Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Food Chem Toxicol ; 142: 111475, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32522589

RESUMO

Persimmon (Diospyros kaki L.f.) trees are widely cultivated for their edible fruits in Asia. D. kaki leaves are abundant in phytochemicals that have numerous medicinal properties. Hepatocyte growth factor (HGF) and its receptor Met lead to poor prognosis via the promotion of metastasis and chemoresistance in hepatocellular carcinoma (HCC). Therefore, inhibitors targeting the HGF/Met pathway are regarded as promising drugs against HCC. Here, we investigated the effects of D. kaki leaves on HGF-induced epithelial-to-mesenchymal transition (EMT) and stemness traits in HCC. The ethanol extract of D. kaki leaves (EEDK) markedly suppressed HGF-mediated cell migration and invasion through upregulation of CDH1 and downregulation of SNAI1, VIM, MMP1, MMP2, and MMP9. Moreover, EEDK increased the cytotoxicity of sorafenib, which was reduced by HGF, and decreased the expression of the stemness markers KRT19 and CD44. Additionally, we found a clear correlation between stemness and EMT markers in HCC patients. Importantly, EEDK reduced Met activity and attenuated HGF-mediated activation of JNK/c-Jun. Our findings provide new evidence that EEDK can ameliorate HCC with poor prognosis and aggressive phenotype by blocking HGF/Met signaling.


Assuntos
Carcinoma Hepatocelular/patologia , Diospyros/química , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Extratos Vegetais/farmacologia , Folhas de Planta/química , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/metabolismo
2.
Bioorg Med Chem Lett ; 27(4): 1081-1088, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28110870

RESUMO

The epithelial-mesenchymal transition (EMT) is an important cellular process during which polarized epithelial cells become motile mesenchymal cells, which promote cancer metastasis. Ginger, the rhizome of Zingiber officinale, is extensively used in cooking worldwide and also as a traditional medicinal herb with antioxidant, anti-inflammatory and anticancer properties. Several pungent compounds have been identified in ginger, including zingerone, which has anticancer potential. However, the role of zingerone in EMT is unclear. We investigated the synergistic effect of zingerone and its derivative on EMT. Transforming growth factor-beta 1 (TGF-ß1) induces the EMT to promote hepatocellular carcinoma metastasis, including migration and invasion. To understand the repressive role of the combination of zingerone and its derivative (ZD 2) in hepatocellular carcinoma metastasis, we investigated the potential use of each compound of ginger, such as zingerone, ZD 2 and 6-shogaol, or the mixture of zingerone and ZD 2 (ZD 2-1) as inhibitors of TGF-ß1 induced EMT development in SNU182 hepatocellular carcinoma cells in vitro. We show that ZD 2-1, but not zingerone, ZD 2 and 6-shogaol significantly increased expression of the epithelial marker E-cadherin and repressed Snail upregulation and expression of the mesenchymal marker N-cadherin during initiation of the TGF-ß1 induced EMT. In addition, ZD 2-1 inhibited the TGF-ß1 induced increase in cell migration and invasion of SNU182 hepatocellular carcinoma cells. Furthermore, ZD 2-1 significantly inhibited TGF-ß1 regulated matrix metalloproteinase-2/9 and activation of Smad2/3. We also found that ZD 2-1 inhibited nuclear translocation of NF-κB, activation of p42/44 MAPK/AP1 signaling pathway in the TGF-ß1 induced EMT. Our findings provide new evidence that combined treatment with ZD 2, novel zingerone derivative, and zingerone synergistically suppresses hepatocellular carcinoma metastasis in vitro by inhibiting the TGF-ß1 induced EMT.


Assuntos
Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Guaiacol/análogos & derivados , Neoplasias Hepáticas/patologia , Invasividade Neoplásica/prevenção & controle , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Linhagem Celular Tumoral , Sinergismo Farmacológico , Transição Epitelial-Mesenquimal/fisiologia , Guaiacol/química , Guaiacol/farmacologia , Humanos , Fator de Crescimento Transformador beta1/fisiologia
3.
Oncotarget ; 7(35): 56767-56780, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27462923

RESUMO

Delphinidin is a major anthocyanidin compound found in various fruits. It has anti-inflammatory, anti-oxidant, and various other biological activities. In this study, we identified the epigenetic modulators that mediate the apoptotic effect of delphinidin in human prostate cancer cells. We found that treatment of LNCaP cells (a p53 wild-type, human prostate cancer cell line) with delphinidin increased caspase-3, -7, and -8 activity, whereas it decreased histone deacetylase activity. Among class I HDACs, the activity of HDAC3 was specifically inhibited by delphinidin. Moreover, the induction of apoptosis by delphinidin was dependent on caspase-mediated cleavage of HDAC3, which results in the acetylation and stabilization of p53. We also observed that delphinidin potently upregulated pro-apoptotic genes that are positively regulated by p53, and downregulated various anti-apoptotic genes. Taken together, these results show that delphinidin induces p53-mediated apoptosis by suppressing HDAC activity and activating p53 acetylation in human prostate cancer LNCaP cells. Therefore, delphinidin may be useful in the prevention of prostate cancer.


Assuntos
Antocianinas/farmacologia , Apoptose , Histona Desacetilases/metabolismo , Neoplasias da Próstata/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Antineoplásicos Fitogênicos/farmacologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Epigênese Genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/genética , Humanos , Masculino , Extratos Vegetais/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Interferência de RNA , Proteína Supressora de Tumor p53/genética
4.
J Agric Food Chem ; 63(25): 5964-9, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26050847

RESUMO

Although cisplatin can dramatically improve the survival rate in cancer patients, its use is limited by its nephrotoxicity. Previous investigations showed that Panax ginseng contains components that exhibit protective activity against cisplatin-induced nephropathy. The aim of the present study is to investigate the effect of microwave-assisted processing on the protective effect of ginseng and identify ginsenosides that are active against cisplatin-induced kidney damage to evaluate the potential of using ginseng in the management of nephrotoxicity. The LLC-PK1 cell damage by cisplatin was significantly decreased by treatment with microwave-processed ginseng (MG) and ginsenosides Rg3, Rg5, and Rk1. Reduced expression of p53 and c-Jun N-terminal kinase proteins by cisplatin in LLC-PK1 cells was markedly ameliorated after Rg3 and Rg5/Rk1 treatment. Additionally, elevated expression of cleaved caspase-3 was significantly reduced by ginsenosides Rg5, Rk1, and with even greater potency, Rg3. Moreover, MG and its fraction containing active ginsenosides showed protective effects against cisplatin-induced nephropathy in mice. We found that ginsenosides Rg3, Rg5, and Rk1 generated during the heat treatment of ginseng ameliorate renal damage by regulating inflammation and apoptosis. Results of current experiments provide evidence of the renoprotective effects and therapeutic potential of MG and its active ginsenosides, both in vitro and in vivo.


Assuntos
Cisplatino/toxicidade , Ginsenosídeos/administração & dosagem , Nefropatias/prevenção & controle , Panax/química , Substâncias Protetoras/administração & dosagem , Animais , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Ginsenosídeos/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Rim/efeitos dos fármacos , Rim/lesões , Nefropatias/etiologia , Nefropatias/genética , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/farmacologia , Suínos
5.
Bioorg Med Chem Lett ; 25(12): 2559-64, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25959811

RESUMO

Cardamonin (2',4'-dihydroxy-6'-methoxychalcone) is derived from Alpinia katsumadai Hayata (Zingiberaceae), a plant that has been used in Traditional Chinese Medicine for thousands of years. Several anticancer agents have been reported to induce autophagy, which either protects cells or further sensitizes cells to drug treatment. However, the possible autophagic and antiproliferative effects of cardamonin on the human colorectal carcinoma HCT116 cell line are unclear. In the present study, experiments were conducted to determine the effects of cardamonin on cell proliferation, cell cycle distribution, and stimulation of autophagy in cultures of the HCT116 cell line. The results showed that cardamonin inhibited cell proliferation, induced G2/M phase cell cycle arrest, and enhanced autophagy in HCT116 cells. We found evidence that cardamonin-induced autophagic and antiproliferative effects are regulated by the tumor protein p53. We also found that the enhanced activation of c-Jun N-terminal kinase (JNK) by cardamonin was partially regulated by p53 and was critical for cardamonin-induced autophagic and antiproliferative effects in HCT116 cells. These findings suggest that cardamonin or other anticancer agents that increase p53/JNK-dependent stimulation of autophagy could be used to effectively treat patients with colorectal carcinoma.


Assuntos
Antineoplásicos/química , Chalconas/química , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalconas/farmacologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células HCT116 , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Planta Med ; 81(4): 286-91, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25719941

RESUMO

The adverse effects of anticancer drugs can prompt patients to end their treatment despite the efficacy. Cisplatin is a platinum-based molecule widely used to treat various forms of cancer, but frequent and long-term use of cisplatin is limited due to severe nephrotoxicity. In the present study, we investigated the protective effect and mechanism of tetrahydrocurcumin on cisplatin-induced kidney damage, oxidative stress, and inflammation to evaluate its possible use in renal damage. Cisplatin-induced LLC-PK1 renal cell damage was significantly reduced by tetrahydrocurcumin treatment. Additionally, the protective effect of tetrahydrocurcumin on cisplatin-induced oxidative renal damage was investigated in rats. Tetrahydrocurcumin was orally administered every day at a dose of 80 mg/kg body weight for ten days, and a single dose of cisplatin was administered intraperitoneally (7.5 mg/kg body weight) in 0.9 % saline on day four. The creatinine clearance levels, which were markers of renal dysfunction, in cisplatin-treated rats were recovered nearly back to normal levels after administration of tetrahydrocurcumin. Moreover, tetrahydrocurcumin exhibited protective effects against cisplatin-induced oxidative renal damage in rats by inhibiting cyclooxygenase-2 and caspase-3 activation. These results collectively provide therapeutic evidence that tetrahydrocurcumin ameliorates renal damage by regulating inflammation and apoptosis.


Assuntos
Cisplatino/efeitos adversos , Curcuma/química , Curcumina/análogos & derivados , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fitoterapia , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Biomarcadores/metabolismo , Caspase 3/metabolismo , Cisplatino/uso terapêutico , Curcumina/farmacologia , Curcumina/uso terapêutico , Ciclo-Oxigenase 2/metabolismo , Técnicas In Vitro , Rim/metabolismo , Nefropatias/metabolismo , Células LLC-PK1 , Masculino , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ratos Wistar , Suínos
7.
Food Chem Toxicol ; 50(3-4): 1027-35, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155221

RESUMO

Aurentiacin is a chalcone isolated from Syzygium samarangense. In the present study, we examined the anti-inflammatory effects of aurentiacin in lipopolysaccharide (LPS)-stimulated mouse macrophages. Aurentiacin significantly inhibited LPS-induced nitric oxide (NO) production in RAW264.7 cells concomitantly with the suppression of inducible nitric oxide synthase (iNOS) expression. Aurentiacin also reduced the mRNA levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Electrophoretic mobility shift assays (EMSAs) and reporter gene assays indicated that DNA binding and transcriptional activities of nuclear factor-κB (NF-κB)/p65 were decreased by aurentiacin in LPS-stimulated RAW264.7 cells. Moreover, results from chromatin immunoprecipitation (ChIP) assays over the promoter region of the iNOS gene were in agreement with the EMSA results. Pretreatment with aurentiacin prevented the nuclear translocation of p65 by blocking the phosphorylation of I-κB kinase (IKK). Aurentiacin also attenuated the phosphorylation (Ser536) and acetylation (Lys310) of p65 and phosphorylation of MAPKs. In an inflammatory animal model, the intraperitoneal (i.p.) injection of aurentiacin suppressed the release of pro-inflammatory cytokines. Moreover, the level of iNOS protein ex vivo was decreased by aurentiacin similar to the result in vitro. Taken together, these results suggest that aurentiacin shows anti-inflammatory activity related to the inhibition of NF-κB activation.


Assuntos
Chalconas/uso terapêutico , Inflamação/tratamento farmacológico , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos Peritoneais/efeitos dos fármacos , Syzygium/química , Animais , Western Blotting , Linhagem Celular , Chalconas/farmacologia , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Quinase I-kappa B/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-5/genética , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/genética
8.
J Ethnopharmacol ; 134(2): 313-22, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21172413

RESUMO

AIM OF STUDY: Modified Yeoldahanso-tang (MYH) is a Korean herbal formula, containing 10 herbs: Pueraria lobata (Willd.) Ohwi, Angelica tenuissima Nakai, Scutellaria baicalensis Georgi, Platycodon grandiflorum (Jacq), Angelicae Dahurica, Cimicifuga heracleifolia Kom, Raphanus sativa L., Polygala tenuifolia (Willd.), Acorus gramineus Soland. and Dimocarpus longan Lour. The constitutive ratio of the ten herbs is at 6:4:2:1:2:2:2:4:6:6 in dry weight. MYH has been used to treat amnesia, hypochondria and dementia in Korea. In this study, we explored the possibility of using MYH in the prevention and treatment of Parkinson's disease (PD). Specifically, we made an effort to demonstrate the neuroprotective effects of MYH using experimental methods similar to those used in a recent study of PD. MATERIALS AND METHODS: 1-Methyl-4-phenylpyridinium (MPP+) (400µM) was used to induce cytotoxicity in NGF (nerve growth factor)-differentiated PC12 cells. Cell viability was measured using a MTT assay. Induction of autophagy by MYH in NGF-differentiated PC12 cells was measured using an immunoblotting assay with LC3 and beclin 1 antibodies. The proteasomal inhibitor lactacystin (10µM) was used to cause UPS dysfunction in NGF-differentiated PC12 cells. Clearance of aggregated proteins by MYH was measured using an immunoblotting assay with an ubiquitin antibody. 1-Methyl-4-phenyl-1,2,3,6-tetrahydrophenylpyridine (MPTP) (20mg/kg, 4 times i.p.) caused substantia nigra injuries in C57BL/6 mice. Dopamine (DA) neurons were identified using a tyrosine hydroxylase-immunohistochemistry (TH-IHC) assay with a rabbit anti-TH antibody. RESULTS: Our findings indicate that MYH provides protection against MPP+-induced injury in NGF-differentiated PC12 cell. And MYH provides neuroprotection against lactacystin-induced NGF-differentiated PC12 cell death, which effect is partially mediated by autophagy enhancement through enhanced degradation of aggregated proteins. Additionally, in a C57BL/6 mice model with MPTP-induced substantia nigra injuries, MYH inhibits both the loss of TH-positive neurons in the substantia nigra pars compacta (SNpc) and the reduction of the optical density of TH-IR fibers in the striatum (ST). CONCLUSIONS: All of our results indicate that MYH treatment has neuroprotective effects that are partially mediated by autophagy enhancement. MYH may be a promising herbal formula for the prevention and treatment of neurodegenerative diseases, especially PD.


Assuntos
Autofagia/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico , 1-Metil-4-fenilpiridínio , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Encéfalo/patologia , Diferenciação Celular/efeitos dos fármacos , Dopamina/metabolismo , Masculino , Medicina Tradicional Coreana , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Neural/metabolismo , Fármacos Neuroprotetores/farmacologia , Células PC12 , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Extratos Vegetais/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/metabolismo , Coelhos , Ratos , Ubiquitina/metabolismo
9.
J Ethnopharmacol ; 130(1): 13-8, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20347949

RESUMO

AIM OF THE STUDY: To investigate the neuroprotective effect of aqueous extract of modified Wu-Zi-Yan-Zong granule (MWG), a traditional Chinese herbal medicine, against CoCl(2)-induced neurotoxicity in PC12 cells. MATERIALS AND METHODS: Cell viability assay, apoptosis rate assay, ROS detection and mitochondrial membrane potential (MMP) assay were performed. In addition, cytochrome c, caspase-3, PARP and MAPKs were also detected by Western blotting. RESULTS: MWG extract increased viability and suppresses early and middle/late stage apoptosis in a dose-dependent manner in CoCl(2)-induced PC12 cells. Moreover, MWG extract decreased the level of intracellular reactive oxygen species (ROS), increased MMP, regulated Bcl-2 family protein expression (Bcl-2 and Bcl-XL) and inhibited the release of cytochrome c from the mitochondria. In addition, MWG extract attenuated activation of caspase-3 and poly ADP-ribose polymerase (PARP) and inhibited the phosphorylation of ERK, c-Jun NH(2)-terminal kinase (JNK) and p38 MAPKs. CONCLUSIONS: MWG extract exhibited significant neuroprotective effect on PC12 cells, and this effect may be associated with the suppression of ROS generation and inhibition of mitochondria-mediated caspase and MAPK signaling pathways.


Assuntos
Cobalto/farmacologia , Medicamentos de Ervas Chinesas , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Potenciais da Membrana/efeitos dos fármacos , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA