Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Cell Signal ; 34: 55-65, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28302565

RESUMO

Protein arginine methyltransferase 5 (PRMT5) is an arginine methylation methyltransferase that regulates various physiological processes. Abnormal PRMT5 activity has been reported in inflammation and various types of cancers. Because osteoclast differentiation is characterized by the activation of inflammation-related pathways, we speculated that PRMT5 may play a role in this process. In the present study, we found that PRMT5 was upregulated during osteoclast differentiation. Knockdown of PRMT5 with siRNA in bone marrow mononuclear cells (BMMs) resulted in inhibition of receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation. Consistent with the PRMT5 knockdown results, the PRMT5 inhibitor EPZ015666 (EPZ) suppressed osteoclast differentiation and bone resorption. Intraperitoneal administration of EPZ prevented ovariectomy-induced bone loss. Moreover, RANKL-induced NF-κB and MAPK activation was inhibited by EPZ. Expression microarrays showed that the expression of several osteoclast formation-related genes was altered by EPZ treatment, including chemokine C-X-C motif ligand 10 (CXCL10). Administration of recombinant CXCL10 partially reversed the osteoclastogenesis inhibition effect of the PRMT5 inhibitor. Intriguingly, RSAD2, which is a reported antiviral protein, was apparently suppressed when PRMT5 was inhibited. Knockdown of RSAD2 with siRNA in BMMs led to inhibition of osteoclast differentiation. Subsequent ChIP-qPCR identified that both PRMT5 inhibition and knockdown resulted in decreased H3R8 or/and H4R3 methylation at CXCL10 and RSAD2 promotors. In conclusion, our study found that PRMT5 is an activator of osteoclast differentiation and inhibition of PRMT5 partially suppressed osteoclastogenesis through downregulation of CXCL10 and RSAD2.


Assuntos
Quimiocina CXCL10/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas/metabolismo , Animais , Células da Medula Óssea/citologia , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL10/genética , Regulação para Baixo/efeitos dos fármacos , Feminino , Isoquinolinas/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteoprotegerina/sangue , Ovariectomia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/genética , Proteínas/genética , Pirimidinas/farmacologia , Ligante RANK/sangue , Ligante RANK/genética , Ligante RANK/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
2.
Nat Med ; 22(11): 1314-1320, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27748748

RESUMO

Pediatric glioblastoma is one of the most common and most deadly brain tumors in childhood. Using an integrative genetic analysis of 53 pediatric glioblastomas and five in vitro model systems, we identified previously unidentified gene fusions involving the MET oncogene in ∼10% of cases. These MET fusions activated mitogen-activated protein kinase (MAPK) signaling and, in cooperation with lesions compromising cell cycle regulation, induced aggressive glial tumors in vivo. MET inhibitors suppressed MET tumor growth in xenograft models. Finally, we treated a pediatric patient bearing a MET-fusion-expressing glioblastoma with the targeted inhibitor crizotinib. This therapy led to substantial tumor shrinkage and associated relief of symptoms, but new treatment-resistant lesions appeared, indicating that combination therapies are likely necessary to achieve a durable clinical response.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-met/genética , RNA Mensageiro/metabolismo , Adolescente , Adulto , Anilidas/farmacologia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Criança , Pré-Escolar , Crizotinibe , DNA de Neoplasias , Feminino , Glioblastoma/tratamento farmacológico , Humanos , Lactente , Masculino , Camundongos , Camundongos SCID , Proteínas Associadas aos Microtúbulos/genética , Proteínas Quinases Ativadas por Mitógeno , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas/genética , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Pirazóis/uso terapêutico , Piridinas/uso terapêutico , Quinolinas/farmacologia , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/genética , Análise de Sequência de DNA , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
3.
J Mol Biol ; 427(11): 2121-34, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25861760

RESUMO

Ubiquitination is one of the most prevalent posttranslational modifications in eukaryotic cells, with functional importance in protein degradation, subcellular localization and signal transduction pathways. Immunoaffinity enrichment coupled with quantitative mass spectrometry enables the in-depth characterization of protein ubiquitination events at the site-specific level. We have applied this strategy to investigate cellular response triggered by two distinct type agents: small molecule inhibitors of the tumor-associated kinases MEK and PI3K or the pro-inflammatory cytokine IL-17. Temporal profiling of protein ubiquitination events across a series of time points covering the biological response permits interrogation of signaling through thousands of quantified proteins, of which only a subset display significant and physiologically meaningful regulation. Distinctive clusters of residues within proteins can display distinct temporal patterns attributable to diverse molecular functions, although the majority of differential ubiquitination appears as a coordinated response across the modifiable residues present within an individual substrate. In cells treated with a combination of MEK and PI3K inhibitors, we found differential ubiquitination of MEK within the first hour after treatment and a series of mitochondria proteins at later time points. In the IL-17 signaling pathway, ubiquitination events on several signaling proteins including HOIL-1 and Tollip were observed. The functional relevance of these putative IL-17 mediators was subsequently validated by knockdown of HOIL-1, HOIP and TOLIP, each of which decreased IL-17-stimulated cytokine production. Together, these data validate proteomic profiling of protein ubiquitination as a viable approach for identifying dynamic signaling components in response to intracellular and extracellular perturbations.


Assuntos
Proteínas Mitocondriais/metabolismo , Proteínas/imunologia , Proteômica/métodos , Espectrometria de Massas em Tandem/métodos , Ubiquitina/metabolismo , Apoptose/efeitos dos fármacos , Azetidinas/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Indazóis/farmacologia , Interleucina-17/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Piperidinas/farmacologia , Proteínas/metabolismo , Transdução de Sinais , Sulfonamidas/farmacologia , Fatores de Transcrição , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
PLoS One ; 8(8): e72377, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24013388

RESUMO

To understand the effects of the interaction between Mycoplasma and cells on the host cellular function, it is important to elucidate the influences of infection of cells with Mycoplasma on nuclear enzymes such as DNA Topoisomerase type I (Topo I). Human Topo I participates in DNA transaction processes and is the target of anti-cancer drugs, the camptothecins (CPTs). Here we investigated the mechanism by which infection of human tumor cells with Mycoplasma fermentans affects the activity and expression of cellular Topo I, and the anti-cancer efficacy of CPT. Human cancer cells were infected or treated with live or sonicated M. fermentans and the activity and expression of Topo I was determined. M. fermentans significantly reduced (by 80%) Topo I activity in the infected/treated tumor cells without affecting the level of Topo I protein. We demonstrate that this reduction in enzyme activity resulted from ADP-ribosylation of the Topo I protein by Poly-ADP-ribose polymerase (PARP-1). In addition, pERK was activated as a result of the induction of the MAPK signal transduction pathway by M. fermentans. Since PARP-1 was shown to be activated by pERK, we concluded that M. fermentans modified the cellular Topo I activity by activation of PARP-I via the induction of the MAPK signal transduction pathway. Moreover, the infection of tumor cells with M. fermentans diminished the inhibitory effect of CPT. The results of this study suggest that modification of Topo I activity by M. fermentans may alter cellular gene expression and the response of tumor cells to Topo I inhibitors, influencing the anti-cancer capacity of Topo I antagonists.


Assuntos
Camptotecina/farmacologia , DNA Topoisomerases Tipo I/metabolismo , Infecções por Mycoplasma/enzimologia , Mycoplasma fermentans/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores da Topoisomerase I/farmacologia , Proteínas de Bactérias/fisiologia , Benzamidas/farmacologia , DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Interações Hospedeiro-Patógeno , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Infecções por Mycoplasma/microbiologia , Poli(ADP-Ribose) Polimerase-1 , Poli Adenosina Difosfato Ribose/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Inibidores de Proteínas Quinases/farmacologia , Proteínas/metabolismo
5.
Am J Physiol Endocrinol Metab ; 304(10): E1042-52, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23531613

RESUMO

Although catabolic signaling has a well-established role in muscle wasting during cancer cachexia, the suppression of anabolic signaling also warrants further investigation. In cachectic tumor-bearing mice, circulating IL-6 levels are associated with suppressed muscle protein synthesis and mTORC1 signaling. We have found AMPK and IGF-I/insulin signaling, two well-known regulators of the mammalian target of rapamycin (mTOR), are altered with the progression of cachexia. How IL-6 can induce suppression of mTORC1 signaling remains to be established. The purpose of this study was to examine mTOR complex 1 (mTORC1) activation and regulation by IL-6 during cancer cachexia. IL-6 effects on mTOR activation were examined in Apc(Min/+) mouse skeletal muscle and C2C12 myotubes. Systemic IL-6 overexpression in Apc(Min/+) mice produced a dose-dependent suppression of mTOR signaling that corresponded to induction of STAT3 and AMPK phosphorylation. This result was also evident in IL-6-treated myotubes. Basal mTOR activation and mTOR responsiveness to glucose administration were suppressed in cachectic skeletal muscle. However, insulin induction of mTOR activity was maintained in IL-6-treated myotubes. Whereas IL-6 suppression of myotube mTOR activity was rescued by AMPK inhibition, inhibition of STAT3 signaling was not sufficient to rescue IL-6 suppression of mTOR activity. Last, treadmill exercise training was able to prevent IL-6-induced inhibition of mTOR signaling in Apc(Min/+) mice independently of activated STAT. In conclusion, we report dose-dependent suppression of mTOR activity by IL-6 and suppressed mTOR responsiveness to glucose administration in Apc(Min/+) mice. IL-6 suppression of mTOR activity was dependent on AMPK activation and independent of STAT signaling in myotubes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Caquexia/metabolismo , Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Caquexia/enzimologia , Interleucina-6/sangue , Interleucina-6/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/enzimologia , Neoplasias Experimentais/enzimologia , Fosforilação , Condicionamento Físico Animal/fisiologia , Proteínas/antagonistas & inibidores , Proteínas/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
6.
J Biol Chem ; 288(10): 7086-95, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23355489

RESUMO

Ataxia telangiectasia mutated (ATM) is activated upon DNA double strand breaks (DSBs) and phosphorylates numerous DSB response proteins, including histone H2AX on serine 139 (Ser-139) to form γ-H2AX. Through interaction with MDC1, γ-H2AX promotes DSB repair by homologous recombination (HR). H2AX Ser-139 can also be phosphorylated by DNA-dependent protein kinase catalytic subunit and ataxia telangiectasia- and Rad3-related kinase. Thus, we tested whether ATM functions in HR, particularly that controlled by γ-H2AX, by comparing HR occurring at the euchromatic ROSA26 locus between mouse embryonic stem cells lacking either ATM, H2AX, or both. We show here that loss of ATM does not impair HR, including H2AX-dependent HR, but confers sensitivity to inhibition of poly(ADP-ribose) polymerases. Loss of ATM or H2AX has independent contributions to cellular sensitivity to ionizing radiation. The ATM-independent HR function of H2AX requires both Ser-139 phosphorylation and γ-H2AX/MDC1 interaction. Our data suggest that ATM is dispensable for HR, including that controlled by H2AX, in the context of euchromatin, excluding the implication of such an HR function in genomic instability, hypersensitivity to DNA damage, and poly(ADP-ribose) polymerase inhibition associated with ATM deficiency.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Recombinação Homóloga , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Western Blotting , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cromonas/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos da radiação , Endodesoxirribonucleases/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Morfolinas/farmacologia , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Radiação Ionizante , Serina/metabolismo , Proteínas Supressoras de Tumor/genética
7.
Biochem Biophys Res Commun ; 430(1): 352-7, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23159620

RESUMO

Metformin, a widely used anti-diabetic drug, is emerging as a potential anticancer agent but the mechanisms involved remain incompletely understood. Here, we demonstrate that the potency of metformin induced AMPK activation, as shown by the phosphorylation of its substrates acetyl-CoA carboxylase (ACC) at Ser(79) and Raptor at Ser(792), was dramatically enhanced in human pancreatic ductal adenocarcinoma (PDAC) cells PANC-1 and MiaPaCa-2 cultured in medium containing physiological concentrations of glucose (5 mM), as compared with parallel cultures in medium with glucose at 25 mM. In physiological glucose, metformin inhibited mTORC1 activation, DNA synthesis and proliferation of PDAC cells stimulated by crosstalk between G protein-coupled receptors and insulin/IGF signaling systems, at concentrations (0.05-0.1 mM) that were 10-100-fold lower than those used in most previous reports. Using siRNA-mediated knockdown of the α(1) and α(2) catalytic subunits of AMPK, we demonstrated that metformin, at low concentrations, inhibited DNA synthesis through an AMPK-dependent mechanism. Our results emphasize the importance of using medium containing physiological concentrations of glucose to elucidate the anticancer mechanism of action of metformin in pancreatic cancer cells and other cancer cell types.


Assuntos
Proteínas Quinases Ativadas por AMP/biossíntese , Antineoplásicos/farmacologia , Replicação do DNA/efeitos dos fármacos , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Neoplasias Pancreáticas/metabolismo , Proteínas/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática , Técnicas de Silenciamento de Genes , Glucose/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Serina-Treonina Quinases TOR
8.
J Biol Chem ; 287(13): 9742-9752, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22223645

