Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Oxid Med Cell Longev ; 2022: 5184135, 2022.
Article in English | MEDLINE | ID: mdl-35186188

ABSTRACT

OBJECTIVE: To investigate the protective effects and regulatory mechanism of miR-488-3p on doxorubicin-induced cardiotoxicity. METHODS: The C57BL/6 mice and primary cardiomyocytes were used to construct doxorubicin-induced cardiomyocyte injury models in vivo and in vitro. The levels of miR-488-3p and its downstream target genes were analyzed by quantitative real-time PCR. Mouse cardiac function, cell survival, cellular injury-related proteins, and the apoptosis level of cardiomyocytes were analyzed by echocardiography, MTT analysis, Western blotting, and DNA laddering separately. RESULTS: Cardiomyocyte injury caused by a variety of stimuli can lead to the reduction of miR-488-3p level, especially when stimulated with doxorubicin. Doxorubicin led to significant decrease in cardiac function, cell autophagic flux blockage, and apoptosis in vivo and in vitro. The expression of miR-488-3p's target gene, CyclinG1, increased remarkably in the doxorubicin-treated neonatal mouse cardiomyocytes. Overexpression of miR-488-3p inhibited CyclinG1 expression, increased cardiomyocyte viability, and attenuated doxorubicin-induced cardiomyocyte autophagic flux blockage and apoptosis. CONCLUSIONS: miR-488-3p is one of the important protective miRNAs in doxorubicin-induced cardiotoxicity by inhibiting the expression of CyclinG1, which provides insight into the possible clinical application of miR-488-3p/CyclinG1 as therapeutic targets in doxorubicin-induced cardiovascular diseases.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Cyclin G1/antagonists & inhibitors , Doxorubicin/adverse effects , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , Animals , Humans , Male , Mice , Rats
2.
Radiat Res ; 195(4): 378-384, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33543294

ABSTRACT

Radiotherapy plays an important role in the treatment of hepatocellular carcinoma (HCC). Cyclin G1 is a novel member of the cyclin family, and it is abnormally expressed in HCC. In this study we investigated the role of cyclin G1 in the radiotherapy of HCC cells. The expression of cyclin G1 was silenced by transfection of cyclin G1-siRNA into HepG2 cells and Huh7 cells, and the expression of cyclin G1 mRNA and protein was measured by qRT-PCR and Western blot analysis. The proliferation was analyzed using MTT assay, and the radiosensitivity of HCC cells was detected using colony formation assay and a xenograft tumor model. The expression of apoptosis-related proteins (Bcl-2 and Bax) was detected by Western blot analysis, and caspase-3 was detected using fluorimetry. The expression of cyclin G1 mRNA and protein in HepG2/Huh7-cyclin G1-siRNA cells was found to be significantly decreased compared to that in HepG2/Huh7 cells. Silencing the expression of cyclin G1 inhibited the proliferation of HCC cells and enhanced radiosensitivity in HCC cells in vitro and in vivo. Knockdown of cyclin G1 expression significantly decreased Bcl-2 expression, and increased Bax expression and caspase-3 activity in HCC cells. Silencing of cyclin G1 expression enhances the radiosensitivity of HCC cells in vitro and in vivo. The mechanism for this may be related to the regulation of apoptosis-related proteins.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Cyclin G1/genetics , Liver Neoplasms/radiotherapy , Radiation Tolerance/genetics , Animals , Apoptosis/radiation effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Caspase 3/genetics , Cell Line, Tumor , Cyclin G1/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2-Associated X Protein/genetics
3.
Cell Physiol Biochem ; 43(1): 94-107, 2017.
Article in English | MEDLINE | ID: mdl-28848145

ABSTRACT

BACKGROUND/AIMS: CyclinG1 (CycG1) is frequently overexpressed in solid tumors and overexpression of CycG1 promotes cell survival upon paclitaxel exposure by inducing polyploidy. Whether and how CycG1 regulates polyploidization caused by small molecular targeted inhibitors remains unclear. METHODS: Immunohistochemistry and immunoblotting were utilized to examine protein expression. Cell proliferation was measured by ATPlite assay, and cell cycle distribution and apoptosis were measured by flow cytometry and/or DNA fragmentation assays. RESULTS: Overexpression of CycG1 in breast cancer cells caused apoptosis-resistant polyploidy upon treatment with Aurora kinase inhibitor, ZM447439 (ZM). Addition of ABT-263, a small-molecule BH3 mimetic, to ZM, produced a synergistic loss of cell viability with greater sustained tumor growth inhibition in breast cancer cell lines. Decrease of Mcl-1 and increase of NOXA caused by ZM treatment, were responsible for the synergy. Furthermore, CycG1 was highly expressed in Triple-Negative-Breast-Cancer patients treated with paclitaxel and was paralleled by decreased cell survival. CONCLUSION: CycG1 is a crucial factor in ZM-induced polyploidy resistance, and ABT-263/ZM combination hold therapeutic utility in the CycG1-amplified subset of breast cancer and CycG1, thus, is a promising target in breast cancer.


Subject(s)
Cyclin G1/metabolism , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Adult , Aniline Compounds/toxicity , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Benzamides/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin G1/antagonists & inhibitors , Cyclin G1/genetics , Female , Humans , MCF-7 Cells , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Polyploidy , Prognosis , Quinazolines/pharmacology , RNA Interference , Sulfonamides/toxicity , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/mortality , bcl-X Protein/metabolism
4.
Oncotarget ; 5(21): 10607-20, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25431954

ABSTRACT

To successfully target Notch receptors as part of a multidrug anticancer strategy, it will be essential to fully characterize the factors that are modulated by Notch signaling. We recently reported that Notch3 silencing in HCC results in p53 up-regulation in vitro and, therefore, we focused on the mechanisms that associate Notch3 to p53 protein expression. We explored the regulation of p53 by Notch3 signalling in three HCC cell lines HepG2, SNU398 and Hep3B.We found that Notch3 regulates p53 at post-transcriptional level controlling both Cyclin G1 expression and the feed-forward circuit involving p53, miR-221 and MDM2. Moreover, our results were validated in human HCCs and in a rat model of HCC treated with Notch3 siRNAs. Our findings are becoming an exciting area for further in-depth research toward targeted inactivation of Notch3 receptor as a novel therapeutic approach for increasing the drug-sensitivity, and thereby improving the treatment outcome of patients affected by HCC. Indeed, we proved that Notch3 silencing strongly increases the effects of Nutilin-3.With regard to therapeutic implications, Notch3-specific drugs could represent a valuable strategy to limit Notch signaling in the context of hepatocellular carcinoma over-expressing this receptor.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin G1/metabolism , Liver Neoplasms/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Notch/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Cyclin G1/antagonists & inhibitors , Cyclin G1/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver Neoplasms/genetics , Male , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
Alcohol Clin Exp Res ; 37(4): 599-608, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23126531

ABSTRACT

BACKGROUND: Clinical studies demonstrate synergistic liver damage by alcohol and hepatitis C virus (HCV); however, the mechanisms by which alcohol promotes HCV infection remain obscure. The liver-specific microRNA-122 (miR-122) regulates HCV replication and expression of host genes, including Cyclin G1. Here, we hypothesized that alcohol regulates miR-122 expression and thereby modulates HCV RNA replication. METHODS: The J6/JFH/Huh-7.5 model of HCV infection was used in this study. Real-time quantitative polymerase chain reaction, Western blotting, electrophoretic mobility shift assay, and confocal microscopy were used for experimental analysis. RESULTS: We found that acute alcohol exposure (25 mM) significantly increased intracellular HCV RNA as well as miR-122 levels in Huh-7.5 and Huh-7.5/CYP2E1 expressing cells in the presence and absence of J6/JFH-HCV infection. Expression of the miR-122 target, Cyclin G1, was inhibited by alcohol both in J6/JFH-infected and uninfected Huh-7.5 cells. The use of a miR-122 inhibitor increased Cyclin G1 expression and prevented the alcohol-induced increase in HCV RNA and protein levels, suggesting a mechanistic role for alcohol-induced miR122 in HCV replication. We discovered that siRNA-mediated silencing of Cyclin G1 significantly increased intracellular HCV RNA levels compared with controls, suggesting a mechanistic role for Cyclin G1 in HCV replication. Alcohol-induced increase in miR-122 was associated with increased nuclear translocation and DNA binding of the nuclear regulatory factor-κB and could be prevented by NF-κB inhibition. CONCLUSIONS: Our novel data indicate a miR-122-mediated mechanism for alcohol increasing HCV RNA replication. We show for the first time that Cyclin G1, a miR-122 target gene, has regulatory effects on HCV replication and that alcohol increases HCV replication by regulating miR-122 and Cyclin G1.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclin G1/antagonists & inhibitors , Ethanol/pharmacology , Hepacivirus/genetics , Liver Neoplasms/genetics , MicroRNAs/biosynthesis , RNA, Viral/genetics , Virus Replication/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cyclin G1/biosynthesis , Cyclin G1/genetics , Gene Expression Regulation, Viral , Hep G2 Cells , Hepacivirus/drug effects , Humans , Liver Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/physiology , Up-Regulation/drug effects , Up-Regulation/genetics , Virus Replication/drug effects
6.
Cancer Biol Ther ; 9(7): 554-61, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20150764

ABSTRACT

BACKGROUND: microRNA-122 (miR-122) plays an important role in both of hepatic physiology and pathology. Downregulation of miR-122 was reported in human primary hepatocellular carcinoma (HCC) and restoration of miR-122 could suppress the growth of cancer cells. In this study, we presented a novel strategy for cancer therapy based on gene transfer of miR-122 by adenoviral vector. METHODS: We generated a recombinant adenoviral vector expressing miR-122 (Ad-miR122). The miR-122 expression was measured by quantitative Real-Time PCR (qRT-PCR). Cell survival rate was determined by MTT assay. RESULTS: Our data showed that Ad-miR122 could express functional miR-122 in tumor cells at a high level. Infection of tumor cells with Ad-miR122 resulted in inhibition of growth of cancer cells originating from liver (HepG2, Hep3B, Huh7 and PLC/PRF/5), lung (NCI-H460) and uterine cervix (HeLa). This antitumor activity was related to the induction of apoptosis and/or cell cycle arrest in cancer cells. Infection with Ad-miR122 resulted in decreased expression of Bcl-W and CCNG1 in cancer cells. CONCLUSION: The antitumor activity of Ad-miR122 was probably due to the induction of apoptosis and/or cell cycle arrest in cancer cells through inhibiting Bcl-W and/or CCNG1 expression. We concluded that expression of therapeutic microRNA, such as miR-122, via adenoviral vector is a promising strategy for cancer treatment.


Subject(s)
Adenoviridae/genetics , Apoptosis , Cell Cycle , MicroRNAs/physiology , Neoplasms/genetics , Neoplasms/therapy , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cyclin G1/antagonists & inhibitors , Cyclin G1/genetics , Cyclin G1/metabolism , Genetic Therapy , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL