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.
Acta Pharmacol Sin ; 41(1): 22-33, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31431734

ABSTRACT

Long non-coding RNAs (lncRNAs) have been identified as essential mediators in neurological dysfunction. Our previous study shows that berberine (BBR) hampers the nuclear-to-cytosolic translocation of high-mobility group box 1 (HMGB1) in the process of poststroke inflammation. In this study, we explored the role of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (Malat1) in the process of BBR-induced inhibition of HMGB1 in ischemic brain. Before the 60-min MCAO surgery, the mice were pretreated with BBR (50 mg· kg-1 per day, ig) for 14 days or ICV injected with specific lentiviral vector or shRNA. We showed that MCAO caused marked increase in the expression Malat1 and HMGB1 in the ipsilateral cortex, which was significantly attenuated by pretreatment with BBR. Knockdown of Malat1 attenuated the inflammatory injury after brain ischemia, whereas overexpression of Malat1 exacerbated ischemic brain inflammation. Overexpression of Malat1 also reversed BBR-induced reduction of HMGB1 and proinflammatory cytokines. The above results suggested a potential correlation between Malat1 and stroke inflammation. Based on informatics analysis we predicted that HMGB1 was a direct downstream target of miR-181c-5p, whereas Malat1 acted as a competitive endogenous RNA (ceRNA) for miR-181c-5p targeted the 3'-UTR of HMGB1 to promote inflammation after ischemic stroke. Knockdown of Malat1 significantly decreased HMGB1 level, which could be abrogated by transfection with miR-181c-5p inhibitors. Taken together, our results demonstrate for the first time that Malat1/miR-181c-5p/HMGB1 axis may be a key pathway of BBR-induced antiinflammation effects in stroke, and they may provide a novel avenue for targeted therapy.


Subject(s)
Berberine/pharmacology , HMGB1 Protein/antagonists & inhibitors , Inflammation/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Administration, Oral , Animals , Berberine/administration & dosage , Cells, Cultured , HEK293 Cells , HMGB1 Protein/metabolism , Humans , In Situ Hybridization, Fluorescence , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Optical Imaging , RNA, Long Noncoding/genetics
2.
Arzneimittelforschung ; 61(11): 622-30, 2011.
Article in English | MEDLINE | ID: mdl-22232851

ABSTRACT

oancer falls to respond to chemotherapy by acquiring multidrug resistance in over 90% of patients. A previous study revealed that multidrug resistance modulator HZ08 had great multidrug resistance reversal effect in vitro and in vivo. It could enhance adriamycin (doxorubicin) induced intrinsic apoptosis pathway and rectify cell cycle and some apoptosis related proteins in human breast resistant cancer MCF-7/ADM cells. This study detected Rh123 accumulation to assess the effect of HZ08 on P-glycoprotein function in human chronic leukaemia cell line K562/A02. Moreover, mitochondria membrane potential, cytochrome c release and caspase-3 activity were analyzed for HZ08 treatment with or without vincristine. Since pretreatment with HZ08 could also reverse the multidrug resistance to vincristine in K562/A02 cells, the individual influence of HZ08 was further detected on apoptotic regulator like Bcl-2, Bax, p53, cell cycle checkpoints and proliferation regulatory factors like survivin, hTERT, c-Myc, c-Fos, c-Jun. Finally, it revealed that HZ08 increased vincristine induced activation in intrinsic apoptosis pathway by inhibition of P-gp mediated efflux. In addition, the outstanding reversal effect of HZ08 should also attribute to its individual effect on apoptosis and proliferation related regulatory factors. It renders HZ08 possibility of application in pretreatment to reverse multidrug resistance while avoiding unexpected drug interactions and accumulative toxicity.


Subject(s)
Apoptosis/drug effects , Isoquinolines/pharmacology , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Survival , Cytochromes c/metabolism , Drug Resistance, Neoplasm/drug effects , Fluorescent Dyes , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , K562 Cells , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Rhodamine 123 , Vincristine/pharmacology
3.
Eur J Pharmacol ; 647(1-3): 21-30, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20816813

ABSTRACT

In early studies, it was demonstrated that R-HZ08, S-HZ08 and the racemate had strong reverse efficacy of multidrug resistance in vitro and in vivo (Yan et al., 2008b). The effect was supposed to have direct interaction with multidrug resistance-associated protein (MRP1) in MCF-7/ADM and P-glycoprotein in K562/A02. According to our latest study, we found HZ08 could enhance chemotherapy induced apoptosis by synergistic action on reactive oxygen species generation, GSH depletion, mitochondrial membrane potential depolarization, cytochrome c release and caspase activation. Moreover, the potential selective effect of HZ08 on resistant cells suggested that HZ08 have specific targets for resistance reversal via apoptosis regulation. Therefore, we traced individual influence of HZ08, not only on apoptosis pathway per se but also on apoptosis related intracellular regulation systems. Then we found HZ08 could increase cells in G(0)/G(1) phase and regulate apoptosis related proteins (Bcl-2, Bax) as well as upstream functional molecules (c-Myc and c-Fos), which are usually abnormal in malignancy and responsible for multidrug resistance in MCF-7/ADM. Thereby, chemotherapy induced apoptosis was promoted. R-HZ08 showed better effect than S-HZ08 or the racemate did in most of targets above. Furthermore, HZ08 did not change the concentration of intracellular Ca(2+) which means it would not have side effect as verapamil does. Considering multidrug resistance is multifactorial, HZ08, especially R-HZ08, which could sensitize apoptosis by multiple improvements of upstream malignant characters, will be a promising drug to enhance the effect of chemotherapy in the treatment of multidrug resistant tumor.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Drug Resistance, Multiple/drug effects , Isoquinolines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Therapy, Combination , Female , Humans , Isoquinolines/chemistry , Membrane Potential, Mitochondrial/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/pharmacology , Reactive Oxygen Species/metabolism , Verapamil/metabolism , Verapamil/pharmacology
4.
Acta Pharmacol Sin ; 31(3): 273-80, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20140005

ABSTRACT

AIM: To investigate the protective effects of ginsenoside Rb(3), a triterpenoid saponin isolated from the leaves of Panax notoginseng, on ischemic and reperfusion injury model of PC12 cells and elucidate the related mechanisms. METHODS: PC12 cells exposed to oxygen and glucose deprivation (OGD) and restoration (OGD-Rep) were used as an in vitro model of ischemia and reperfusion. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) leakage were used to evaluate the protective effects of ginsenoside Rb(3). Cellular apoptosis and mitochondrial membrane potential (MMP) were analyzed using flow cytometry. Intracellular calcium ion concentration ([Ca(2+)](i)) was detected using fluorophotometer system. Caspase-3, -8, and -9 activities were measured using assay kits with an ELISA reader. Western blotting assay was used to evaluate the release of cytochrome c and expression of caspase-3, Bcl-2 and Bax proteins. RESULTS: It was shown that ginsenoside Rb(3) (0.1-10 micromol/L) significantly increased cell viability and inhibited LDH release in a dose-dependent manner on the ischemic model. In addition, ginsenoside Rb(3) also significantly inhibited ischemic injury-induced apoptosis, [Ca(2+)](i) elevation, and decrease of MMP. Meanwhile, pretreatment with ginsenoside Rb(3) significantly induced an increase of Bcl-2 protein expression and a decrease of cytosolic cytochrome c, cleaved-caspase 3 and Bax protein expression, the caspase-3, -8, and -9 activity were also inhibited. CONCLUSION: The results indicated that ginsenoside Rb(3) could markedly protected OGD-Rep induced ischemic injury and the mechanisms maybe related to its suppression of the intracellular Ca(2+) elevation and inhibition of apoptosis and caspase activity. Ginsenoside Rb(3) could be a promising candidate in the development of a novel class of anti-ischemic agent.


Subject(s)
Cytoprotection , Ginsenosides/pharmacology , Glucose/metabolism , Oxygen/metabolism , Reperfusion Injury/drug therapy , Animals , Calcium/metabolism , Caspases/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Ginsenosides/therapeutic use , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , PC12 Cells , Panax notoginseng/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...