Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(48): 12791-12796, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29133416

RESUMO

Metastasis is a major cause of cancer-related deaths. Approximately 80% of patients with colorectal cancer develop liver metastasis and 20% develop lung metastasis. We found that at different stages of colon cancer, IFNγ secretion from peripheral blood mononuclear cells was decreased compared with healthy controls. The ribosomal S6 kinase (RSK) family of kinases has multiple cellular functions, and we examined their roles in this observed IFNγ decrease. Flow cytometry analysis of wild-type (WT) and RSK2 knockout (KO) mice revealed significantly lower levels of IFNγ in the RSK2 KO mice compared with the WT mice. Since IFNγ is a component of immunity, which contributes to protection against metastatic carcinomas, we conducted a colon cancer liver metastasis experiment. We found significantly greater metastasis in RSK2 KO mice compared with WT mice. Transcription factor T-bet can directly activate Ifnγ gene transcription. In vitro kinase assay results showed that RSK2 phosphorylated T-bet at serines 498 and 502. We show that phosphorylation of T-bet by RSK2 is required for IFNγ expression, because knockdown of RSK2 expression or overexpression of mutant T-bet reduces IFNγ mRNA expression. To verify the function of the phosphorylation sites, we overexpressed a constitutively active mutant T-bet (S498E/S502E) in bone marrow. Mutant T-bet restored the IFNγ mRNA levels and dramatically reduced the metastasis rate in these mice. Overall, these results indicate that phosphorylation of T-bet is required for the inhibition of colon cancer metastasis and growth through a positive regulation of RSK2/T-bet/IFNγ signaling.


Assuntos
Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Interferon gama/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Proteínas Quinases S6 Ribossômicas/genética , Proteínas com Domínio T/genética , Animais , Transplante de Medula Óssea , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Feminino , Humanos , Interferon gama/imunologia , Isoenzimas/genética , Isoenzimas/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Fosforilação , Proteínas Quinases S6 Ribossômicas/imunologia , Serina/metabolismo , Transdução de Sinais , Proteínas com Domínio T/imunologia , Transfecção , Irradiação Corporal Total
2.
J Biol Chem ; 289(41): 28192-201, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25122774

RESUMO

Non-small cell lung cancer (NSCLC) is the most lethal cancer, causing more than 150,000 deaths in the United States in 2013. The receptor tyrosine kinase inhibitors such as gefitinib are not perfect clinical therapeutic agents for NSCLC treatment due to primary or acquired tyrosine kinase inhibitor resistance. Herein, 3,6,2',4',5'-pentahydroxyflavone (36245-PHF) was identified as a multiple kinase inhibitor for NSCLC treatment based on the computational screening of a natural products database. 36245-PHF was shown to inhibit PI3K and Aurora A and B kinases and overcome gefitinib-resistant NSCLC growth. Our data clearly showed that 36245-PHF markedly inhibited anchorage-independent growth of gefitinib-resistant NSCLC cell lines and exerted a substantial chemotherapeutic effect following oral administration in a gefitinib-resistant NSCLC xenograft model. The evidence from three different subsequent methodological approaches, in vitro, ex vivo, and in vivo, all confirmed that 36245-PHF as a multiple protein kinase inhibitor. Overall, we identified 36245-PHF as a multiple protein kinase inhibitor and as a novel therapeutic agent to overcome gefitinib-resistant NSCLC growth, which could provide a new option for clinical NSCLC oral treatment.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Flavonoides/farmacologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Animais , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Gefitinibe , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos
3.
J Biol Chem ; 287(31): 25881-92, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22685297

RESUMO

TRAF2 has an important function in mediating the TNF-R signaling pathway toward activation of NF-κB and JNKs. Here we reveal a novel function of TRAF2 in the epidermal growth factor (EGF) signaling pathway. Knockdown of TRAF2 blocked EGF-induced AP-1 activity and anchorage- independent cell transformation. Notably, we showed that EGF induces ribosomal S6 kinase 2 (RSK2) ubiquitination, and knocking down TRAF2 suppresses ubiquitination of RSK2 induced by EGF. We also found that TRAF2 affects RSK2 activity through RSK2 ubiquitination. RSK2 plays a critical role in AP-1 activity mediated through CREB and c-Fos, which regulates anchorage-independent cell transformation. In addition, TRAF2 is overexpressed in colon cancer and required for colon cancer development, suggesting that TRAF2 might be a potential molecular target for cancer prevention and treatment.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator de Crescimento Epidérmico/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Poliubiquitina/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Carga Tumoral , Ubiquitinação
4.
J Biol Chem ; 286(9): 6946-54, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21183680

RESUMO

The ribosomal S6 kinase 2 (RSK2) is a member of the p90 ribosomal S6 kinase (p90RSK) family of proteins and plays a critical role in proliferation, cell cycle, and cell transformation. Here, we report that RSK2 phosphorylates caspase-8, and Thr-263 was identified as a novel caspase-8 phosphorylation site. In addition, we showed that EGF induces caspase-8 ubiquitination and degradation through the proteasome pathway, and phosphorylation of Thr-263 is associated with caspase-8 stability. Finally, RSK2 blocks Fas-induced apoptosis through its phosphorylation of caspase-8. These data provide a direct link between RSK2 and caspase-8 and identify a novel molecular mechanism for caspase-8 modulation by RSK2.


Assuntos
Apoptose/fisiologia , Caspase 8/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais/fisiologia , Ubiquitinação/fisiologia , Divisão Celular/fisiologia , Células HEK293 , Células HeLa , Humanos , Fosforilação/fisiologia , Estabilidade Proteica , Treonina/metabolismo
5.
Carcinogenesis ; 32(5): 779-85, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21349818

RESUMO

In addition to capsaicin, a transient receptor potential channel vanilloid subfamily 1 (TRPV1) agonist, two kinds of antagonists against this receptor are used as therapeutic drugs for pain relief. Indeed, a number of small molecule TRPV1 antagonists are currently undergoing Phase I/II clinical trials to determine their effect on relieving chronic inflammatory pain and migraine headache pain. However, we previously reported that the absence of TRPV1 in mice results in a striking increase in skin carcinogenesis, suggesting that chronic blockade of TRPV1 might increase the risk of tumor development. In this study, we found that a typical TRPV1 antagonist, AMG9810, promotes mouse skin tumor development. The topical application of AMG9810 resulted in a significant increase in the expression level of the epidermal growth factor receptor (EGFR) and its downstream Akt/mammalian target of rapamycin (mTOR)-signaling pathway. This increase was not only observed in AMG9810-treated tumor tissue but was also found in skin tissue treated with AMG9810. In telomerase-immortalized primary human keratinocytes, AMG9810 promoted proliferation that was mediated through the EGFR/Akt/mTOR-signaling pathway. In summary, our data suggest that the TRPV1 antagonist, AMG9810, promotes mouse skin tumorigenesis mediated through EGFR/Akt/mTOR signaling. Thus, the application of this compound for pain relief might increase the risk of skin cancer.


Assuntos
Acrilamidas/toxicidade , Compostos Bicíclicos Heterocíclicos com Pontes/toxicidade , Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Canais de Cátion TRPV/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Acrilamidas/administração & dosagem , Animais , Western Blotting , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Carcinógenos/toxicidade , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Células Cultivadas , Cocarcinogênese , Feminino , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Pelados , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/metabolismo , Canais de Cátion TRPV/metabolismo
6.
J Biol Chem ; 285(50): 39108-16, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20921231

RESUMO

MST1 (mammalian STE20-like kinase 1) is a serine/threonine kinase that is cleaved and activated by caspases during apoptosis. Overexpression of MST1 induces apoptotic morphological changes such as chromatin condensation, but the mechanism is not clear. Here we show that MST1 induces apoptotic chromatin condensation through its phosphorylation of histone H2AX at Ser-139. During etoposide-induced apoptosis in Jurkat cells, the cleavage of MST1 directly corresponded with strong H2AX phosphorylation. In vitro kinase assay results showed that MST1 strongly phosphorylates histone H2AX. Western blot and kinase assay results with a mutant S139A H2AX confirmed that MST1 phosphorylates H2AX at Ser-139. Direct binding of MST1 and H2AX can be detected when co-expressed in HEK293 cells and was also confirmed by an endogenous immunoprecipitation study. When overexpressed in HeLa cells, both the MST1 full-length protein and the MST1 kinase domain (MST1-NT), but not the kinase-negative mutant (MST1-NT-KN), could induce obvious endogenous histone H2AX phosphorylation. The caspase-3 inhibitor benzyloxycarbonyl-DEVD-fluoromethyl ketone (Z-DEVD-fmk) attenuates phosphorylation of H2AX by MST1 but cannot inhibit MST1-NT-induced histone H2AX phosphorylation, indicating that cleaved MST1 is responsible for H2AX phosphorylation during apoptosis. Histone H2AX phosphorylation and DNA fragmentation were suppressed in MST1 knockdown Jurkat cells after etoposide treatment. Taken together, our data indicated that H2AX is a substrate of MST1, which functions to induce apoptotic chromatin condensation and DNA fragmentation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/metabolismo , Histonas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Apoptose , Cromatina/química , Fragmentação do DNA , Etoposídeo/farmacologia , Células HEK293 , Células HeLa , Histonas/química , Humanos , Células Jurkat , Fosforilação , Serina/química
7.
FASEB J ; 24(9): 3490-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20385620

RESUMO

The ribosomal S6 kinase 2 (RSK2) is a well-known serine/threonine kinase and a member of the p90 ribosomal S6 kinase (p90RSK) family of proteins. It is activated downstream of the MEK/ERKs cascade by mitogenic stimuli such as EGF or TPA. Here, we show that RSK2 is activated by treatment with tumor necrosis factor-alpha (TNF-alpha) and directly phosphorylates IkappaBalpha at Ser-32, leading to IkappaBalpha degradation. The phosphorylation of IkappaBalpha promotes the activation and translocation of the nuclear factor-kappaB (NF-kappaB) subunits p65 and p50 to the nucleus. The net result is an increased NF-kappaB activity, which serves as a mechanism for RSK2 blockade of TNF-alpha-induced apoptosis and enhanced cell survival.


Assuntos
Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Inibidor de NF-kappaB alfa , Subunidade p50 de NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo
8.
Nat Struct Mol Biol ; 12(8): 699-707, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16007099

RESUMO

Inactivation of the p16(INK4a) tumor suppressor protein is critical for the development of human cancers, including human melanoma. However, the molecular basis of the protein's inhibitory effect on cancer development is not clear. Here we investigated a possible mechanism for p16(INK4a) inhibition of neoplastic transformation and UV-induced skin cancer. We show that p16(INK4a) suppresses the activity of c-Jun N-terminal kinases (JNKs) and that it binds to the glycine-rich loop of the N-terminal domain of JNK3. Although p16(INK4a) does not affect the phosphorylation of JNKs, its interaction with JNK inhibits c-Jun phosphorylation induced by UV exposure. This, in turn, interferes with cell transformation promoted by the H-Ras-JNK-c-Jun-AP-1 signaling axis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Melanoma/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Linhagem Celular Tumoral , Imunofluorescência , Vetores Genéticos , Glutationa Transferase , Humanos , Immunoblotting , Camundongos , Modelos Moleculares , Fosforilação , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
9.
Mol Cancer Res ; 18(7): 1028-1038, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32269074

RESUMO

The identification of oncogenic biomolecules as drug targets is an unmet need for the development of clinically effective novel anticancer therapies. In this study, we report for the first time that opsin 4/melanopsin (OPN4) plays a critical role in the pathogenesis of non-small cell lung cancer (NSCLC) and is a potential drug target. Our study has revealed that OPN4 is overexpressed in human lung cancer tissues and cells, and is inversely correlated with patient survival probability. Knocking down expression of OPN4 suppressed cells growth and induced apoptosis in lung cancer cells. We have also found that OPN4, a G protein-coupled receptor, interacted with Gα11 and triggered the PKC/BRAF/MEK/ERKs signaling pathway in lung adenocarcinoma cells. Genetic ablation of OPN4 attenuated the multiplicity and the volume of urethane-induced lung tumors in mice. Importantly, our study provides the first report of AE 51310 (1-[(2,5-dichloro-4-methoxyphenyl)sulfonyl]-3-methylpiperidine) as a small-molecule inhibitor of OPN4, suppressed the anchorage-independent growth of lung cancer cells and the growth of patient-derived xenograft tumors in mice. IMPLICATIONS: Overall, this study unveils the role of OPN4 in NSCLC and suggests that targeting OPN4 with small molecules, such as AE 51310 would be interesting to develop novel anticancer therapies for lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Opsinas de Bastonetes/metabolismo , Bibliotecas de Moléculas Pequenas/administração & dosagem , Regulação para Cima/efeitos dos fármacos , Células A549 , Adenocarcinoma de Pulmão/induzido quimicamente , Adenocarcinoma de Pulmão/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/induzido quimicamente , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/metabolismo , Camundongos , Bibliotecas de Moléculas Pequenas/farmacologia , Uretana/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 80(19): 4158-4171, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32816906

RESUMO

The key functional molecules involved in inflammatory bowel disease (IBD) and IBD-induced colorectal tumorigenesis remain unclear. In this study, we found that the apoptosis repressor with caspase recruitment domain (ARC) protein plays critical roles in IBD. ARC-deficient mice exhibited substantially higher susceptibility to dextran sulfate sodium (DSS)-induced IBD compared with wild-type mice. The inflammatory burden induced in ARC-deficient conditions was inversely correlated with CCL5 and CXCL5 levels in immune cells, especially CD4-positive T cells. Pathologically, ARC expression in immune cells was significantly decreased in clinical biopsy specimens from patients with IBD compared with normal subjects. In addition, ARC levels inversely correlated with CCL5 and CXCL5 levels in human biopsy specimens. ARC interacted with TNF receptor associated factor (TRAF) 6, regulating ubiquitination of TRAF6, which was associated with NF-κB signaling. Importantly, we identified a novel ubiquitination site at lysine 461, which was critical in the function of ARC in IBD. ARC played a critical role in IBD and IBD-associated colon cancer in a bone marrow transplantation model and azoxymethane/DSS-induced colitis cancer mouse models. Overall, these findings reveal that ARC is critically involved in the maintenance of intestinal homeostasis and protection against IBD through its ubiquitination of TRAF6 and subsequent modulation of NF-κB activation in T cells. SIGNIFICANCE: This study uncovers a crucial role of ARC in the immune system and IBD, giving rise to a novel strategy for IBD and IBD-associated colon cancer therapy.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Colorretais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Proteínas Musculares/metabolismo , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Azoximetano/toxicidade , Transplante de Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Quimiocina CCL5/metabolismo , Quimiocina CXCL5/metabolismo , Colite/induzido quimicamente , Neoplasias Colorretais/induzido quimicamente , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/química , Proteínas Musculares/genética , Ubiquitinação
11.
Theranostics ; 10(21): 9721-9740, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32863956

RESUMO

Rationale: Melanoma is an aggressive tumor of the skin and drug resistance is still a major problem in melanoma therapy. Novel targets and effective agents to overcome drug resistant melanoma are urgently needed in clinical therapy. Methods: Gene Expression Omnibus (GEO) database analysis, pathway enrichment analysis, and survival rate analysis were utilized to identify a candidate target. An anchorage-independent cell growth assay, flow cytometry, Western blot, and a xenograft mouse model were used to study the function of Aurora kinase B (AURKB) in both drug-sensitive and drug-resistant melanoma. Next, HI-511, a novel dual-target inhibitor targeting both AURKB and BRAF V600E, was designed and examined by an in vitro kinase assay. Methods as indicated above in addition to a BRAF V600E/PTEN-loss melanoma mouse model were used to demonstrate the effect of HI-511 on melanoma development in vitro and in vivo. Results: AURKB is highly expressed in melanoma and especially in vemurafenib-resistant melanoma and the expression was correlated with patient survival rate. Knocking down AURKB inhibited cell growth and induced apoptosis in melanoma, which was associated with the BRAF/MEK/ERKs and PI3-K/AKT signaling pathways. Importantly, we found that HI-511, a novel dual-target inhibitor against AURKB and BRAF V600E, suppresses both vemurafenib-sensitive and vemurafenib-resistant melanoma growth in vitro and in vivo by inducing apoptosis and mediating the inhibition of the BRAF/MEK/ERKs and PI3K/AKT signaling pathways. Conclusion: AURKB is a potential target for melanoma treatment. HI-511, a novel dual-target inhibitor against both AURKB and BRAF V600E, could achieve durable suppression of melanoma growth, even drug-resistant melanoma growth.


Assuntos
Aurora Quinase B/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vemurafenib/farmacologia
12.
Cancer Res ; 67(18): 8725-35, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875713

RESUMO

The c-jun-NH(2)-kinases (JNK) play a critical role in tumor promoter-induced cell transformation and apoptosis. Here, we showed that the nuclear factor of activated T3 (NFAT3) is phosphorylated by JNK1 or JNK2 at Ser(213) and Ser(217), which are located in the conserved SP motif. The transactivation domain of NFAT3 is found between amino acids (aa) 113 and 260 and includes the phosphorylation targets of JNK1 and JNK2. NFAT3 transactivation activity was suppressed in JNK1(-/-) or JNK2(-/-) mouse embryonic fibroblast (MEF) cells compared with wild-type MEF cells. Moreover, a 3xNFAT-luc reporter gene assay indicated that NFAT3 transcriptional activity was increased in a dose-dependent manner by JNK1 or JNK2. Double mutations at Ser(213) and Ser(217) suppressed NFAT3 transactivation activity; and SP600125, a JNK inhibitor, suppressed NFAT3-induced 3xNFAT-luciferase activity. Knockdown of JNK1 or JNK2 suppressed foci formation in NIH3T3 cells. Importantly, ectopic expression of NFAT3 inhibited AP-1 activity and suppressed foci formation. Furthermore, knockdown of NFAT3 enhanced Ras-JNK1 or JNK2-induced foci formation in NIH3T3 cells. Taken together, these results provided direct evidence for the anti-oncogenic potential of the NFAT3 transcription factor.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes Reporter , Humanos , Camundongos , Fatores de Transcrição NFATC/biossíntese , Fatores de Transcrição NFATC/genética , Células NIH 3T3 , Fosforilação , Serina/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/antagonistas & inibidores , Ativação Transcricional
13.
J Atten Disord ; 23(4): 384-397, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-25646023

RESUMO

OBJECTIVE: This study examined the effect of a 12-week table tennis exercise on motor skills and executive functions in children with ADHD. METHOD: Fifteen children with ADHD received the intervention, whereas 15 children with ADHD and 30 typically developing children did not. The Test of Gross Motor Development-2, Stroop, and Wisconsin Card Sorting Test (WCST) were conducted before and after the intervention. RESULTS: After the intervention, the ADHD training group scored significantly higher in the locomotor as well as object-control skills, Stroop Color-Word condition, and WCST total correct performance compared with the ADHD non-training group, and we noted improvements in the locomotor as well as object-control skills, Stroop Color-Word condition, and three aspects of the WCST performances of the ADHD training group over time. CONCLUSION: A 12-week table tennis exercise may have clinical relevance in motor skills and executive functions of children with ADHD.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/terapia , Terapia por Exercício/métodos , Destreza Motora/fisiologia , Criança , Desenvolvimento Infantil/fisiologia , Função Executiva/fisiologia , Exercício Físico/fisiologia , Feminino , Humanos , Masculino , Projetos Piloto , Teste de Classificação de Cartas de Wisconsin
14.
EBioMedicine ; 49: 145-156, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31707149

RESUMO

BACKGROUND: Barrett's esophagus (BE), a complication of gastroesophageal reflux disease (GERD), predisposes patients to esophageal adenocarcinoma (EAC). Reliable biomarkers for early detection and discovery of potential drug targets are urgently needed for improved BE and EAC patient outcomes. METHODS: Patient biopsy samples were evaluated for COX1/2, and thromboxane A2 synthase (TBXAS) expression. Circulating prostaglandins biosynthesis was determined using enzyme immunoassay kits. Anchorage-independent cell growth assay, crystal violet staining assay, and xenograft experiments were conducted to assess BE and EAC cell growth. A surgical mouse model of reflux (i.e., esophagoduodenostomy) was established and samples were analyzed using an enzyme immunoassay kit, immunohistochemistry, immunoblotting, or RT-PCR. Esophageal biopsy samples (pre- and post-intervention) were obtained from a randomized clinical trial in which participants were administered esomeprazole (40 mg) twice daily in combination with an acetylsalicylic acid (ASA) placebo or 81 or 325 mg ASA for 28 days. Esophageal biopsy specimens before and after the intervention period were analyzed. FINDINGS: COX2 and TBXAS are highly expressed in BE and EAC patients accompanied by a pronounced elevation of circulating TXA2 levels. ASA suppressed BE and EAC growth by targeting the TXA2 pathway. Additionally, biopsies from 49 patients (with similar baseline characteristics) showed that ASA substantially decreased serum TXA2 levels, resulting in reduced inflammation. INTERPRETATION: This study establishes the importance of the COX1/2-driven TXA2 pathway in BE and EAC pathophysiology and lays the groundwork for introducing a TXA2-targeting strategy for EAC prevention and early detection. FUNDING: Hormel Foundation, Exact Sciences, Pentax Medical, Intromedic and National Cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Esôfago de Barrett/tratamento farmacológico , Carcinogênese/patologia , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Terapia de Alvo Molecular , Transdução de Sinais , Tromboxano A2/metabolismo , Adenocarcinoma/sangue , Animais , Aspirina/farmacologia , Esôfago de Barrett/sangue , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Neoplasias Esofágicas/sangue , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fator de Transcrição STAT3/metabolismo , Tromboxano A2/sangue
15.
Mol Carcinog ; 47(10): 797-805, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18381589

RESUMO

Targeted molecular cancer therapies can potentially deliver treatment directly to a specific protein or gene to optimize efficacy and reduce adverse side effects often associated with traditional chemotherapy. Key oncoprotein and oncogene targets are rapidly being identified based on their expression, pathogenesis and clinical outcome. One such protein target is cyclooxygenase-2 (COX-2), which is highly expressed in various cancers. Research findings suggest that resveratrol (RSVL; 3,5,4'-trihydroxy-trans-stilbene) demonstrates nonselective COX-2 inhibition. We report herein that RSVL directly binds with COX-2 and this binding is absolutely required for RSVL's inhibition of the ability of human colon adenocarcinoma HT-29 cells to form colonies in soft agar. Binding of COX-2 with RSVL was compared with two RSVL analogues, 3,3',4',5',5-pentahydroxy-trans-stilbene (RSVL-2) or 3,4',5-trimethoxy-trans-stilbene (RSVL-3). The results indicated that COX-2 binds with RSVL-2 more strongly than with RSVL, but does not bind with RSVL-3. RSVL or RSVL-2, but not RSVL-3, inhibited COX-2-mediated PGE(2) production in vitro and ex vivo. HT-29 human colon adenocarcinoma cells express high levels of COX-2 and either RSVL or RSVL-2, but not RSVL-3, suppressed anchorage independent growth of these cells in soft agar. RSVL or RSVL-2 (not RSVL-3) suppressed growth of COX-2(+/+) cells by 60% or 80%, respectively. Notably, cells deficient in COX-2 were unresponsive to RSVL or RSVL-2. These data suggest that the anticancer effects of RSVL or RSLV-2 might be mediated directly through COX-2.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Estilbenos/farmacologia , Linhagem Celular Tumoral , Cromatografia de Afinidade , Humanos , Resveratrol , Espectrometria de Fluorescência
16.
Mol Carcinog ; 47(3): 172-83, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18095272

RESUMO

The cancer preventive action of (-)-epigallocatechin gallate (EGCG), found in green tea, is strongly supported by epidemiology and laboratory research data. However, the mechanism by which EGCG inhibits carcinogenesis and cell transformation is not clear. In this study, we report that EGCG suppressed epidermal growth factor (EGF)-induced cell transformation in JB6 cells. We also found that EGCG inhibited EGF-induced Fyn kinase activity and phosphorylation in vitro and in vivo. Fyn was implicated in the process because EGF-induced JB6 cell transformation was inhibited by small interfering RNA (siRNA)-Fyn-JB6 cells. With an in vitro protein-binding assay, we found that EGCG directly bound with the GST-Fyn-SH2 domain but not the GST-Fyn-SH3 domain. The K(d) value for EGCG binding to the Fyn SH2 domain was 0.367 +/- 0.122 microM and B(max) was 1.35 +/- 0.128 nmol/mg. Compared with control JB6 Cl41 cells, EGF-induced phosphorylation of p38 MAP kinase (p38 MAPK) (Thr180/Tyr182), ATF-2 (Thr71) and signal transducer and activator of transcription 1 (STAT1) (Thr727) was decreased in siRNA-Fyn-JB6 cells. EGCG could inhibit the phosphorylation of p38 MAPK, ATF-2, and STAT1. The DNA binding ability of AP-1, STAT1, and ATF-2 was also decreased in siRNA-Fyn-JB6 cells. Overall, these results demonstrated that EGCG interacted with Fyn and inhibited Fyn kinase activity and thereby regulated EGF-induced cell transformation. Inhibition of Fyn kinase activity is a novel and important mechanism that may be involved in EGCG-induced inhibition of cell transformation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Antioxidantes/farmacologia , Catequina/análogos & derivados , Transformação Celular Neoplásica/efeitos dos fármacos , Fator 2 Ativador da Transcrição/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína de Ligação a CREB/metabolismo , Catequina/metabolismo , Catequina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Relação Dose-Resposta a Droga , Fator de Crescimento Epidérmico/toxicidade , Glutationa Transferase/metabolismo , Cinética , Camundongos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Estrutura Terciária de Proteína/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
17.
Mol Biol Cell ; 16(9): 4437-53, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16000378

RESUMO

The ocular lens is the only organ that does not develop spontaneous tumor. The molecular mechanism for this phenomenon remains unknown. Through examination of the signaling pathways mediating stress-induced apoptosis, here we presented evidence to show that different from most other tissues in which the extracellular signal-regulated kinases (ERKs) pathway is generally implicated in mediation of survival signals activated by different factors, the RAF/MEK/ERK signaling pathway alone plays a key role in stress-activated apoptosis of lens epithelial cells. Treatment of N/N1003A cells with calcimycin, a calcium mobilizer, activates the RAF/MEK/ERK pathway through RAS, which is indispensable for the induced apoptosis because inhibition of this pathway by either pharmacological drug or dominant negative mutants greatly attenuates the induced apoptosis. Calcimycin also activates p38 kinase and JNK2, which are not involved in calcium-induced apoptosis. Downstream of ERK activation, p53 is essential. Activation of RAF/MEK/ERK pathway by calcimycin leads to distinct up-regulation of p53. Moreover, overexpression of p53 enhances calcimycin-induced apoptosis, whereas inhibition of p53 expression attenuates calcimycin-induced apoptosis. Up-regulation of p53 directly promotes Bax expression, which changes the integrity of mitochondria, leading to release of cytochrome c, activation of caspase-3 and eventually execution of apoptosis. Overexpression of alphaB-crystallin, a member of the small heat-shock protein family, blocks activation of RAS to inhibit ERK1/2 activation, and greatly attenuates calcimycin-induced apoptosis. Together, our results provide 1) a partial explanation for the lack of spontaneous tumor in the lens, 2) a novel signaling pathway for calcium-induced apoptosis, and 3) a novel antiapoptotic mechanism for alphaB-crystallin.


Assuntos
Apoptose/fisiologia , Sinalização do Cálcio/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Cadeia B de alfa-Cristalina/fisiologia , Quinases raf/fisiologia , Proteínas ras/fisiologia , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Calcimicina/farmacologia , Caspase 3 , Inibidores de Caspase , Linhagem Celular , Citocromos c/antagonistas & inibidores , MAP Quinase Quinase Quinase 1/metabolismo , MAP Quinase Quinase Quinase 2/metabolismo , Nitrilas/farmacologia , Coelhos
18.
Cancer Res ; 66(18): 9260-9, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16982771

RESUMO

Many beneficial properties have been attributed to (-)-epigallocatechin gallate (EGCG), including chemopreventive, anticarcinogenic, and antioxidant actions. In this study, we investigated the effects of EGCG on the function of glucose-regulated protein 78 (GRP78), which is associated with the multidrug resistance phenotype of many types of cancer cells. Our investigation was directed at elucidating the mechanism of the EGCG and GRP78 interaction and providing evidence about whether EGCG modulates the activity of anticancer drugs through the inhibition of GRP78 function. We found that EGCG directly interacted with GRP78 at the ATP-binding site of protein and regulated its function by competing with ATP binding, resulting in the inhibition of ATPase activity. EGCG binding caused the conversion of GRP78 from its active monomer to the inactive dimer and oligomer forms. Further, we showed that EGCG interfered with the formation of the antiapoptotic GRP78-caspase-7 complex, which resulted in an increased etoposide-induced apoptosis in cancer cells. We also showed that EGCG significantly suppressed the transformed phenotype of breast cancer cells treated with etoposide. Overall, these results strongly suggested that EGCG could prevent the antiapoptotic effect of GRP78, which usually suppresses the caspase-mediated cell death pathways in drug-treated cancer cells, contributing to the development of drug resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Catequina/análogos & derivados , Etoposídeo/farmacologia , Proteínas de Choque Térmico/antagonistas & inibidores , Chaperonas Moleculares/antagonistas & inibidores , Adenosina Trifosfatases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Neoplasias da Mama/tratamento farmacológico , Caspase 7/metabolismo , Inibidores de Caspase , Catequina/metabolismo , Catequina/farmacologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Humanos , Camundongos , Chaperonas Moleculares/metabolismo , Conformação Proteica/efeitos dos fármacos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo
19.
Cancer Res ; 66(11): 5729-36, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16740711

RESUMO

The oncoprotein c-Jun is a component of the activator protein-1 transcription factor complex, which is involved in cellular proliferation, transformation, and death. The stabilization of c-Jun is critically important for its function. The phosphorylation of c-Jun by c-Jun NH(2)-terminal kinase 1 and extracellular signal-regulated protein kinases reduces c-Jun ubiquitination resulting in increased stabilization of c-Jun. In this report, we showed that COOH-terminal Src kinase (CSK) binds with and phosphorylates c-Jun at Y26 and Y170. Phosphorylation of c-Jun by CSK, in opposition to c-Jun NH(2)-terminal kinase 1 and extracellular signal-regulated protein kinases, promoted c-Jun degradation and reduced stability. By promoting c-Jun degradation, CSK helps to maintain a low steady-state level of c-Jun, thereby inhibiting activator protein-1 activity and cell transformation caused by c-Jun. These results indicated that this function of CSK controls cell proliferation under normal growth conditions and may have implications for CSK loss of function in carcinogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Células HeLa , Humanos , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Células NIH 3T3 , Fosforilação , Ligação Proteica , Fator de Transcrição AP-1/metabolismo , Ubiquitina/metabolismo
20.
Oncogene ; 37(26): 3501-3513, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29563609

RESUMO

Inflammation is a complex biological host reaction to tissue damage, infection and trauma. Extensive study of the inflammatory response has led to the identification of several protein kinases that are essential for signaling and could be potential therapeutic targets. The RSK family of kinases has multiple cellular functions. In our study, we found that RSK2 is a mediator for inflammation signaling and interacts with TRAF6. In vitro kinase assay results indicated that RSK2 strongly phosphorylates TRAF6 at serines 46, 47 and 48. Ectopic overexpression of TRAF6 or knocking down RSK2 expression confirmed that RSK2 is a positive regulator of TRAF6 K63 ubiquitination. TRAF6 is also required for RSK2 ubiquitination. TRAF6 bridges the TNF receptor superfamily and intracellular signaling for the induction of proinflammatory cytokines. We developed a colon inflammation model using RSK2 wild type (WT) and knockout (KO) mice. As expected, F4/80 and CD3 infiltration were significantly upregulated in WT mice compared to RSK2 KO mice. Furthermore, inflammation signaling, including Ikkα/ß, p38 and JNKs, was dramatically upregulated in WT mice. Colon tissue immunoprecipitation results further confirmed that TRAF6 K63 ubiquitination was lower in RSK2 KO mice. Overall, these results indicate that phosphorylation of TRAF6 (S46, 47, 48) by RSK2 is required for TRAF6 K63 ubiquitination and inflammation signaling.


Assuntos
Colite/patologia , Colo/patologia , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Colo/imunologia , Feminino , Células HEK293 , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação , Células RAW 264.7 , Ubiquitinação/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA