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










Base de dados
Intervalo de ano de publicação
1.
Bone Joint Res ; 11(5): 304-316, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35579298

RESUMO

AIMS: The association of auraptene (AUR), a 7-geranyloxycoumarin, on osteoporosis and its potential pathway was predicted by network pharmacology and confirmed in experimental osteoporotic mice. METHODS: The network of AUR was constructed and a potential pathway predicted by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) terms enrichment. Female ovariectomized (OVX) Institute of Cancer Research mice were intraperitoneally injected with 0.01, 0.1, and 1 mM AUR for four weeks. The bone mineral density (BMD) level was measured by dual-energy X-ray absorptiometry. The bone microstructure was determined by histomorphological changes in the femora. In addition, biochemical analysis of the serum and assessment of the messenger RNA (mRNA) levels of osteoclastic markers were performed. RESULTS: In total, 65.93% of the genes of the AUR network matched with osteoporosis-related genes. Osteoclast differentiation was predicted to be a potential pathway of AUR in osteoporosis. Based on the network pharmacology, the BMD and bone mineral content levels were significantly (p < 0.05) increased in the whole body, femur, tibia, and lumbar spine by AUR. AUR normalized the bone microstructure and the serum alkaline phosphatase (ALP), bone-specific alkaline phosphatase (bALP), osteocalcin, and calcium in comparison with the OVX group. In addition, AUR treatment reduced TRAP-positive osteoclasts and receptor activator of nuclear factor kappa-B ligand (RANKL)+nuclear factor of activated T cells 1 (NFATc1)+ expression in the femoral body. Moreover, the expressions of initiators for osteoclastic resorption and bone matrix degradation were significantly (p < 0.05) regulated by AUR in the lumbar spine of the osteoporotic mice. CONCLUSION: AUR ameliorated bone loss by downregulating the RANKL/NFATc1 pathway, resulting in improvement of osteoporosis. In conclusion, AUR might be an ameliorative cure that alleviates bone loss in osteoporosis via inhibition of osteoclastic activity. Cite this article: Bone Joint Res 2022;11(5):304-316.

2.
Histopathology ; 67(4): 520-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25786562

RESUMO

AIMS: From the viewpoint of histogenesis, lung adenocarcinoma can be subdivided into two groups: terminal respiratory unit (TRU) and non-TRU types. We recently reported a non-TRU type adenocarcinoma designated as ciliated adenocarcinoma (we now prefer central type adenocarcinoma). We suggest reasons that mucinous adenocarcinoma should encompass central type adenocarcinoma to represent its biological characteristics as non-TRU type adenocarcinoma. METHODS AND RESULTS: Mucin (MUC)5AC and MUC5B were expressed more significantly in non-TRU type adenocarcinoma (P < 0.01). Thirty-five (76.1%) and 45 cases (97.8%) of 46 non-TRU type adenocarcinoma showed positivity for MUC5AC and MUC5B. Twelve (7.6%) and eight (5.1%) cases of 157 TRU type adenocarainoma showed positivity for MUC5B and MUC5AC. NKX2-1 gene expression was measured with quantitative reverse transcription-polymerase chain reaction (qRT-PCR). ΔΔCt of NKX2-1 gene expression was 6.79 for TRU type adenocarcinoma and 0.6 for non-TRU type adenocarcinoma. Overall survival and disease-free survival were poorer in non-TRU type adenocarcinoma (P = 0.02 and P = 0.03). A multivariate test also showed that non-TRU type adenocarcinoma is an independent prognostic factor (P = 0.04). CONCLUSION: MUC5AC and MUC5B were specific makers for non-TRU adenocarcinoma, including both central type adenocarcinoma and mucinous adenocarcinoma. We suggest that non-TRU type adenocarcinoma presents a poorer prognosis, so it should be regarded separately from TRU type adenocarcinoma.


Assuntos
Adenocarcinoma Mucinoso/patologia , Biomarcadores Tumorais/análise , Neoplasias Pulmonares/patologia , Mucina-5AC/biossíntese , Mucina-5B/biossíntese , Adenocarcinoma Mucinoso/classificação , Adenocarcinoma Mucinoso/mortalidade , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/classificação , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Mucina-5AC/análise , Mucina-5B/análise , Prognóstico , Modelos de Riscos Proporcionais , Reação em Cadeia da Polimerase em Tempo Real
3.
Toxicol In Vitro ; 23(4): 622-6, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19268519

RESUMO

The mechanisms involving the inhibitory effects of ascorbic acid (AA) on carcinogenesis have not fully defined, except for its free-radical scavenging activity against oxidative DNA damage. In this study, we examined the effects of AA on the expression of the aryl hydrocarbon receptor (AhR)-regulated gene cytochrome P4501A1 (CYP1A1), which catalyzes the activation of genotoxic metabolites that can lead to mutagenesis. Cultured human HepG2 cells were incubated with AA with or without the potent AhR agonist/CYP1A1 inducer 2,3,7,8-tetrachloridibenxo-p-dioxin (TCDD). AA was highly effective at suppressing CYP1A1 induction following coincubation of the cells with 1nM TCDD. The preventive effects of AA were seen at the level of mRNA and protein expression as well as CYP1A1-specific 7-ethoxyresorufin O-deethylase (EROD) activity. A transient transfection assay using a dioxin response element (DRE)-linked luciferase reporter and an electrophoretic mobility shift assay revealed that AA reduced the amount of AhR that could form a complex with the DRE sequence in the promoter region of the CYP1A1 gene. In addition, AA inhibited the TCDD-induced Ecto-ATPase activity, which is known to be requiring for AhR translocation to the nucleus. These results suggest that AA may exert at least part of its anticarcinogenesis effect by controlling the expression of CYP1A1 at the transcription level.


Assuntos
Ácido Ascórbico/farmacologia , Citocromo P-450 CYP1A1/genética , Dibenzodioxinas Policloradas/toxicidade , Antígenos CD/metabolismo , Apirase/metabolismo , Células Cultivadas , Citocromo P-450 CYP1A1/metabolismo , Humanos , Regiões Promotoras Genéticas , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/fisiologia
4.
Oncol Rep ; 20(6): 1569-75, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19020743

RESUMO

Overexpression of epidermal growth factor (EGF) and urokinase plasminogen activator receptor (uPAR) have been observed in human gastric cancers. However, the interaction between EGF and uPAR in gastric cancer has not been well elucidated. In this study, we investigated the effect of EGF on uPAR expression and the underlying signal pathways in human gastric cancer AGS cells. EGF induced uPAR mRNA expression in a time- and concentration-dependent manner. EGF also induced uPAR promoter activity. In addition, EGF induced the activation of extracellular signal regulated kinase-1/2 (ERK-1/2) and P38 mitogen-activated protein kinase (MAPK) but not the activation of c-Jun amino terminal kinase. A specific inhibitor of MEK-1 (an upstream effector of ERK-1/2) and a dominant negative MEK-1 were able to suppress the EGF-induced uPAR promoter activity. Site-directed mutagenesis and electrophoretic mobility shift assays demonstrated that the binding sites of transcription factors, activator protein-1 (AP-1) and nuclear factor (NF)-kappaB, are involved in the EGF-induced uPAR transcription. Suppression of the EGF-induced uPAR promoter activity by the AP-1 decoy oligonuclotide, as well as expression vectors encoding mutated-type NF-kappaB-inducting kinase and I-kappaB, confirmed that the activation of AP-1 and NF-kappaB are essential for the EGF-induced uPAR upregulation. The AGS cells pretreated with EGF showed a remarkably enhanced invasiveness and this effect was partially abrogated by uPAR neutralizing antibodies and by the inhibitors of ERK-1/2, AP-1, and NF-kappaB. The above results suggest that EGF induces uPAR expression via ERK-1/2, AP-1, and NF-kappaB signaling pathways and, in turn, stimulates cell invasiveness in human gastric cancer AGS cells.


Assuntos
Carcinoma/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Fator de Transcrição AP-1/metabolismo , Humanos , Invasividade Neoplásica , Oligonucleotídeos/química , Regiões Promotoras Genéticas
5.
Anticancer Res ; 27(5A): 3411-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17970088

RESUMO

The overexpression of urokinase-type plasminogen activator receptor (uPAR) is closely related to tumor cell invasion. Therefore, strategies for down-regulating uPAR expression may be of clinical utility. This study examined the effects of triptolide, which is a diterpenoid triepoxide extracted from the Chinese herb Tripterygium wilfordii Hook F., on the induction of uPAR in human gastric cancer AGS cells. Triptolide inhibited the phorbol 12-myristate 13-acetate (PMA)-induced uPAR mRNA and protein expression in a dose-dependent manner, and reduced the uPAR transcriptional activity. The stability of the uPAR transcripts was not altered by the triptolide treatment. The signals involved in uPAR induction by PMA were investigated to determine the mechanisms for the triptolide-mediated regulation of uPAR. The inhibitors of extracellular signal-regulated kinases 1 and 2 (Erk-1/2, PD98059), c-Jun N-terminal kinases (JNK, SP600125) and nuclear factor-kappa B (NF-kappaB, Bay11-7082) inhibited the PMA-induced expression of uPAR, which suggests that PMA induces uPAR through multiple signals. Triptolide suppressed the PMA-induced activation of NF-kappaB but not Erk-1/2 and JNKI The inhibitory effect of triptolide on the activation of NF-kappaB was confirmed by an electrophoretic mobility shift assay and NF-kappaB dependent transcription studies. AGS cells treated with PMA showed a remarkably enhanced invasiveness, which was partially abrogated by triptolide and uPAR-neutralizing antibodies. This suggests that triptolide may exert at least part of its anti-invasive effect in gastric cancer by controlling the expression of uPAR through the suppression of NF-kappaB activation.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Diterpenos/farmacologia , NF-kappa B/antagonistas & inibidores , Fenantrenos/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Neoplasias Gástricas/tratamento farmacológico , Acetato de Tetradecanoilforbol/antagonistas & inibidores , Carcinógenos/antagonistas & inibidores , Carcinógenos/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Interações Medicamentosas , Compostos de Epóxi/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Invasividade Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos
6.
Int J Mol Med ; 19(4): 689-97, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17334646

RESUMO

The gastric pathogen, helicobacter pylori (H. pylori), has been associated with the progression of gastric cancer. It was previously reported that H. pylori induced urokinase plasminogen activator receptor (uPAR) expression and stimulated cell invasiveness in human gastric cancer AGS cells. However, the precise mechanisms for how H. pylori upregulates uPAR are unclear. This study investigated the underlying signal pathways in H. pylori-induced uPAR in human gastric cancer AGS cells. The intracellular H2O2 content, as determined using H2O2-sensitive probe 2',7'-dichlorodihydrofluorescein, increased after the H. pylori treatment. N-acetyl cysteine (NAC), an antioxidant, prevented the H. pylori-induced production of H2O2 and uPAR expression. In addition, exogenous H2O2 was found to increase uPAR mRNA expression and its promoter activity. Site-directed mutagenesis of the potential NF-kappaB element in the uPAR promoter showed that the redox-sensitive transcription factor NF-kappaB was essential for H. pylori-induced uPAR expression. The expression of vectors encoding a mutated-type NF-kappaB-inducing kinase and I-kappaB, and a specific inhibitor of NF-kappaB (BAY11-7082) decreased the H. pylori-induced uPAR promoter activity. Chromatin immunoprecipitation and the electrophoretic mobility shift assay confirmed that H. pylori increased the DNA binding activity of NF-kappaB. With the aid of NAC and H2O2, it was determined that reactive oxygen species (ROS) is an upstream signaling molecule for activating the NF-kappaB induced by H. pylori. The enhanced AGS cell invasiveness by H. pylori was partially abrogated by an NAC and BAY11-7082 treatment. These results suggest that the ROS and NF-kappaB signaling pathway is important in H. pylori-induced uPAR expression and the increased cell invasiveness of human gastric cancer AGS cells.


Assuntos
Carcinoma/patologia , Helicobacter pylori , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/agonistas , Neoplasias Gástricas/patologia , Acetilcisteína/farmacologia , Carcinoma/enzimologia , Carcinoma/microbiologia , Sequestradores de Radicais Livres/farmacologia , Humanos , NF-kappa B/antagonistas & inibidores , Invasividade Neoplásica , Nitrilas/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/microbiologia , Sulfonas/farmacologia , Células Tumorais Cultivadas
7.
Life Sci ; 80(21): 1957-65, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17379255

RESUMO

Monocyte chemotactic protein-1 (MCP-1) is a potent chemoattractant for monocytes and plays a key role in various inflammatory responses, including atherosclerosis. In this study, we examined the effect of (-)-epigallocatechin-3-gallate (EGCG), a major green tea catechin, on the expression of MCP-1 in human endothelial ECV304 cells. EGCG markedly inhibited the phorbol 12-myristate 13-acetate (PMA)-induced MCP-1 mRNA and protein levels in a dose-dependent manner. EGCG was also found to reduce the MCP-1 transcriptional activity. The upregulation of MCP-1 by PMA was significantly inhibited by blockade of P38 mitogen-activated protein kinase (MAPK) and NF-kappaB, but not by blockade of extracellular-signal-regulated kinase and c-Jun N-terminal kinase pathway. Furthermore, The PMA-induced p38 MAPK and NF-kappaB activation were obviously attenuated after pretreating ECV304 cells with EGCG. The conditioned media from the endothelial ECV304 cells treated with PMA could remarkably stimulate the migration of THP-1 monocytes and this effect was partially abrogated by MCP-1 neutralizing antibodies. Moreover, the media from the EGCG-pretreated ECV304 cells lost the stimulatory activity for THP-1 migration. These results suggest that EGCG may exert an anti-inflammatory effect in endothelial cells by controlling MCP-1 expression, at least in part, mediated through the suppression of p38 MAPK and NF-kappaB activation.


Assuntos
Catequina/análogos & derivados , Quimiocina CCL2/metabolismo , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/metabolismo , Northern Blotting , Western Blotting , Catequina/farmacologia , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/genética , Meios de Cultura/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Int J Oncol ; 29(5): 1247-52, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17016658

RESUMO

Platelet-derived growth factor (PDGF) has been known to induce vascular endothelial growth factor (VEGF) expression in human vascular smooth muscle cells (hVSMCs). We previously reported that Erk-1/2 and AP-1 pathways are crucial in the PDGF-induced VEGF expression in hVSMCs . In this study, we investigated the effect of epigallocatechin-3-gallate (EGCG), the major green tea catechin, on the PDGF-induced VEGF expression in hVSMCs and the underlying mechanisms. EGCG were found to inhibit dose-dependently the VEGF expression and activation of PDGF receptor, Erk-1/2 and AP-1 induced by PDGF. In addition, cell free studies demonstrated that EGCG could directly inhibit the Erk-1/2 activity. Conditioned media from the hVSMCs treated with PDGF could remarkably stimulate the in vitro growth of human umbilical vein endothelial cells (HUVECs) but the media from the EGCG-pretreated hVSMCs lost its stimulatory activity for HUVEC proliferation. These results suggest that EGCG may exert the anti-angiogenic effect by inhibiting the PDGF-induced VEGF expression at multiple signaling levels.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticarcinógenos/farmacologia , Catequina/análogos & derivados , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Catequina/farmacologia , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Cordão Umbilical/citologia , Fator A de Crescimento do Endotélio Vascular/genética
9.
Int J Oncol ; 28(1): 135-41, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16327989

RESUMO

Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) have been shown to communicate with each other via cytokine signaling during neovascularization. In this study, we investigated the effect of platelet-derived growth factor (PDGF), a cytokine released from tumors and ECs, on vascular endothelial growth factor (VEGF) expression in human VSMCs and underlying signal transduction pathways. PDGF induced VEGF expression in a time- and concentration-dependent manner. PDGF induced the activation of extra-cellular signal-regulated kinase-1/2 (ERK-1/2), but not the activation of c-jun amino terminal kinase (JNK) and P38 mitogen-activated protein kinase (MAPK). Specific inhibitor of mitogen-activated protein kinase kinase (MEK)-1 was found to suppress VEGF expression and promoter activity. The expression of vectors encoding a mutated-type MEK-1 decreased the VEGF promoter activity. Electrophoretic mobility shift assay revealed that PDGF dose-dependently increased the DNA binding activity of AP-1. Transient transfection studies using an AP-1 decoy oligonucleotide confirmed that the activation of AP-1 is involved in PDGF-induced VEGF upregulation. Conditioned media from the human VSMCs pretreated with PDGF could remarkably stimulate the in vitro growth of human umbilical vein endothelial cells and this effect was partially abrogated by VEGF neutralizing antibodies. The above results suggest that ERK-1/2 and AP-1 signaling pathways are involved in the PDGF-induced VEGF expression in human VSMCs and that these paracrine signaling pathways induce endothelial cell proliferation.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Fator de Transcrição AP-1/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proliferação de Células , Células Endoteliais , Humanos , Músculo Liso Vascular/fisiologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Transdução de Sinais , Regulação para Cima
10.
Biochem Biophys Res Commun ; 333(3): 874-80, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15963460

RESUMO

The gastric pathogen Helicobacter pylori (H. pylori) is suggested to be associated with gastric cancer progression. In this study, we investigated the effect of H. pylori on urokinase plasminogen activator receptor (uPAR) expression which has been known to correlate closely with gastric cancer invasion. H. pylori induced the uPAR expression in a time- and concentration-dependent manner. Specific inhibitors and inactive mutants of MEK-1 and JNK were found to suppress the H. pylori-induced uPAR expression and the uPAR promoter activity. Electrophoretic mobility shift assay and transient transfection study using an AP-1 decoy oligonucleotide confirmed that the activation of AP-1 is involved in the H. pylori-induced uPAR upregulation. The AGS cells treated with H. pylori showed a remarkably enhanced invasiveness, and this effect was partially abrogated by uPAR-neutralizing antibodies. These results suggest that H. pylori induces uPAR expression via Erk-1/2, JNK, and AP-1 signaling pathways and, in turn, stimulates the cell invasiveness in human gastric cancer AGS cells.


Assuntos
Helicobacter pylori/fisiologia , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Ativação Enzimática , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fator de Transcrição AP-1/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética
11.
Int J Oncol ; 26(6): 1669-74, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15870884

RESUMO

Overexpression of urokinase plasminogen activator receptor (uPAR) is known to correlate closely with tumor cell invasion and metastasis. In gastric cancer, however, the mechanism for induction of uPAR remains to be elucidated. In this study, to investigate the effect of reactive oxygen species (ROS) on uPAR expression and the underlying signal pathways in human gastric cancer AGS cells, phenazine methosulfate (PMS) and H2O2 were used as ROS generator. PMS and H2O2 induced the uPAR expression in time- and concentration-dependent manner. PMS and H2O2 also induced the activation of extracellular signal-regulated kinases (Erk)-1/2. A specific inhibitor of MEK-1 (PD980590) was found to suppress the PMS-induced uPAR expression and the uPAR promoter activity. Expression of vectors encoding a mutated-type MEK-1 resulted in decreases in the uPAR promoter activity. Electrophoretic mobility shift assay revealed that PMS increased time-dependently the DNA binding activity of AP-1. Transient transfection studies using AP-1 decoy confirmed that the activation of AP-1 is involved in PMS-induced uPAR upregulation. The AGS cells pretreated with PMS showed a remarkably enhanced invasiveness and this effect was partially abrogated by uPAR neutralizing antibodies. The above results suggest that ROS induces uPAR expression via Erk-1/2 and AP-1 signaling pathways and, in turn, stimulates the cell invasiveness in human gastric cancer AGS cells.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/genética , Transdução de Sinais/fisiologia , Neoplasias Gástricas/metabolismo , Fator de Transcrição AP-1/fisiologia , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , RNA Mensageiro/análise , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Neoplasias Gástricas/patologia
12.
Oncogene ; 23(39): 6603-11, 2004 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-15208668

RESUMO

Recent studies have suggested that the expression of interleukin-8 (IL-8) directly correlates with the vascularity of human gastric carcinomas. In this study, the effect of IL-1beta on IL-8 expression in human gastric cancer TMK-1 cells and the underlying signal transduction pathways were investigated. IL-1beta induced the IL-8 expression in a time- and concentration-dependent manner. IL-1beta induced the activation of extracellular signal-regulated kinases-1/2 and P38 mitogen-activated protein kinase (MAPK), but not the activation of c-jun amino-terminal kinse and Akt. Specific inhibitors of MEK-1 (PD980590) and P38 MAPK (SB203580) were found to suppress the IL-8 expression and the IL-8 promoter activity. Expression of vectors encoding a mutated-type MEK-1 and P38 MAPK resulted in decrease in the IL-8 promoter activity. IL-1beta also induced the production of reactive oxygen species (ROS). N-acetyl cysteine (NAC) prevented the IL-1beta-induced ROS production and IL-8 expression. In addition, exogenous H2O2 could induce the IL-8 expression. Deletional and site-directed mutagenesis studies on the IL-8 promoter revealed that activator protein-1 (AP-1) and nuclear factor (NF)-kappaB sites were required for the IL-1beta-induced IL-8 transcription. Electrophoretic mobility shift assay confirmed that IL-1beta increased the DNA-binding activity of AP-1 and NF-kappaB. Inhibitor (PD980590, SB203580) and ROS scavenger (NAC) studies revealed that the upstream signalings for the transcription factors AP-1 and NF-kappaB were MAPK and ROS, respectively. Conditioned media from the TMK-1 cells pretreated with IL-1beta could remarkably stimulate the in vitro growth of HUVEC and this effect was partially abrogated by IL-8-neutralizing antibodies. The above results suggest that MAPK-AP-1 and ROS-NF-kappaB signaling pathways are involved in the IL-1beta-induced IL-8 expression and that these paracrine signaling pathways induce endothelial cell proliferation.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Interleucina-1/fisiologia , Interleucina-8/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais/fisiologia , Neoplasias Gástricas/metabolismo , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Humanos , RNA Mensageiro/genética , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/patologia
13.
Anticancer Res ; 24(2B): 747-53, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15161022

RESUMO

Overexpression of matrix metalloproteinases (MMPs) has been known to correlate closely with tumor cell invasion and strategies to down-regulate their expression may ultimately be of clinical utility. In this study, we investigated the effects of (-)-epigallocatechin gallate (EGCG), a major green tea catechin, on the cell invasiveness and MMP-9 induction in human gastric cancer AGS cells. EGCG inhibited the phorbol 12-myristate 13-acetate (PMA)-induced cell invasiveness and MMP-9 expression in a dose-dependent manner. EGCG treatment was found to reduce the MMP-9 transcriptional activity. To further study the mechanisms for the EGCG-mediated regulation of MMP-9, the effects of EGCG on transcription factor AP-1 and mitogen-activated protein kinase (MAPK) activities were examined. The results showed that EGCG suppressed the PMA-induced AP-1 activation. EGCG also abrogated the PMA-induced activation of extracellular-regulated protein kinase (Erk) and c-jun N-terminal kinase (JNK), which are upstream modulators of AP-1. These results suggest that EGCG may exert at least part of its anti-invasive effect in gastric cancer by controlling MMP expression through the suppression of MAPK and AP-1 activation.


Assuntos
Antineoplásicos/farmacologia , Catequina/análogos & derivados , Catequina/farmacologia , Inibidores de Metaloproteinases de Matriz , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/enzimologia , Fator de Transcrição AP-1/antagonistas & inibidores , Carcinógenos/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Indução Enzimática/efeitos dos fármacos , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Fosforilação , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/fisiologia , Ativação Transcricional/efeitos dos fármacos , Transfecção
14.
Eur J Pharmacol ; 487(1-3): 1-6, 2004 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15033370

RESUMO

(-)-Epigallocatechin-3-gallate (EGCG), a main flavanol of green tea, potently suppressed the urokinase-type plasminogen activator (uPA) expression in human fibrosarcoma HT 1080 cells. EGCG induced not only the suppression of the uPA promoter activity but also the destabilization of uPA mRNA. EGCG inhibited the phosphorylation of extracellular signal-regulated kinases 1 and 2 (Erk-1/2) and P38 mitogen-activated protein kinase (MAPK), but not the phosphorylation of c-jun N-terminal kinase (JNK) and Akt. Specific inhibitors of Erk-1/2 (2'-amino-3'-methoxyflavone, PD98059) and P38 MAPK (pyridinylimidazole, SB203580) were found to suppress the uPA expression and the uPA promoter activity. However, the specific inhibitors did not affect the uPA mRNA stability. These results suggest that EGCG could regulate the uPA expression by at least two different mechanisms: EGCG may inhibit the Erk-1/2 and P38 MAPK, leading to suppression of the uPA promoter activity, and EGCG may destabilize the uPA mRNA in an Erk-1/2- and p38 MAPK-independent way.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Catequina/análogos & derivados , Catequina/farmacologia , Fibrossarcoma/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Northern Blotting , Western Blotting , Cloranfenicol O-Acetiltransferase/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Indicadores e Reagentes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica/prevenção & controle , Fosforilação , RNA Mensageiro/biossíntese , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Int J Mol Med ; 13(1): 81-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14654975

RESUMO

Silibinin, the flavonoid found in the milk thistle, has been shown to suppress cell growth and exhibit anti-cancer effects. Some flavonoids were reported to inhibit angiogenesis which is essential for tumor growth and metastasis. In this study, to clarify the underlying mechanisms for the anti-cancer effect of silibinin, we examined the effects of silibinin on human endothelial ECV304 cells. Silibinin was found to suppress the growth and induce the apoptosis of ECV304 cells. The induction of apoptosis by silibinin was confirmed by ladder-patterned DNA fragmentation, cleaved and condensed nuclear chromatin and DNA hypoploidy. Silibinin could effectively inhibit constitutive NF-kappaB activation as revealed by electrophoretic mobility shift assay and NF-kappaB-dependent luciferase reporter study. Consistent with this, silibinin treatment resulted in a significant decrease in the nuclear level of p65 subunit of NF-kappaB. In addition, silibinin treatment caused a change in the ratio of Bax/Bcl-2 in a manner that favors apoptosis. Silibinin also induced the cytochrome c release, activation of caspase-3 and caspase-9 and cleavage of PARP. These results suggest that silibinin may exert, at least partly, its anti-cancer effect by inhibiting angiogenesis through induction of endothelial apoptosis via modulation of NF-kappaB, Bcl-2 family and caspases.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Células Endoteliais/fisiologia , NF-kappa B/metabolismo , Silimarina/metabolismo , Citocromos c/metabolismo , DNA/metabolismo , Humanos , NF-kappa B/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Silibina , Proteína X Associada a bcl-2
16.
Oncol Rep ; 10(5): 1467-71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12883725

RESUMO

Overexpression of urokinase plasminogen activator (uPA) is known to correlate closely with tumor cell invasion and metastasis. In gastric cancer, however, the mechanism for induction of uPA remains to be elucidated. In this study, we investigated the intracellular signaling for uPA expression in human gastric carcinoma cells (AGS, SNU-1, SNU-5, and SNU-638). SNU-638 cells which expressed a high level of uPA was found to be highly invasive on a matrigel, while AGS, SNU-1, and SNU-5 cells with low levels of uPA expression were only slightly invasive. SNU-638 cells showed a much higher P38 MAPK activity than the 3 other cell lines. However, there was no significant difference in the activities of P44/42 MAPK (Erk-1/2), JNK and Akt among the above cell lines. Treatment of SNU-638 cells with SB203580, a specific P38 MAPK inhibitor, reduced both the promoter activity and mRNA expression of uPA. Expression of a vector encoding a mutated-type P38alpha MAPK resulted in decrease in the uPA promoter activity in SNU-638 cells. These results suggest that P38 MAPK signaling pathway is important for uPA expression in gastric SNU-638 cells by enhancing the promoter activity of uPA.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neoplasias Gástricas/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Cloranfenicol O-Acetiltransferase/metabolismo , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Laminina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Regiões Promotoras Genéticas , Proteoglicanas/farmacologia , Piridinas/farmacologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...