Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Food Nutr Res ; 672023.
Artigo em Inglês | MEDLINE | ID: mdl-37050924

RESUMO

Background: 1,2,3,4,6-Penta-O-galloyl-ß-D-glucose (ß-PGG) is a polyphenol ellagic compound with a variety of pharmacological effects and has an inhibitory effect on lots of cancers. Objective: To explore the antitumor effects and mechanism of 1,2,3,4,6-Penta-O-galloyl-ß-D-glucose on human hepatocellular carcinoma HepG2 cells. Design: A network pharmacology method was first used to predict the possible inhibition of hepatocellular carcinoma growth by 1,2,3,4,6-Penta-O-galloyl-ß-D-glucose (ß-PGG) through the p53 signaling pathway. Next, the Cell Counting Kit (CCK-8) assay was performed to evaluate changes in the survival rate of human hepatocellular carcinoma HepG2 cells treated with different concentrations of the drug; flow cytometry was used to detect changes in cell cycle, apoptosis, mitochondrial membrane potential (MMP) and intracellular Ca2+ concentration; real-time fluorescence quantification and immunoblotting showed that the expression of P53 genes and proteins associated with the p53 signaling pathway was significantly increased by ß-PGG treatment. Reasult: It was found that ß-PGG significantly inhibited survival of HepG2 cells, promoted apoptosis, decreased MMP and intracellular Ca2+ concentration, upregulated P53 gene and protein expression, increased CASP3 expression, and induced apoptosis in HepG2 cells. Conclusion: This study has shown that network pharmacology can accurately predict the target of ß-PGG's anti-hepatocellular carcinoma action. Moreover, it was evident that ß-PGG can induce apoptosis in HepG2 cells by activating the p53 signaling pathway to achieve its anti-hepatocellular carcinoma effect in vitro.

2.
Front Oncol ; 12: 934958, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992839

RESUMO

Background: Gastric cancer (GC) is ranked as the third leading cause of cancer-related mortality worldwide. 1,2,3,4,6-Pentagalloyl-ß-D-glucose (ß-PGG) has various pharmacological activities and has been shown to suppress cancer development. However, the mechanism by which ß-PGG inhibits gastric cancer has not been elucidated. Objective: This study explored the potential targets and mechanism of ß-PGG in GC using the network pharmacology approach combined with in-vitro experiments. Methods: The PharmMapper software was used to predict the potential targets of ß-PGG, and GC-related genes were identified on the GeneCards database. PPI analysis of common genes was performed using the STRING database. The potential regulatory mechanism of ß-PGG in GC was explored through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The binding ability of key genes and target proteins was verified by molecular docking. The effects of ß-PGG on genes and proteins were evaluated using the CCK-8 assay, cell cycle analysis, apoptosis assay, real-time fluorescence quantification polymerase chain reaction (qRT-PCR), and Western blotting. Results: Eight hub genes involved in cell cycle progression and apoptosis were identified. Cancer-related signaling pathways were identified using the Cytoscape tool. Some of those genes were significantly enriched in the p53 signaling pathway. The CCK-8 assay showed that ß-PGG inhibited the proliferation of GC cells. Cell cycle and apoptosis experiments revealed that ß-PGG induced cell cycle arrest and apoptosis of gastric cancer cells. qRT-PCR and Western blot analysis showed that ß-PGG inhibited ß-PGG cells by modulating the p53 signaling pathway. Conclusion: In the present study, the targets and mechanism of ß-PGG in gastric cancer were explored. The results indicate that ß-PGG can be used to develop treatments for GC.

3.
Front Med (Lausanne) ; 8: 747263, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34977060

RESUMO

Objective: Sepsis is life threatening and leads to complex inflammation in patients with immunocompromised conditions, such as cancer, and receiving immunosuppressants for autoimmune diseases and organ transplant recipients. Increasing evidence has shown that RNA-Sequencing (RNA-Seq) can be used to define subendotype in patients with sepsis; therefore, we aim to use RNA-Seq to identify transcriptomic features among immunocompromised patients with sepsis. Methods: We enrolled patients who were admitted to medical intensive care units (ICUs) for sepsis at a tertiary referral centre in central Taiwan. Whole blood on day-1 and day-8 was obtained for RNA-Seq. We used Gene Set Enrichment Analysis (GSEA) to identify the enriched pathway of day-8/day-1 differentially expressed genes and MiXCR to determine the diversity of T cell repertoire. Results: A total of 18 immunocompromised subjects with sepsis and 18 sequential organ failure assessment (SOFA) score-matched immunocompetent control subjects were enrolled. The ventilator-day, ICU-stay, and hospital-day were similar between the two groups, whereas the hospital mortality was higher in immunocompromised patients than those in immunocompetent patients (50.0 vs. 5.6%, p < 0.01). We found that the top day-8/day-1 upregulated genes in the immunocompetent group were mainly innate immunity and inflammation relevant genes, namely, PRSS33, HDC, ALOX15, FCER1A, and OLR1, whereas a blunted day-8/day-1 dynamic transcriptome was found among immunocompromised patients with septic. Functional pathway analyses of day-8/day-1 differentially expressed genes identified the upregulated functional biogenesis and T cell-associated pathways in immunocompetent patients recovered from sepsis, whereas merely downregulated metabolism-associated pathways were found in immunocompromised patients with septic. Moreover, we used MiXCR to identify a higher diversity of T cell receptor (TCR) in immunocompetent patients both on day-1 and on day-8 than those in immunocompromised patients. Conclusions: Using RNA-Seq, we found compromised T cell function, altered metabolic signalling, and decreased T cell diversity among immunocompromised patients with septic, and more mechanistic studies are warranted to elucidate the underlying mechanism.

4.
Oncotarget ; 8(14): 23905-23926, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28108741

RESUMO

A variety of studies indicated that inorganic arsenic and its methylated metabolites have paradoxical effects, namely, carcinogenic and anticancer effects. Epidemiological studies have shown that long term exposure to arsenic can increase the risk of cancers of lung, skin or bladder in man, which is probably associated with the arsenic metabolism. In fact, the enzymatic conversion of inorganic arsenic by Arsenic (+3 oxidation state) methyltransferase (AS3MT) to mono- and dimethylated arsenic species has long been considered as a major route for detoxification. However, several studies have also indicated that biomethylation of inorganic arsenic, particularly the production of trivalent methylated metabolites, is a process that activates arsenic as a toxin and a carcinogen. On the other hand, arsenic trioxide (As2O3) has recently been recognized as one of the most effective drugs for the treatment of APL. However, elaboration of the cytotoxic mechanisms of arsenic and its methylated metabolites in eradicating cancer is sorely lacking. To provide a deeper understanding of the toxicity and carcinogenicity along with them use of arsenic in chemotherapy, caution is required considering the poor understanding of its various mechanisms of exerting toxicity. Thereby, in this review, we have focused on arsenic metabolic pathway, the roles of the methylated arsenic metabolites in toxicity and in the therapeutic efficacy for the treatments of solid tumors, APL and/or non-APL malignancies.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/metabolismo , Arsenicais/farmacologia , Animais , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Intoxicação por Arsênico/etiologia , Intoxicação por Arsênico/metabolismo , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico
5.
Int J Mol Sci ; 18(2)2017 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-28125064

RESUMO

Arsenic trioxide (As2O3) has recently become one of the most effective drugs for treatment of patient with acute promyelocytic leukemia (APL), and its molecular mechanism has also been largely investigated. However, it has been reported that As2O3 resistant patients are frequently found in relapsed APL after consolidation therapy, which is due to the point mutations in B-box type 2 motifs of promyelocytic leukemia (PML) gene. In the present study, we for the first time establish whether organic arsenic species phenylarsine oxide (PAO) could induce the mutant PML-IV (A216V) protein solubility changes and degradation. Here, three different PML protein variants (i.e., PML-IV, PML-V and mutant PML-A216V) were overexpressed in HEK293T cells and then exposed to PAO in time- and dose-dependent manners. Interestingly, PAO is found to have potential effect on induction of mutant PML-IV (A216V) protein solubility changes and degradation, but no appreciable effects were found following exposure to high concentrations of iAsIII, dimethylarsinous acid (DMAIII) and adriamycin (doxorubicin), even though they cause cell death. Our current data strongly indicate that PAO has good effects on the mutant PML protein solubility changes, and it may be helpful for improving the therapeutic strategies for arsenic-resistant APL treatments in the near future.


Assuntos
Arsenicais/farmacologia , Arsenitos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Mutação , Proteína da Leucemia Promielocítica/química , Proteína da Leucemia Promielocítica/genética , Expressão Gênica , Células HEK293 , Humanos , Processamento de Proteína Pós-Traducional , Proteólise , Solubilidade/efeitos dos fármacos
6.
Phytomedicine ; 23(14): 1706-1715, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27912872

RESUMO

BACKGROUND: Typhonium blumei Nicolson & Sivadasan (Araceae) is a traditional Chinese medicinal herb possessing detumescent, detoxifying, and anti-inflammatory activities. It is used in Taiwan as a folk medicine to treat cancer and inflammatory diseases. Typhonium blumei is usually not distinguished from Typhonium roxburghii Schott and they are commonly used interchangeably. PURPOSE: To evaluate and compare the anti-allergic and anti-inflammatory properties of T. blumei and T. roxburghii, their composition profiles and molecular basis of the anti-allergic effect. METHODS: The methanolic plant extracts were partitioned with different solvents to obtain the nonpolar fractions. The anti-allergic activity of the nonpolar fractions was assessed by A23187- and antigen-induced degranulation assays using RBL-2H3 mast cells. Several molecular targets were investigated: FcεRI receptor expression by flow cytometry, calcium influx by live cells imaging fluorescent microscopy, cytokines mRNA expression by RT-PCR, and protein expression by Western blotting. The anti-inflammatory activity was evaluated using superoxide anion and elastase release assays in human neutrophils. TLC, NMR and GC-MS analyses were conducted to evaluate the chemical composition of the fractions. RESULTS: The nonpolar fractions of both Typhonium species showed potent inhibitory activity in A23187-induced degranulation assay in RBL-2H3 cells. They also inhibited superoxide production and elastase release in human neutrophils. T. blumei nonpolar fractions inhibited antigen-induced ß-hexosaminidase and histamine release. The nonpolar fractions of T. blumei significantly inhibited calcium influx upon activation with either A23187 or an antigen. The fractions did not affect FcεRI receptor expression, mRNA level of IL-4 and MCP-1 cytokine production or MAPK proteins expression, but did suppress the calcium signaling pathway via PI3K/PLCγ2. The active fractions were rich in fatty acids with palmitic, linoleic and α-linolenic acids identified as the major fatty acids in both plants. The content of omega-3 unsaturated fatty acids was higher in T. roxburghii nonpolar fractions compared to T. blumei. CONCLUSION: Both species possess potent anti-allergic and anti-inflammatory activities. The inhibition of degranulation in mast cells was attributed to calcium influx modulation. The obtained results support the traditional use of T. blumei in the treatment of inflammatory diseases as well as its substitution with T. roxburghii.


Assuntos
Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Araceae/química , Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Ácidos Graxos/farmacologia , Animais , Antialérgicos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Citocinas/metabolismo , Medicamentos de Ervas Chinesas/uso terapêutico , Ácidos Graxos/análise , Ácidos Graxos/uso terapêutico , Liberação de Histamina/efeitos dos fármacos , Humanos , Interleucina-4/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Fitoterapia , Ratos , Receptores de IgE/metabolismo , Transdução de Sinais , beta-N-Acetil-Hexosaminidases/metabolismo
7.
Chem Res Toxicol ; 28(3): 351-3, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25166275

RESUMO

Arsenic is a known carcinogen; however, there is no information on the toxic effects of inorganic arsenic and its intermediate metabolites, monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)), during the differentiation of embryonic stem (ES) cells into cardiomyocytes. The objective of this study was to evaluate the effects of arsenic compounds on ES cell differentiation into cardiomyocytes in vitro and to predict the associated toxic effects. Although iAs(III) is known to be toxic, here we found that iAs(III) and DMA(III) did not influence ES cellular differentiation, whereas MMA(III) inhibited ES cell differentiation into cardiomyocytes, suggesting that MMA(III) has adverse effects on embryonic stem cells.


Assuntos
Arsênio/toxicidade , Ácido Cacodílico/análogos & derivados , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Miócitos Cardíacos/citologia , Compostos Organometálicos/toxicidade , Animais , Ácido Cacodílico/toxicidade , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Camundongos , Células-Tronco Embrionárias Murinas/citologia
8.
Metallomics ; 7(1): 165-73, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25412289

RESUMO

Arsenic trioxide has been successfully used for the treatment of patients with acute promyelocytic leukemia (APL) worldwide. Recently, it has also been further developed to treat solid tumors in clinical trials. However, the therapeutic effects on malignant tumors appeared to be unsatisfactory, as these cells exhibited resistance towards arsenic. In this study, we explored new therapeutic strategies for treatment of human breast cancer MCF-7 cells based on arsenic metabolites. The MCF-7 cells were exposed to three arsenic species, namely, inorganic arsenite (iAs(III)) and its intermediate metabolites monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)) either alone or in combination with cryptotanshinone (CPT) to establish their anticancer effects against MCF-7 cells. Surprisingly, MCF-7 cells were shown to be resistant to both iAs(III) and CPT when used alone; however, they were shown to be relatively sensitive to treatment when exposed to MMA(III) and DMA(III) alone. Conversely, the combination of MMA(III) with CPT showed significantly enhanced anticancer effects on MCF-7 cells at low doses, but no appreciable effect was observed upon exposure to the other two arsenic species with CPT. In addition, remarkable redistribution of pro-apoptosis related proteins Bax and Bak was observed in the mitochondria, together with activation of poly(ADP-ribose) polymerase (PARP) and caspase-9 after exposure to the combination of MMA(III) with CPT. Furthermore, we clearly found that induction of apoptosis in MCF-7 cells was predominantly triggered by endoplasmic reticulum (ER) stress after exposure to the combination of MMA(III) with CPT.


Assuntos
Apoptose/efeitos dos fármacos , Arsênio/toxicidade , Neoplasias da Mama/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fenantrenos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Feminino , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA