Enzastaurin has a direct effect on human tumor cells, inducing apoptosis and suppressing the proliferation of cultured tumor cells. Enzastaurin treatment also suppresses the phosphorylation of GSK3betaser9, ribosomal protein S6(S240/244), and AKT(Thr308). Oral dosing with Enzastaurin to yield plasma concentrations similar to those achieved in clinical trials significantly suppresses the growth of human glioblastoma and colon carcinoma xenografts. As in cultured tumor cells, Enzastaurin treatment suppresses the phosphorylation of GSK3beta in these xenograft tumor tissues. Enzastaurin treatment also suppresses GSK3beta phosphorylation to a similar extent in peripheral blood mononuclear cells (PBMCs) from these treated mice. These data show that Enzastaurin has a direct antitumor effect and that Enzastaurin treatment suppresses GSK3beta phosphorylation in both tumor tissue and in PBMCs, suggesting that GSK3beta phosphorylation may serve as a reliable pharmacodynamic marker for Enzastaurin activity. With previously published reports, these data support the notion that Enzastaurin suppresses tumor growth through multiple mechanisms: direct suppression of tumor cell proliferation and the induction of tumor cell death coupled to the indirect effect of suppressing tumor-induced angiogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Indóis/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Feminino , Glioblastoma/enzimologia , Glioblastoma/patologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteína Quinase C beta , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Biosci Biotechnol Biochem ; 66(11): 2437-43, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12506984

RESUMO

An expression library of Arabidopsis thaliana cDNAs was randomly introduced into A. thaliana. The transformant pool was used to obtain a line, c105, with reduced apical dominance and irregular positioning of leaves and flowers. The inserted DNA was a 3'-fragment of the ribosomal protein S6 gene with antisense orientation. The transcriptional level of the ribosomal protein S6 was lower in c105 than in the wild-type plant. Introduction of the same fragment into the wild-type plant gave phenotypes similar to those of c105, so the phenotypes of c105 were due to the S6 antisense expression. The phenotypes suggest selectively reduced function of specific proteins rather than an overall decrease in protein function caused by defective ribosomal biogenesis.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/crescimento & desenvolvimento , Proteína S6 Ribossômica/antagonistas & inibidores , Arabidopsis/genética , Northern Blotting , Sondas de DNA/genética , DNA Complementar/metabolismo , DNA de Plantas/genética , DNA de Plantas/metabolismo , Flores/genética , Flores/crescimento & desenvolvimento , Regulação da Expressão Gênica de Plantas , Mutação , Fenótipo , Folhas de Planta/genética , Folhas de Planta/crescimento & desenvolvimento , Plantas Geneticamente Modificadas/metabolismo , RNA Antissenso/biossíntese , RNA Antissenso/genética , RNA Ribossômico/biossíntese , RNA Ribossômico/genética , Proteína S6 Ribossômica/genética , Proteína S6 Ribossômica/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...