Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846797

RESUMO

Prednisolone is an anti­inflammatory drug used to treat a number of conditions, including liver disease and cancer. Numerous studies have demonstrated that glucocorticoids such as prednisolone modified by ionizing radiation can promote anticancer activity in cancer cells. To the best of our knowledge, however, the effect of ionizing radiation on prednisolone structure and cancer cells has not yet been identified. The present study created a novel prednisolone derivative using γ­irradiation, and its anticancer properties were investigated in liver cancer cells. The present study confirmed the structure of the new prednisolone derivative using liquid chromatogram­mass spectrometry. MTT assays determined the cytotoxic effects of γ­irradiated (IR)­prednisolone in liver cancer cells. Flow cytometry analysis evaluated apoptosis, mitochondrial membrane potential and cell cycle distribution. Western blotting was used to analyze the proteins associated with apoptosis. The chromatogram profile revealed that IR­prednisolone produced a number of peaks compared with the single peak of the original prednisolone. In contrast to prednisolone, the MTT results showed that IR­prednisolone significantly prevented the growth of liver cancer cells. IR­prednisolone promoted apoptosis and arrested the cell cycle at the G0/G1 stage in Huh7 cells. IR­prednisolone also altered the mitochondrial membrane potential and activated caspase­associated proteins, which activated the intrinsic apoptotic signaling pathway. In conclusion, IR­prednisolone promoted anticancer effects in liver cancer cells via apoptosis activation. The present study demonstrated that IR­prednisolone may be a potential anticancer agent against liver cancer, although specific molecules have yet to be identified.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Hepáticas/metabolismo , Prednisolona/metabolismo , Prednisolona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Hep G2 , Humanos , Neoplasias Hepáticas/prevenção & controle , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Radiação Ionizante , Fase de Repouso do Ciclo Celular/efeitos dos fármacos
2.
Phytomedicine ; 55: 172-178, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668427

RESUMO

BACKGROUND: Centipedegrass extract (CGE) is rich in several polyphenolic compounds including C-glycosylflavonoids, such as maysin and its derivatives, and exerts antioxidant, anti-adipogenic and anticancer effects. However, the effect of CGE on the immune system is unclear. PURPOSE: CGE might inhibit NO production induced by lipopolysaccharide (LPS). In this study, we propose a molecular mechanism for regulation of aberrant immune responses by CGE in LPS-stimulated RAW264.7 cells. STUDY DESIGN: We will preparation of Centipedegrass extract and purify partially in rich of maysin and its derivatives. And examine the effect of the CGE on immune system using LPS-induced RAW cells and animals. METHODS: LPS-induced nitric oxide (NO) and interleukin-6 levels were measured by enzyme-linked immunosorbent assay. The mRNA and protein levels of immune mediators were analyzed by reverse-transcription polymerase chain reaction and immunoblotting, respectively. RESULTS: CGE inhibited LPS-induced NO production in a concentration-dependent manner by suppressing inducible nitric oxide synthase (iNOS) expression in LPS-stimulated cells; this effect was mediated by inhibition of the JAK/STAT pathway. However, CGE did not regulate the expression of other factors, including phosphorylated p38, c-jun N-terminal kinase, or extracellular signal-regulated kinase 1/2. In addition, CGE increased T cells percentage in peripheral blood after oral administration. CONCLUSION: These results indicate that CGE suppresses LPS-induced production of NO and expression of iNOS by directly inhibiting JAK2 kinase activity and enhancing effects on the immune system in mice.


Assuntos
Inibidores de Janus Quinases/farmacologia , Extratos Vegetais/imunologia , Extratos Vegetais/farmacologia , Poaceae/química , Administração Oral , Animais , Relação Dose-Resposta a Droga , Janus Quinase 2/metabolismo , Inibidores de Janus Quinases/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Células RAW 264.7
3.
Oncol Rep ; 41(3): 1837-1850, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30569155

RESUMO

Kenalog is a synthetic glucocorticoid drug used to treat various cancers including ocular and choroidal melanoma. However, the drug achieves rarely sustainable results for patients. To overcome this difficulty, the structure of Kenalog was altered by ionizing radiation (IR) to develop a more effective anticancer agent for treatment of various skin cancers. The anticancer effect of modified Kenalog (Kenalog­IR) was assessed in melanoma cancer cells in vitro. The assessment of mitochondrial functions by MTT assay revealed significant inhibition of melanoma cancer cell viability by Kenalog­IR compared to Kenalog. Moreover, Kenalog­IR­induced apoptotic cell death was associated with the intrinsic mitochondrial pathway by triggering the release of intrinsic apoptosis molecules through activation of caspase­related molecules in concentration and time­dependent manners. Furthermore, it was observed that Kenalog­IR­induced apoptosis was associated with the generation of reactive oxygen species (ROS) with increased G2/M cell cycle arrest. Collectively, Kenalog­IR may be a potential suppressor of skin­related cancer in particular melanoma cancer.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Glucocorticoides/farmacologia , Radiação Ionizante , Triancinolona Acetonida/farmacologia , Antineoplásicos/química , Antineoplásicos/efeitos da radiação , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular , Glucocorticoides/química , Glucocorticoides/efeitos da radiação , Glucocorticoides/uso terapêutico , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Molecular , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Triancinolona Acetonida/química , Triancinolona Acetonida/efeitos da radiação , Triancinolona Acetonida/uso terapêutico
4.
PLoS One ; 13(4): e0194341, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29617386

RESUMO

Dexamethasone (Dex) is widely used in the management of leukemia and lymphoma. While Dex is commonly used for hematological malignancies, the effects of Dex in solid cancer cells remain controversial. To develop a more effective anticancer drug for solid cancers, Dex was modified by ionizing radiation and the anticancer activity of ionizing-radiation-irradiated Dex (Dex-IR) was investigated in human lung cancer cells. Using the MTT assay, the proliferation of non-small cell lung cancer cells was significantly inhibited after treatment with Dex-IR compared with Dex. Furthermore, Dex-IR induced apoptotic cell death and cell cycle arrest of H1650 human lung cancer cells. The invasiveness of H1650 cells was significantly reduced and the matrix metalloproteinase activity was strongly suppressed. These results indicate that Dex-IR acts as a tumor suppressor by both inducing apoptosis and arresting the cell cycle. Although the structure of Dex-IR remains to be determined, our results suggest it may be useful as a novel anticancer agent for the treatment of solid cancers.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Dexametasona/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dexametasona/farmacologia , Dexametasona/efeitos da radiação , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...