RESUMO

An intensive recent effort to develop ATP-competitive mTOR inhibitors has resulted in several potent and selective molecules such as Torin1, PP242, KU63794, and WYE354. These inhibitors are being widely used as pharmacological probes of mTOR-dependent biology. To determine the potency and specificity of these agents, we have undertaken a systematic kinome-wide effort to profile their selectivity and potency using chemical proteomics and assays for enzymatic activity, protein binding, and disruption of cellular signaling. Enzymatic and cellular assays revealed that all four compounds are potent inhibitors of mTORC1 and mTORC2, with Torin1 exhibiting ∼20-fold greater potency for inhibition of Thr-389 phosphorylation on S6 kinases (EC(50) = 2 nM) relative to other inhibitors. In vitro biochemical profiling at 10 µM revealed binding of PP242 to numerous kinases, although WYE354 and KU63794 bound only to p38 kinases and PI3K isoforms and Torin1 to ataxia telangiectasia mutated, ATM and Rad3-related protein, and DNA-PK. Analysis of these protein targets in cellular assays did not reveal any off-target activities for Torin1, WYE354, and KU63794 at concentrations below 1 µM but did show that PP242 efficiently inhibited the RET receptor (EC(50), 42 nM) and JAK1/2/3 kinases (EC(50), 780 nM). In addition, Torin1 displayed unusually slow kinetics for inhibition of the mTORC1/2 complex, a property likely to contribute to the pharmacology of this inhibitor. Our results demonstrated that, with the exception of PP242, available ATP-competitive compounds are highly selective mTOR inhibitors when applied to cells at concentrations below 1 µM and that the compounds may represent a starting point for medicinal chemistry efforts aimed at developing inhibitors of other PI3K kinase-related kinases.


Assuntos
Inibidores Enzimáticos/farmacocinética , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Trifosfato de Adenosina , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Proteômica/métodos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Mol Cancer Res ; 9(12): 1696-707, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22009179

RESUMO

DNA-dependent protein kinase (DNA-PK) plays a pivotal role in the repair of DNA double-strand breaks (DSB) and is centrally involved in regulating cellular radiosensitivity. Here, we identify DNA-PK as a key therapeutic target for augmenting accelerated senescence in irradiated human cancer cells. We find that BEZ235, a novel inhibitor of DNA-PK and phosphoinositide 3-kinase (PI3K)/mTOR, abrogates radiation-induced DSB repair resulting in cellular radiosensitization and growth delay of irradiated tumor xenografts. Importantly, radiation enhancement by BEZ235 coincides with a prominent p53-dependent accelerated senescence phenotype characterized by positive ß-galactosidase staining, G(2)-M cell-cycle arrest, enlarged and flattened cellular morphology, and increased p21 expression and senescence-associated cytokine secretion. Because this senescence response to BEZ235 is accompanied by unrepaired DNA DSBs, we examined whether selective targeting of DNA-PK also induces accelerated senescence in irradiated cells. Significantly, we show that specific pharmacologic inhibition of DNA-PK, but not PI3K or mTORC1, delays DSB repair leading to accelerated senescence after radiation. We additionally show that PRKDC knockdown using siRNA promotes a striking accelerated senescence phenotype in irradiated cells comparable with that of BEZ235. Thus, in the context of radiation treatment, our data indicate that inhibition of DNA-PK is sufficient for the induction of accelerated senescence. These results validate DNA-PK as an important therapeutic target in irradiated cancer cells and establish accelerated senescence as a novel mechanism of radiosensitization induced by DNA-PK blockade.


Assuntos
Senescência Celular/genética , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Imidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Nus , Complexos Multiproteicos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , RNA Interferente Pequeno/genética , Serina-Treonina Quinases TOR , Transplante Heterólogo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
10.
Cell Signal ; 23(12): 1927-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21763421

RESUMO

cAMP and mTOR signalling pathways control a number of critical cellular processes including metabolism, protein synthesis, proliferation and cell survival and therefore understanding the signalling events which integrate these two signalling pathways is of particular interest. In this study, we show that the pharmacological elevation of [cAMP](i) in mouse embryonic fibroblasts (MEFs) and human embryonic kidney 293 (HEK293) cells inhibits mTORC1 activation via a PKA-dependent mechanism. Although the inhibitory effect of cAMP on mTOR could be mediated by impinging on signalling cascades (i.e. PKB, MAPK and AMPK) that inhibit TSC1/2, an upstream negative regulator of mTORC1, we show that cAMP inhibits mTORC1 in TSC2 knockout (TSC2(-/-)) MEFs. We also show that cAMP inhibits insulin and amino acid-stimulated mTORC1 activation independently of Rheb, Rag GTPases, TSC2, PKB, MAPK and AMPK, indicating that cAMP may act independently of known regulatory inputs into mTOR. Moreover, we show that the prolonged elevation in [cAMP](i) can also inhibit mTORC2. We provide evidence that this cAMP-dependent inhibition of mTORC1/2 is caused by the dissociation of mTORC1 and 2 and a reduction in mTOR catalytic activity, as determined by its auto-phosphorylation on Ser2481. Taken together, these results provide an important insight into how cAMP signals to mTOR and down-regulates its activity, which may lead to the identification of novel drug targets to inhibit mTOR that could be used for the treatment and prevention of human diseases such as cancer.


Assuntos
AMP Cíclico/fisiologia , Proteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Adenilato Quinase/metabolismo , Aminoácidos/farmacologia , Aminoácidos/fisiologia , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Colforsina/farmacologia , AMP Cíclico/química , AMP Cíclico/metabolismo , Fatores de Iniciação em Eucariotos , Deleção de Genes , Técnicas de Inativação de Genes , Humanos , Insulina/farmacologia , Insulina/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Proteínas Quinases/metabolismo , Proteínas/agonistas , Proteínas/antagonistas & inibidores , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Serina-Treonina Quinases TOR , Fatores de Transcrição/agonistas , Fatores de Transcrição/antagonistas & inibidores , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
11.
J Lipid Res ; 52(8): 1450-60, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21636835

RESUMO

Both insulin and the cell death-inducing DNA fragmentation factor-α-like effector (CIDE) family play important roles in apoptosis and lipid droplet formation. Previously, we reported that CIDEA and CIDEC are differentially regulated by insulin and contribute separately to insulin-induced anti-apoptosis and lipid droplet formation in human adipocytes. However, the upstream signals of CIDE proteins remain unclear. Here, we investigated the signaling molecules involved in insulin regulation of CIDEA and CIDEC expression. The phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and PI-103 blocked both insulin-induced downregulation of CIDEA and upregulation of CIDEC. The Akt inhibitor API-2 and the c-Jun N-terminal kinase (JNK) inhibitor SP600125 selectively inhibited insulin regulation of CIDEA and CIDEC expression, respectively, whereas the MAPK/ERK kinase inhibitor U0126 and the p38 inhibitor SB203580 did not. Small interfering RNA-mediated depletion of Akt1/2 prevented insulin-induced downregulation of CIDEA and inhibition of apoptosis. Depletion of JNK2, but not JNK1, inhibited insulin-induced upregulation of CIDEC and lipid droplet enlargement. Furthermore, insulin increased both Akt and JNK phosphorylation, which was abrogated by the PI3K inhibitors. These results suggest that insulin regulates CIDEA and CIDEC expression via PI3K, and it regulates expression of each protein via Akt1/2- and JNK2-dependent pathways, respectively, in human adipocytes.


Assuntos
Adipócitos/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Regulação da Expressão Gênica , Insulina , Obesidade/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Adipócitos/citologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Clorpropamida/análogos & derivados , Clorpropamida/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Regulação para Baixo , Feminino , Furanos/farmacologia , Inativação Gênica/efeitos dos fármacos , Humanos , Insulina/metabolismo , Insulina/farmacologia , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/farmacologia , Regulação para Cima
12.
Nitric Oxide ; 25(2): 161-8, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21703357

RESUMO

Nitric oxide (NO) exerts bifunctional effects on cell survival. While a high concentration of NO is cytotoxic, a relatively low concentration of NO promotes cytoprotection and cell survival. However, the molecular mechanism underlying the cytoprotective effect of NO remains poorly understood. One of the transcription factors that confer cellular protection against oxidative stress is NF-E2-related factor 2 (Nrf2), which is sequestered in the cytoplasm by forming an inactive complex with Klech-like ECH-associated protein 1 (Keap1). Previous studies suggested that various stimuli could induce the dissociation of Nrf2 from Keap1 in cytosol and/or promote its nuclear translocation by activating several upstream kinases. NO-mediated thiol modification in Keap1 has also been proposed as a possible mechanism of Nrf2 activation. Since NO can modify the function or activity of target proteins through S-nitrosylation of cysteine, we attempted to investigate whether the cytoprotective effect of NO is mediated through Nrf2 activation by directly modifying cysteine residues of Keap1. Our present study reveals that treatment of rat pheochromocytoma (PC12) cells with an NO donor S-nitroso-N-acetylpenicillamine (SNAP) induced nuclear translocation and DNA binding of Nrf2. Under the same experimental conditions, there was NO-mediated S-nitrosylation of Keap1 observed, which coincided with the Nrf2 activation. Moreover, SNAP treatment caused phosphorylation of Nrf2, and pharmacological inhibition of protein kinase C (PKC) abolished the phosphorylation and nuclear localization of Nrf2. In conclusion, NO can activate Nrf2 by S-nitrosylation of Keap1 and alternatively by PKC-catalyzed phosphorylation of Nrf2 in PC12 cells.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Proteínas/metabolismo , S-Nitrosotióis/metabolismo , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Western Blotting , Núcleo Celular/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Dissulfetos/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ativação Enzimática , Imunofluorescência , Heme Oxigenase (Desciclizante)/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteína 1 Associada a ECH Semelhante a Kelch , Células PC12 , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Ratos , Transdução de Sinais , Estaurosporina/farmacologia
13.
Arterioscler Thromb Vasc Biol ; 31(6): 1403-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454807

RESUMO

OBJECTIVE: The adipocyte-secreted hormone adiponectin exerts important cardioprotective and antidiabetic effects. Little is known about its effect on vascular smooth muscle cells (VSMC), key cells in restenosis, hypertension, and atherosclerosis. METHODS AND RESULTS: Using human coronary artery VSMC, we found that recombinant adiponectin in the high-molecular-weight or trimeric forms but not the globular form induced VSMC differentiation through a mechanism similar to the classic feedback signaling used by rapamycin, a drug known to effectively inhibit restenosis on drug-eluting stents. Using a combination of pharmacological agents, small interfering RNA, and overexpression approaches, we demonstrated that adiponectin activated 5'-AMP-activated protein kinase α2 isoform, leading to inhibition of mammalian target of rapamycin complex 1 and S6K1. This in turn stabilized insulin receptor substrate-1, driving Akt2-mediated inhibition of FoxO4 and subsequent contractile protein induction. Although adiponectin and rapamycin have similarly beneficial effects on VSMC phenotype in both cell and organ culture, a direct comparison of the effects of rapamycin versus adiponectin on endothelial cells revealed distinct differences: rapamycin inhibited Akt phosphorylation, whereas adiponectin maintained it. Importantly, Akt activity preserves endothelial function. CONCLUSION: Adiponectin promotes VSMC differentiation and preserves endothelial cell Akt signaling, suggesting that targeting the adiponectin pathway may have advantages over rapamycin in developing new drug-eluting stent therapeutics.


Assuntos
Adiponectina/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/fisiologia , Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Fatores de Transcrição Forkhead , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Recombinantes/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Transcrição/fisiologia
14.
Mol Biol Rep ; 38(2): 1375-82, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20680465

RESUMO

The transcription factor AP-1 plays an important role in cellular proliferation, transformation and death. In this study, we report a novel human gene, AC3-33 (GenBank name: c3orf33, FLJ31139), which encodes a secretory protein that can inhibit Elk1 transcriptional activity via ERK1/2 pathway. The AC3-33 mRNA encodes a protein of 251 amino acids, which is a classical secretory protein. Functional investigation reveals that overexpression of AC3-33 significantly inhibit AP-1 activity and DNA-binding ability. Further investigation indicated that overexpression of AC3-33 significantly inhibit transcriptional activity of Elk1 and c-jun, but not c-fos. As for the upstream of signaling pathway of Elk-1, our study demonstrated that overexpression of AC3-33 significantly down-regulates phosphorylation of ERK1/2, but not JNK/SAPK or p38 MAPK. These results clearly indicate that AC3-33 is a novel member of the secretory family and inhibits Elk1 transcriptional activity via ERK1/2 MAPK.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas/metabolismo , Proteínas Elk-1 do Domínio ets/antagonistas & inibidores , Linhagem Celular , Biblioteca Gênica , Humanos , Proteínas de Membrana , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual , Transcrição Gênica , Proteínas Elk-1 do Domínio ets/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
J Biol Chem ; 285(52): 40461-71, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20880848

RESUMO

Methylating drugs such as temozolomide (TMZ) are widely used in the treatment of brain tumors including malignant glioblastoma. The mechanism of TMZ-induced glioblastoma cell death and apoptosis, however, is not fully understood. Here, we tested the potential involvement of AMP-activated protein kinase (AMPK) in this process. We found that methylating agents TMZ and N-methyl-N'-nitro-N-nitrosoguanidine induce AMPK activation in primary cultured human glioblastoma and glioblastoma cell lines. TMZ-induced O(6)-methylguanine production is involved in AMPK activation. O(6)-benzylguanine, an O(6)-methylguanine-DNA methyltransferase inhibitor, enhances TMZ-induced O(6)-methylguanine production, leading to enhanced reactive oxygen species production, which serves as an upstream signal for AMPK activation. Activation of AMPK is involved in TMZ-induced glioblastoma cell death and apoptosis. AMPK inhibitor (Compound C) or AMPKα siRNA knockdown inhibits TMZ-induced glioblastoma cell death and apoptosis, whereas AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside enhances it. In further studies, we found that activation of AMPK is involved in TMZ-induced p53 activation and subsequent p21, Noxa, and Bax up-regulation. Activation of AMPK by TMZ also inhibits mTOR complex 1 (mTORC1) signaling and promotes anti-apoptosis protein Bcl-2 down-regulation, which together mediate TMZ-induced pro-cell apoptosis effects. Our study suggests that activation of AMPK by TMZ contributes to glioblastoma cell apoptosis, probably by promoting p53 activation and inhibiting mTORC1 signaling.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Dacarbazina/análogos & derivados , Glioblastoma/metabolismo , Proteínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/metabolismo , Dacarbazina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Guanina/análogos & derivados , Guanina/metabolismo , Guanina/farmacologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Metilnitronitrosoguanidina/farmacologia , Complexos Multiproteicos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Temozolomida , Proteína X Associada a bcl-2/biossíntese
16.
J Virol ; 84(6): 2787-97, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20053736

RESUMO

The Epstein-Barr virus (EBV)-encoded LMP1 oncogene has a role in transformation, proliferation, and metastasis of several EBV-associated tumors. Furthermore, LMP1 is critically involved in transformation and growth of EBV-immortalized B cells in vitro. The oncogenic properties of LMP1 are attributed to its ability to upregulate anti-apoptotic proteins and growth signals. The transcriptional regulation of LMP1 is dependent on the context of cellular and viral proteins present in the cell. Here, we investigated the effect of several signaling pathways on the regulation of LMP1 expression. Inhibition of p38 signaling, using p38-specific inhibitors SB203580 and SB202190, downregulated LMP1 in estrogen-induced EREB2.5 cells. Similarly, p38 inhibition decreased trichostatin A-induced LMP1 expression in P3HR1 cells. Exogenous expression of p38 in lymphoblastoid cell lines (LCLs) led to an increase in LMP1 promoter activity in reporter assays, and this activation was mediated by the previously identified CRE site in the promoter. Inhibition of p38 by SB203580 and p38-specific small interfering RNA (siRNA) also led to a modest decrease in endogenous LMP1 expression in LCLs. Chromatin immunoprecipitation indicated decreased binding of CREB-ATF1 to the CRE site in the LMP1 promoter after inhibition of the p38 pathway in EREB2.5 cells. Taken together, our results suggest that an increase in p38 activation upregulates LMP1 expression. Since p38 is activated in response to stimuli such as stress or possibly primary infection, a transient upregulation of LMP1 in response to p38 may allow the cells to escape apoptosis. Since the p38 pathway itself is activated by LMP1, our results also suggest the presence of an autoregulatory loop in LMP1 upregulation.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 4 , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Oncogênicas Virais , Proteínas da Matriz Viral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Regiões Promotoras Genéticas , Proteínas/genética , Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Regulação para Cima , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
17.
J Nutr Biochem ; 21(2): 98-106, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19157820

RESUMO

The main aims of this study were to elucidate the effect of green tea catechins on Nudix-type motif 6 (NUDT6) suppression and to characterize NUDT6's biological activity. Our microarray data showed that the green tea component epicatechin-3-gallate suppressed NUDT6 expression, and this was confirmed by RT-PCR. Subsequently, the use of different catechins showed that the effect of epigallocatechin-3-gallate (EGCG) was stronger than that of other catechins. At the posttranscriptional level, EGCG decreased the RNA stability of NUDT6, indicating it as a potential mechanism of NUDT6 suppression. Further cloning of the 3' untranslated region of human NUDT6 mRNA resulted in reduced luciferase activity by EGCG treatment. This effect was at least, in part, mediated by the extracellular-signal-regulated kinase and p38MAPK pathways. Finally, increased cell proliferation and cell growth in soft agar were observed in NUDT6-overexpressing cells. These findings provide a novel mechanism for the suppression of the proliferative gene NUDT6 by green tea catechins in human colorectal cancer.


Assuntos
Anticarcinógenos/farmacologia , Catequina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Proteínas/fisiologia , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , Chá/química , Regiões 3' não Traduzidas/efeitos dos fármacos , Catequina/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/prevenção & controle , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Meia-Vida , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/genética , Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
18.
J Cell Physiol ; 222(2): 365-73, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19859909

RESUMO

Mitogenic effect of augmenter of liver regeneration (ALR), a protein produced and released by hepatocytes, on hepatocytes in vivo but not in vitro suggests that the effect is mediated by nonparenchymal cells. Since mediators produced by Kupffer cells are implicated in hepatic regeneration, we investigated receptor for ALR and its functions in rat Kupffer cells. Kupffer cells were isolated from rat liver by enzymatic digestion and centrifugal elutriation. Radioligand ([(125)I] ALR) receptor binding, ALR-induced GTP/G-protein association, and nitric oxide (NO), tumor necrosis factor (TNF)-alpha, and interleukin-6 (IL-6) synthesis were determined. High-affinity receptor for ALR, belonging to the G-protein family, with K(d) of 1.25 +/- 0.18 nM and B(max) of 0.26 +/- 0.02 fmol/microg DNA was identified. ALR stimulated NO, TNF-alpha, and IL-6 synthesis via cholera toxin-sensitive G-protein, as well as p38-MAPK activity and nuclear translocation of NFkappaB. While inhibitor of NFkappaB (MG132) inhibited ALR-induced NO synthesis, MG132 and p38-MAPK inhibitor (SB203580) abrogated ALR-induced TNF-alpha and IL-6 synthesis. ALR also prevented the release of mediator(s) from Kupffer cells that cause inhibition of DNA synthesis in hepatocytes. Administration of ALR to 40% partially hepatectomized rats increased expression of TNF-alpha, IL-6, and inducible nitric oxide synthase (iNOS) and caused augmentation of hepatic regeneration. These results demonstrate specific G-protein coupled binding of ALR and its function in Kupffer cells and suggest that mediators produced by ALR-stimulated Kupffer cells may elicit physiologically important effects on hepatocytes.


Assuntos
Toxina da Cólera/farmacologia , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Regeneração Hepática , Fígado/metabolismo , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Proliferação de Células , Hepatectomia , Hepatócitos/efeitos dos fármacos , Imidazóis/farmacologia , Interleucina-6/metabolismo , Células de Kupffer/efeitos dos fármacos , Leupeptinas/farmacologia , Fígado/efeitos dos fármacos , Fígado/cirurgia , Regeneração Hepática/efeitos dos fármacos , Masculino , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Comunicação Parácrina , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Ensaio Radioligante , Ratos , Ratos Endogâmicos Lew , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Oncol Rep ; 21(6): 1477-81, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19424626

RESUMO

CTRP3/cartducin, a novel secretory protein, is a member of the C1q and tumor necrosis factor (TNF)-related protein (CTRP) superfamily, and plays important roles in regulating both embryonic cartilage development and postnatal longitudinal bone growth. CTRP3/cartducin is expressed in human osteosarcomas. We hypothesized that CTRP3/cartducin might have a role in osteosarcoma tumor growth and metastasis. Murine osteosarcoma cell lines, NHOS and LM8, were used as a model. RT-PCR analysis showed that the mRNA level of CTRP3/cartducin was increased in these two murine osteosarcoma cell lines compared with its level in normal murine osteoblast MC3T3-E1 cells. Western blot analysis showed that the protein level of CTRP3/cartducin was also increased in these two osteosarcoma cell lines. Stimulation of NHOS and LM8 cells by CTRP3/cartducin promoted tumor cell growth but not migration in vitro. Further, CTRP3/cartducin stimulation led to the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) in these two osteosarcoma cell lines. MAPK/ERK kinase 1/2 (MEK1/2) inhibitor, U0126, blocked CTRP3/cartducin-induced cell proliferation. These results suggest that CTRP3/cartducin expression may play a role in osteosarcoma tumor growth associated with activation of the ERK1/2 signaling pathway.


Assuntos
Neoplasias Ósseas/metabolismo , Proliferação de Células , Osteossarcoma/metabolismo , Proteínas/metabolismo , Adipocinas , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Osteossarcoma/genética , Osteossarcoma/patologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
20.
J Neurosci ; 28(41): 10245-56, 2008 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-18842884

RESUMO

The neuropeptide Phe-Met-Arg-Phe-NH(2) (FMRFa) can induce transcription-dependent long-term synaptic depression (LTD) in Aplysia sensorimotor synapses. We investigated the role of the ubiquitin-proteasome system and the regulation of one of its components, ubiquitin C-terminal hydrolase (ap-uch), in LTD. LTD was sensitive to presynaptic inhibition of the proteasome and was associated with upregulation of ap-uch mRNA and protein. This upregulation appeared to be mediated by CREB2, which is generally regarded as a transcription repressor. Binding of CREB2 to the promoter region of ap-uch was accompanied by histone hyperacetylation, suggesting that CREB2 cannot only inhibit but also promote gene expression. CREB2 was phosphorylated after FMRFa, and blocking phospho-CREB2 blocked LTD. In addition to changes in the expression of ap-uch, the synaptic vesicle-associated protein synapsin was downregulated in LTD in a proteasome-dependent manner. These results suggest that proteasome-mediated protein degradation is engaged in LTD and that CREB2 may act as a transcription activator under certain conditions.


Assuntos
Aplysia/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Acetilação , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Células Cultivadas , Técnicas de Cocultura , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Regulação para Baixo , FMRFamida/farmacologia , Gânglios/citologia , Gânglios/metabolismo , Histonas/metabolismo , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios Aferentes/fisiologia , Fosforilação , Regiões Promotoras Genéticas , Inibidores de Proteassoma , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Sinapsinas/metabolismo , Sinaptossomos/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação/efeitos dos fármacos , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA