Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 41(2): 829-838, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535464

ABSTRACT

In cancer research, autophagy acts as a double­edged sword: it increases cell viability or induces cell apoptosis depending upon the cell context and functional status. Recent studies have shown that adenosine (Ado) has cytotoxic effects in many tumors. However, the role of autophagy in Ado­induced apoptosis is still poorly understood. In the present study, Ado­induced apoptotic death and autophagy in hepatoblastoma HepG2 cells was investigated and the relationship between autophagy and apoptosis was identified. In the present study, it was demonstrated that Ado inhibited HepG2 cell growth in a time­ and concentration­dependent manner and activated endoplasmic reticulum (ER) stress, as indicated by G0/G1 cell cycle arrest, the increased mRNA and protein levels of GRP78/BiP, PERK, ATF4, CHOP, cleaved caspase­3, cytochrome c and the loss of mitochon-drial membrane potential (ΔΨm). Ado also induced autophagic flux, revealed by the increased expression of the autophagy marker microtubule­associated protein 1 light chain 3­II (LC3­II), Beclin­1, autophagosomes, and the degradation of p62, as revealed by western blot analysis and macrophage­derived chemokine (MDC) staining. Blocking autophagy using LY294002 notably entrenched Ado­induced growth inhibition and cell apoptosis, as demonstrated with the increased expression of cytochrome c and p62, and the decreased expression of LC3­II. Conversely, the autophagy inducer rapamycin alleviated Ado­induced apoptosis and markedly increased the ΔΨm. Moreover, knockdown of AMPK with si­AMPK partially abolished Ado­induced ULK1 activation and mTOR inhibition, and thus reinforced CHOP expression and Ado­induced apoptosis. These results indicated that Ado­induced ER stress resulted in apoptosis and autophagy concurrently. The AMPK/mTOR/ULK1 signaling pathway played a protective role in the apoptotic procession. Inhibition of autophagy may effectively enhance the anticancer effect of Ado in human hepatoblastoma HepG2 cells.


Subject(s)
Adenosine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Hepatoblastoma/drug therapy , Liver Neoplasms/drug therapy , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine/therapeutic use , Autophagy-Related Protein-1 Homolog/metabolism , Cell Survival/drug effects , Chromones/pharmacology , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Hep G2 Cells , Hepatoblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/pathology , Morpholines/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
2.
Int J Oncol ; 48(1): 421-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26647875

ABSTRACT

Long non-coding RNA MEG3 is suggested to function as a tumor suppressor. However, the activation mechanism of MEG3 is still not well understood and data are not available on its role under adenosine-induced apoptosis. In this study, HepG2 cells were treated with adenosine or 5-Aza­cdR. Methylation status of MEG3 promoter was detected by methylation specific PCR (MSP) and MEG3 expression was determined by qRT-PCR. PcDNA3.1-MEG3 recombinant plasmid was constructed and transfected to hepatoma HepG2 and Huh7 cells. Cell growth, morphological changes, cell-cycle distribution and apoptosis were analyzed by MTT assay, fluorescence microscopy and flow cytometry. The mRNA and protein expression levels were detected by qRT-PCR and western blot analysis. MEG3 binding proteins were screened by the improved MS2 biotin tagged RNA affinity purification method. The co-expression network of MEG3 was generated by GO analysis and ILF3 was identified as MEG3 binding protein by RNA pulldown and western blot analysis. Both adenosine and 5-Aza-CdR increased MEG3 mRNA expression and the CpG island of MEG3 gene in HepG2 cells was typical hypermethylation. Ectopic expression of MEG3 inhibited hepatoma cell growth in a time-dependent manner, resulted in cell cycle arrest and induced apoptosis. Ectopic expression of MEG3 increased p53, caspase-3 mRNA and protein levels, decreased MDM2 and cyclin D1 mRNA and protein levels, as well as ILF3 protein expression in HepG2 cells. These findings are the first to identify that adenosine increases MEG3 expression by inhibition of DNA methylation and its antitumor effects is involved in MEG3 activation. ILF3 may participate in the anticancer regulation of MEG3 by interacting with MEG3.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/biosynthesis , Adenosine/administration & dosage , Azacitidine/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , CpG Islands/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , RNA, Long Noncoding/genetics
3.
Int J Mol Sci ; 15(1): 525-44, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24394318

ABSTRACT

Our previous studies show that adenosine-induced apoptosis is involved in endoplasmic reticulum stress in HepG2 cells. In this study, we have investigated whether knockdown of GRP78 by short hairpin RNA (shRNA) increases the cytotoxic effects of adenosine in HepG2 cells. The adenovirus vector-delivered shRNA targeting GRP78 (Ad-shGRP78) was constructed and transfected into HepG2 cells. RT-PCR assay was used to determine RNA interference efficiency. Effects of knockdown of GRP78 on adenosine-induced cell viabilities, cell-cycle distribution and apoptosis, as well as relative protein expressions were determined by flow cytometry and/or Western blot analysis. The intracellular Ca2+ concentration was detected by laser scanning confocal microscope. Mitochondrial membrane potential (ΔΨm) was measured by a fluorospectrophotometer. The results revealed that GRP78 mRNA was significantly downregulated by Ad-shGRP78 transfection. Knockdown of GRP78 enhanced HepG2 cell sensitivity to adenosine by modulating G0/G1 arrest and stimulating Bax, Bak, m-calpain, caspase-4 and CHOP protein levels. Knockdown of GRP78 worsened cytosolic Ca2+ overload and ΔΨm loss. Knockdown of caspase-4 by shRNA decreased caspase-3 mRNA expression and cell apoptosis. These findings indicate that GRP 78 plays a protective role in ER stress-induced apoptosis and show that the combination of chemotherapy drug and RNA interference adenoviruses provides a new treatment strategy against malignant tumors.


Subject(s)
Adenosine/toxicity , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Heat-Shock Proteins/metabolism , RNA, Small Interfering/metabolism , Adenoviridae/genetics , Antineoplastic Agents/metabolism , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caspases, Initiator/metabolism , Down-Regulation/drug effects , Endoplasmic Reticulum Chaperone BiP , G1 Phase Cell Cycle Checkpoints/drug effects , Genetic Vectors/metabolism , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , RNA Interference , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription Factor CHOP/metabolism
4.
Int J Mol Med ; 30(4): 797-804, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859272

ABSTRACT

Apoptosis plays a critical role in the development and homeostasis of multicellular organisms, and endoplasmic reticulum stress (ERS) is one of the intrinsic apoptosis pathways. Previous studies have shown that adenosine induces apoptosis in several cancer cell lines. However, the molecular mechanism remains poorly understood. In this study, we explored whether adenosine triggers apoptosis of EC109 esophageal carcinoma (EC) cells by ERS. The MTT assay was used to determine cell proliferation; cell cycle detection (FCM) and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay were performed to determine cell apoptosis. The subcellular distribution and expression of the ERS-related proteins GRP78, cleaved caspase-3, cleaved caspase-4, CHOP and NF-κB p65 were detected by western blot techniques. NF-κB activation was measured by electrophoretic mobility shift assay (EMSA). The MTT assay demonstrated that adenosine inhibited EC109 cell proliferation in a dose- and time-dependent manner. FCM and TUNEL assay verified that adenosine caused an apoptotic peak in cell cycle arrest and a higher percentage of apoptotic cells. Western blot analysis confirmed that the expression of GRP78, cleaved caspase-4, CHOP, NF-κB p65 and cleaved caspase-3 were upregulated in a dose-dependent manner after adenosine treatment. EMSA revealed that adenosine activated NF-κB p65. This is the first demonstration that adenosine inhibits cell proliferation, increases GRP78 and NF-κB p65 expression and induces apoptosis by CHOP and caspase-4 pathways. The ERS pathway is involved in adenosine-induced apoptosis in EC109 cells.


Subject(s)
Adenosine/metabolism , Apoptosis , Endoplasmic Reticulum Stress , Esophageal Neoplasms/metabolism , Caspase 3/metabolism , Caspases, Initiator/metabolism , Cell Line, Tumor , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , NF-kappa B/metabolism , Transcription Factor CHOP/metabolism
5.
Oncol Rep ; 26(1): 73-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21479362

ABSTRACT

Endoplasmic reticulum stress (ERS)-mediated cell apoptosis has been implicated in the development of multiple diseases such as cancer, neurodegenerative diseases and ischemic reperfusion damage. Previous studies have demonstrated the adenosine-induced apoptosis in several tumor cell lines. However, the role of ERS in adenosine-induced human hepatoma HepG2 cell apoptosis remains unclear. The present study was designed to determine whether ERS is involved in adenosine-induced HepG2 cell apoptosis. The MTT assay was used to determine proliferation, and DAPI staining of cell nuclei was performed to determine cell apoptosis. The translocation of CHOP and caspase-3 was observed by immunofluorescence analysis, and the protein expression of CHOP, caspase-4 and caspase-3 was detected by Western blotting. The MTT assay demonstrated that adenosine inhibited HepG2 cell proliferation in a dose-dependent manner. DAPI staining of cell nuclei and cell cycle analysis verified cell apoptosis. The immunofluorescence assay demonstrated that adenosine induced the translocation of CHOP and of caspase-3 from the cytoplasm to the nucleus. Western blotting confirmed that CHOP, caspase-4 and caspase-3 were up-regulated in HepG2 cells after treatment with adenosine. However, JNK protein expression was not altered. These results show that ERS is involved in the adenosine-induced HepG2 cell apoptosis.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Caspase 3/metabolism , Caspases, Initiator/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/metabolism , Liver Neoplasms/metabolism , Transcription Factor CHOP/metabolism , Adenosine/metabolism , Adenosine/pharmacology , Cell Proliferation , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Hep G2 Cells , Humans , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology
6.
Biochem Cell Biol ; 88(4): 705-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20651843

ABSTRACT

Adenosine can exhibit cytotoxic activity in vivo and in vitro, though its mechanisms are still uncertain. In this study, we investigated the adenosine-mediated apoptotic signaling pathway and the role of NF-kappaB in human hepatocellular carcinoma HepG2 cells. HepG2 cells were treated with different concentrations of adenosine for 12-48 h, and the effect of adenosine on cell proliferation was evaluated by MTT assay. The cytotoxicity of adenosine alone or in combination with an NF-kappaB inhibitor, pyrrolidine dithiocarbamate (PDTC), was also evaluated by MTT assay and the mode of cell death was detected by Hoechst 33342 staining. Cell cycle progress was performed by flow cytometry with PI staining. The protein expressions of Bcl-2, p53, NF-kappaB subunit p65, and caspase-3 were assayed by Western blot. Caspase-3 activity was measured by spectrophotomteric assay. The results showed that adenosine significantly reduced the viability of HepG2 cells in a dose- and time-dependent manner, with IC 50 (24 and 48 h) of 2.52 and 1.89 mmol x L(-1), respectively. The apoptotic index (percentage of sub-G1 phase) of HepG2 cells in adenosine treatment alone for 12 and 24 h or in combination with PDTC were 8.30%, 22.32% and 20.18%, 30.89%, respectively. All of them were higher than that in the control group (0.81%, p < 0.01). The characteristic changes of cell apoptosis (chromatin condensation and sub-G1 peak) were observed under fluorescent microscopy and flow cytometry. We also found that the apoptotic process triggered by adenosine was involved in G0-G1 cell-cycle arrest, enhanced the activity of caspase-3, upregulated p53 and NF-kappaB p65 expression, and downregulated Bcl-2 expression. Inhibition of NF-kappaB by PDTC decreased NF-kappaB p65 expression, enhanced cell apoptosis ratio, and increased caspase-3 activity. NF-kappaB may play an anti-apoptosis role in adenosine-induced HepG2 cytotoxicity.


Subject(s)
Adenosine/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , NF-kappa B/physiology , Adenosine/metabolism , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Extracellular Space/drug effects , Extracellular Space/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Thiocarbamates/pharmacology
7.
Acta Pharmacol Sin ; 27(4): 477-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16539849

ABSTRACT

AIM: To investigate effects of adenosine on cell proliferation and apoptosis in human HepG2 cells. METHODS: HepG2 cells were incubated in the presence of adenosine (0.1-5 mmol/L) for 12-48 h, and the effect of adenosine on cell proliferation was evaluated by using 3-(4,5-dimethyl-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Hoechst 33342 fluorescent staining, dUTP-fluorescein isothiocyanate (FITC) fluorescence and flow cytometric analysis techniques were used to observe cell apoptosis. The effects of adenosine receptor (A1, A2a, A3 and nonspecific receptor) antagonists (8-cpt, DMPX, MRS1191, and theophylline) and an adenosine transporter protein inhibitor (dipyridamole) on adenosine-induced cell apoptosis were observed. Mitochondrial membrane potential was analyzed using DePsipher fluorescent staining, and caspase activity was detected using a Fluorometric assay kit and a fluorescence microplate reader. RESULTS: Adenosine significantly reduced cell viability in a dose- and time-dependent manner. The cytotoxicity of adenosine was related to the induction of cell apoptosis. Four adenosine receptor antagonists had no effect on cell apoptosis. However, dipyridamole significantly reduced the percentage of adenosine-induced apoptotic cells from 27.3% to 7.1% (P<0.05). At 48 h after treatment, 3 mmol/L adenosine increased caspase-3 activity 3.5-fold; dipyridamole markedly decreased caspase-3 activity 1.6-fold, and decreased apoptotic cell numbers. When HepG2 cells were treated with 3 mmol/L adenosine, mitochondrial membrane potential and the activity of caspase-8 or -9 remained unchanged. CONCLUSION: Our results suggest that adenosine-induced apoptosis in HepG2 cells is related to intracellular events rather than cell surface receptors, and that a caspase-3 cascade activation is required, which is not mediated via a mitochondrial pathway.


Subject(s)
Adenosine/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/metabolism , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dipyridamole/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Liver Neoplasms/metabolism , Membrane Potentials/drug effects , Mitochondria, Liver/physiology , Theophylline/analogs & derivatives , Theophylline/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...