Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Drug Des ; 101(6): 1367-1381, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36798994

RESUMO

Indole-3-carbinol is produced from the cruciferous vegetables and broadly investigated for their various biological effects in in-vitro and in-vivo aspects. However, the anticancer activity of I3C and its molecular mechanisms have not been investigated in human adeno gastro carcinoma (AGS) cells. In our study of AGS cells, nuclear condensation was observed by 4',6-diamidino-2-phenylindole (DAPI) staining, cell death was confirmed by a cell viability assay, and fragmented DNA was observed at the IC50 dose by a DNA fragmentation assay. Apoptosis was evaluated by the qPCR technique. Treatment of the AGS cells with I3C at different concentrations has drastically decreased cell proliferation and differentiation. By releasing cytochrome-c from mitochondria in the intrinsic pathway, I3C prevents the multiplication of AGS cells and initiates apoptosis. The WST-1 assay result showed that I3C treatment against AGS cells had considerably reduced the viability of the cells. Furthermore, RT-qPCR showed the fold change among the expressed proteins compared with reference gene ß-actin. Molecular docking revealed that I3C showed a strong binding affinity for the apoptotic protein 3DCY. The results show the caspase group of proteins contribute to the core of apoptotic machinery. I3C and its metabolites target a variety of components of cell-cycle control via distinct signaling pathways in light of the rapid development of tumors and oncogenesis. The translational significance of I3C and its metabolites in cancer is highlighted by their wide range of antitumor activity and low toxicity. Furthermore, the novel prodrug I3C, which has overlapping underlying mechanisms, could encourage new strategies to decrease oncogenesis.


Assuntos
Anticarcinógenos , Humanos , Anticarcinógenos/farmacologia , Linhagem Celular Tumoral , Simulação de Acoplamento Molecular , Apoptose , Mitocôndrias/metabolismo , Carcinogênese/metabolismo
2.
J Immunol Res ; 2020: 1731457, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33299895

RESUMO

Tumor microenvironment components dictate the growth and progression of various cancers. Tumor-associated macrophages are the most predominant cells in TME and play a major role in cancer invasiveness. Gastric cancer is one of the most common cancers in Asia, and recently, various cases of resistance to fluorouracil treatment have been reported. In this study, we investigated the role of alternatively activated macrophages in the resistance of AGS gastric cancer cells to fluorouracil. THP-1 cells were polarized using recombinant human IL-4, then were cocultured with AGS cells treated with fluorouracil. Cell viability, Western blot, immunofluorescence, and cell invasion were performed for this investigation. Our results demonstrated that polarized macrophages initiated the survival of treated AGS cells and induced the resistance in AGS by upregulating the expression of integrin ß3, focal adhesion protein (FAK), and cofilin proteins. These results reveal that integrin ß3, focal adhesion protein (FAK), and cofilin proteins are potential targets for the improvement of fluorouracil efficacy in gastric cancer treatment.


Assuntos
Fatores de Despolimerização de Actina/genética , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Integrina beta3/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Polaridade Celular , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/imunologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Integrina beta3/metabolismo , Ativação de Macrófagos/imunologia , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
3.
Colloids Surf B Biointerfaces ; 183: 110455, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31493630

RESUMO

Green chemistry is beneficial for the production of eco-friendly and stable nanoparticles using biological agents. The present study was performed to explore the potential of the marine bacterium Paracoccus haeundaensis BC74171T for the extracellular synthesis of gold nanoparticles (AuNPs). Cell-free supernatant-mediated AuNPs were characterized by different techniques and analyzed for their antioxidant activity and antiproliferative effect on normal and cancer cells. Visual observations indicated the formation of AuNPs by the development of a ruby red color and were confirmed by a UV-vis absorbance peak at about 535 nm. The synthesized AuNPs were spherical in shape and had an average size of 20.93 ± 3.46 nm, as determined by transmission electron microscopy and a dynamic light scattering particle size analyzer, respectively. From Fourier transform infrared spectroscopy, the interaction of functional groups was determined and the presence of biomaterial on the AuNP surface was confirmed. Concentration-dependent antioxidant activity of AuNPs was observed by the 2,2-diphenyl-1-picrylhydrazyl method. The AuNPs synthesized do not show growth inhibition on HaCaT and HEK293 normal cells, while they show concentration-dependent growth inhibition in the case of A549 and AGS cancer cells. Thereby, this study proves that AuNP synthesis using P. haeundaensis is a facile method and that the AuNPs synthesized are non-toxic to human cells, which indicates that they can be useful in biomedical applications.


Assuntos
Antibacterianos/farmacologia , Antioxidantes/farmacologia , Proliferação de Células/efeitos dos fármacos , Ouro/química , Nanopartículas Metálicas/química , Paracoccus/metabolismo , Células A549 , Antibacterianos/química , Antioxidantes/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Células HEK293 , Humanos , Testes de Sensibilidade Microbiana , Água do Mar/microbiologia
4.
Int J Biol Macromol ; 137: 844-852, 2019 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-31295479

RESUMO

Fucoidan is a sulfated hetero-polysaccharide, found in cell-wall composition of brown algae. Recent studies have reported the role of fucoidan in the induction of Endoplasmic Reticulum (ER) stress-related cell death in cancer cells but the mechanism of action of fucoidan in cervical HeLa cells is not well-known. The purpose of this study was to investigate if fucoidan induces HeLa cells death through ER-stress-related cell death and G1 phase arrest. 200-600 µg/ml concentrations of fucoidan inhibited the proliferation of HeLa cells after 48 h of treatment while investigated normal cell lines (HaCaT: Keratinocytes and HEK-293: embryonic kidney) were not affected. The exposure of HeLa cells to these concentrations of fucoidan induced phosphorylation of ER stress sensors followed by upregulation of Bip/GRP78, CHOP expression which triggered a buildup of malfolded proteins in ER, therefore, initiating unfolded protein response (UPR) mechanism. In addition, intracellular calcium levels were elevated following the treatment suggesting that this contributed to the ER stress-induced apoptosis. Fucoidan treatment caused G1phase arrest by inducting CDKIs and inhibiting CDKs and Cyclins as well as apoptosis by activating the mitochondrial-dependent pathway in HeLa cells. We demonstrated that Fucoidan inhibits HeLa cells proliferation by inducing apoptosis, G1 phase arrest, ER stress, and mitochondrial-mediated apoptosis.


Assuntos
Antineoplásicos/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Polissacarídeos/farmacologia , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Homeostase/efeitos dos fármacos , Humanos
5.
Int J Oncol ; 53(5): 2300-2308, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226597

RESUMO

In recent decades, various bioactive compounds from plants have been investigated for their potential use in the treatment of diseases in humans. Aster incisus extract (AIE) is the extract of a common plant that is mostly found in Asia. It has traditionally been used for medicinal purposes in South Korea. In this study, we evaluated the potential anticancer effects of a methanolic extract of Aster incisus in a normal human cell line (HaCaT keratinocytes) and in 4 different types of human cancer cell lines (A549, lung cancer; Hep3B, liver cancer; MDA­MB­231, breast cancer; and AGS, gastric cancer). The HaCaT, A549, Hep3B, MDA­MB­231 and AGS cells were treated with various concentrations of AIE and following treatment, cell survival was evaluated. Additional analyses, such as WST-1 assay, western blot analysis, DAPI staining, flow cytometry, immunofluorescence staining and wound healing assay were performed to elucidate the mechanisms and pathways involved in the cell death induced by AIE. Treatment with AIE induced morphological changes and considerably reduced the viability of the both normal and cancer cell lines. Further analysis of the AGS gastric cancer cells revealed that AIE led to the induction of apoptosis and a high accumulation of cells in the G1 cell phase following treatment with AIE in a dose-dependent manner. The results also revealed that AIE successfully suppressed the migration of the AIE-treated AGS cells. The results of western blot analysis indicated that AIE increased the expression of pro-apoptotic proteins, particularly Bid, Bad, Bak, cytochrome c, apoptosis inducing factor (AIF), cleaved caspase­3, -8 and -9 and cleaved poly(ADP-ribose) polymerase (PARP). Additionally, AIE decreased the expression of the anti-apoptotic proteins, Bcl-2 and Bcl-xL. On the whole, the findings of this study demonstrate that AIE induces apoptosis through the activation of the caspase­dependent pathway mediated by the mitochondrial pathway and by arresting the cell cycle in AGS cells.


Assuntos
Adenocarcinoma/tratamento farmacológico , Aster/química , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Medicina Tradicional Coreana/métodos , Extratos Vegetais/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Adenocarcinoma/epidemiologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Incidência , Metanol/química , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , República da Coreia/epidemiologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/epidemiologia
6.
Mediators Inflamm ; 2018: 4675204, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30622433

RESUMO

Aster incisus is a common flower found in almost all regions of South Korea. In the current study, we investigated the potential antioxidant and anti-inflammatory properties of the Aster incisus methanol extract in LPS-stimulated RAW 264.7 cells. We analyzed the phytochemicals contained in the extract by GC-MS. GC-MS results showed that the Aster incisus extract contains 9 known compounds. Later on, DPPH assay, WST-1 assay, nitric oxide (NO) assay, Western blot, and RT-PCR were conducted to investigate the anti-inflammatory effects of the extract. Our WST-1 assay results revealed that Aster incisus did not affect the viability of all tested cell lines up to a concentration of 200 µg/ml; therefore, lower concentrations (50 µg/ml and 150 µg/ml) were used for further assays. Aster incisus scavenged DPPH and inhibited the production of NO. Aster incisus also reduced significantly the production of inflammation-related enzymes (iNOS, Cox-2) and cytokines (TNFα, IL-1ß, and IL-6) and the gene expression of the proinflammatory cytokines. Additionally, further Western blot results indicated that Aster incisus inhibited the expression of p-PI3K, p-IκBα, p-p65 NF-κB, p-ERK1/2, p-SAPK/JNK, and p-Akt. Our results demonstrated that Aster incisus suppressed the expression of the inflammation mediators through the regulation of NF-κB, MAPK, and Akt pathways.


Assuntos
Aster/química , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Camundongos , Células RAW 264.7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...