Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Med ; 10(19): 6795-6806, 2021 10.
Article in English | MEDLINE | ID: mdl-34427987

ABSTRACT

The incidence and mortality of hepatocellular carcinoma (HCC) are gradually increasing during the past years. Recently, some studies have reported that malic enzyme (ME) plays an important role in cancer development, while the involvement of ME2 in HCC remains still undetermined. Here, we demonstrated that ME2 played an oncogenic role in HCC. ME2 was overexpressed in HCC tissues. TCGA database showed that the ME2 transcript level was inversely associated with the survival of HCC patients. Loss-of-function and gain-of-function assays showed that ME2 promoted HCC cell growth and migration. Furthermore, the xenografted tumorigenesis of MHCC97H cells was retarded by ME2 knockdown. ME2 silencing also suppressed the cell cycle process and induced apoptosis. Mechanistically, ME2 potentiated triglyceride synthesis, inhibition of which suppressed the proliferation and migration. We propose that ME2 promotes HCC progression by increasing triglyceride production.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Malate Dehydrogenase/adverse effects , Triglycerides/adverse effects , Animals , Carcinogenesis , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Humans , Liver Neoplasms/mortality , Male , Mice , Mice, Nude , Survival Analysis
2.
Exp Ther Med ; 15(1): 67-74, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29399057

ABSTRACT

Cerebral ischemic injury causes severe brain damage and remains one of the leading causes of morbidity and mortality worldwide. Members of the microRNA-29 (miR-29) family are involved in regulating the process of ischemia and may be developed as biomarkers to diagnose and treat cerebral ischemia. The role of miR-29b in cerebral ischemia injury remains poorly understood. The purpose of the present study was to investigate whether miR-29b overexpression suppressed cerebral ischemic injury and to explore its underlying mechanism of action. The results demonstrated that levels of miR-29b in N2a neuroblastoma cells decreased following oxygen and glucose deprivation/reperfusion (OGD/R) treatment. Transfection with miR-29b mimics significantly increased cell viability, decreased lactate dehydrogenase (LDH) leakage, inhibited apoptosis by decreasing morphological changes occurring in the nuclei and reduced caspase-3 activity in OGD/R-treated N2a cells. Conversely, miR-29b inhibitors enhanced OGD/R-induced cytotoxicity and apoptosis. In addition, the miR-29b mimics blocked the increase in Bax and p53 expression and decreased Bcl-2 expression in OGD/R-treated N2a cells, whereas miR-29b inhibitors exacerbated the changes in the expression of these apoptosis-associated proteins caused by OGD/R. p53 knockdown using p53 small interfering RNA decreased cell viability and increased LDH leakage, reversing the improvements that the miR-29b mimics induced in damaged cells. Taken together, the results of the present study demonstrated that miR-29b attenuates ischemic injury by negatively regulating the p53-dependent apoptosis pathway and may therefore be a novel potential therapeutic target for treating ischemic stroke.

3.
Med Sci Monit ; 23: 6107-6113, 2017 Dec 25.
Article in English | MEDLINE | ID: mdl-29276937

ABSTRACT

BACKGROUND Hepatocellular carcinoma (HCC) is the most important cause of cancer-related deaths worldwide. Pirfenidone is an orally available small molecule with therapeutic potential for fibrotic diseases. MATERIAL AND METHODS In this study, we analyzed the effects of different pirfenidone concentrations on the proliferation of HepG2 HCC cells using Cell Counting Kit-8 (CCK-8) and colony formation assays. Flow cytometry was performed to measure the apoptotic effects of pirfenidone on HepG2 cells. Western blot analysis was performed to detect the expression of ß-catenin and p-ß-catenin. RESULTS Pirfenidone inhibited proliferation and promoted HepG2 cell apoptosis. In addition, Western blot results indicated that pirfenidone suppressed b-catenin expression in HepG2 cells. To assess the mechanism, we treated HepG2 cells with pirfenidone, and pirfenidone plus the ß-catenin activator, SB-216763. The results revealed that SB-216763 accelerated proliferation and inhibited apoptosis in HepG2 cells treated with pirfenidone. Western blot results showed that SB-216763 upregulated ß-catenin expression in HepG2 cells treated with pirfenidone. CONCLUSIONS In conclusions, pirfenidone may be a potential drug for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Pyridones/pharmacology , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Pyridones/metabolism , Signal Transduction/drug effects , Wnt Signaling Pathway/physiology , beta Catenin/drug effects , beta Catenin/metabolism
4.
Mol Med Rep ; 12(1): 309-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738437

ABSTRACT

Pirfenidone (esbiret) is an established anti-fibrotic and anti-inflammatory drug used to treat idiopathic pulmonary fibrosis. In the present study, the dose-dependent effects of pirfenidone on the cell cycle, proliferation and expression of heat shock protein (HSP)-47 and collagen type I in a cultured rat hepatic stellate cell line (HSC-T6) were investigated. Following pirfenidone treatment, cell proliferation was determined using the cell counting kit-8 assay and the cell cycle was measured using flow cytometry. HSP-47 expression was estimated using western blot analysis and collagen type I mRNA was assessed using reverse transcription quantitative polymerase chain reaction. Pirfenidone induced significant dose-dependent inhibition of proliferation in HSC-T6 cells. Cell viability was unaffected by treatment with pirfenidone (0, 10 or 100 µM) for 24 and 72 h. However, after 24 h, HSC-T6 cells exhibited dose-dependent decreases in HSP-47 protein and collagen I mRNA levels. In conclusion, pirfenidone inhibited HSC-T6 cell proliferation, arrested the cell cycle and reduced the expression of HSP-47 and collagen type I, indicating that pirfenidone may be a promising drug in the treatment of liver fibrosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Proliferation/drug effects , Collagen Type I/genetics , Down-Regulation/drug effects , HSP47 Heat-Shock Proteins/genetics , Pyridones/pharmacology , Animals , Blotting, Western , Cell Line , Cell Survival/drug effects , Collagen Type I/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , HSP47 Heat-Shock Proteins/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...