Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 930: 175155, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-35863508

ABSTRACT

Long noncoding RNA-Myosin heavy chain associated RNA transcript (LncRNA-MHRT) has been reported to prevent pathological cardiac hypertrophy. However, the underlying inhibition mechanism has not been fully elucidated. Further, whether MHRT inhibits hypertrophy by regulating post-translational modification of certain proteins remains unclear. Therefore, this study aims to find potential role of MHRT in inhibiting cardiac hypertrophy via regulating modification of certain proteins. Here, Angiotensin II (Ang II) -treated neonatal rat cardiomyocytes and transverse aortic constriction (TAC) mice were used to investigate the effect and mechanism of MHRT in cardiac hypertrophy in vitro and in vivo. Moreover, the regulatory effects of MHRT on SUMOylation of NAD-dependent protein deacetylase sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α)/peroxisome proliferator-activated receptor-α (PPARα), specificity protein 1 (SP1)/histone deacetylase 4 (HDAC4) pathway were investigated. Here, we found that MHRT improved heart function by attenuating pathological cardiac hypertrophy in vivo and in vitro. MHRT also promoted the SUMOylation of SIRT1 protein that activated PGC1-α/PPAR-α pathway. Furthermore, MHRT enhanced SUMOylation of SIRT1 by upregulating SP1/HDAC4. Our findings suggested that SUMOylation of SIRT1 could mediate the protective effect of MHRT in cardiac hypertrophy. The new regulatory pathway provides a potential new therapeutic target for pathological cardiac hypertrophy.


Subject(s)
RNA, Long Noncoding , Sirtuin 1 , Animals , Cardiomegaly/pathology , Mice , Myocytes, Cardiac , Myosin Heavy Chains/genetics , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Long Noncoding/metabolism , Rats , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sumoylation
2.
Anticancer Drugs ; 32(9): 939-949, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34001704

ABSTRACT

Exploring drugs that reverse drug resistance and increase the sensitivity of chemotherapy drugs could significantly improve treatment effect of cancer. Our study explored the reversal effect and possible molecular mechanisms of emodin on cisplatin resistance in A549/DDP cells. The IC50 and resistance index of cells were determined by Cell Counting Kit-8 assay. The ability of cell proliferation was evaluated by wound healing assay. Transwell assay was used to detect cell invasion and migration. Apoptosis induction rate was determined by flow cytometry assay and 4',6- diamidino- 2-phenylindole staining. Intracellular concentration was determined by HPLC. Western blot analysis was applied to determine expressions of nuclear factor kappa beta (NF-κB) and its downstream proteins. In this study, we found that the growth inhibitory effect of cisplatin was significantly enhanced by emodin in A549/DDP cells. The combined use of emodin with DDP can effectively promote lung cancer cells apoptosis and inhibit cell migration and invasion. Further investigation indicated that reinforcement effect of emodin and DDP may be associated with inhibition of NF-κB pathway and drug efflux-related proteins such as P-glycoprotein (P-gp), multidrug resistance-associated protein (MRP) and Glutathione S-transferase (GST). The key role of NF-κB was further confirmed by the application of NF-κB inhibitor Ammonium pyrrolidinedithiocarbamate. The intervention of both can significantly increase A549/DDP cell apoptosis and inhibit DDP-induced upregulation of P-gp, MRP and GST. Emodin reverses the cisplatin resistance of tumor cells by down-regulating expression of P-gp, MRP and GST, increasing the intracellular accumulation in A549/DDP cells, and the effect may be associated with the NF-κB pathways.


Subject(s)
Adenocarcinoma of Lung/pathology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Emodin/pharmacology , Lung Neoplasms/pathology , A549 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glutathione Transferase/drug effects , Humans , Inhibitory Concentration 50 , Multidrug Resistance-Associated Proteins/drug effects , NF-kappa B/drug effects , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology
3.
Curr Pharm Des ; 25(43): 4606-4612, 2020.
Article in English | MEDLINE | ID: mdl-31657676

ABSTRACT

BACKGROUND: The human ether-a-go-go-related gene (hERG) potassium channel is the rapidly activating component of cardiac delayed rectifier potassium current (IKr), which is a crucial determinant of cardiac repolarization. The reduction of hERG current is commonly believed to cause Long QT Syndrome (LQTs). Probucol, a cholesterol-lowering drug, induces LQTs by inhibiting the expression of the hERG channel. Unfortunately, there is currently no effective therapeutic method to rescue probucol-induced LQTs. METHODS: Patch-clamp recording techniques were used to detect the action potential duration (APD) and current of hERG. Western blot was performed to measure the expression levels of proteins. RESULTS: In this study, we demonstrated that 1 µM matrine and oxymatrine could rescue the hERG current and hERG surface expression inhibited by probucol. In addition, matrine and oxymatrine significantly shortened the prolonged action potential duration induced by probucol in neonatal cardiac myocytes. We proposed a novel mechanism underlying the probucol induced decrease in the expression of transcription factor Specificity protein 1 (Sp1), which is an established transactivator of the hERG gene. We also demonstrated that matrine and oxymatrine were able to upregulate Sp1 expression which may be one of the possible mechanisms by which matrine and oxymatrine rescued probucol-induced hERG channel deficiency. CONCLUSION: Our current results demonstrate that matrine and oxymatrine could rescue probucol-induced hERG deficiency in vitro, which may lead to potentially effective therapeutic drugs for treating acquired LQT2 by probucol in the future.


Subject(s)
Alkaloids/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Probucol/adverse effects , Quinolizines/pharmacology , Animals , Cell Line , Humans , Patch-Clamp Techniques , Rats, Sprague-Dawley , Matrines
4.
Cell Physiol Biochem ; 41(5): 2004-2015, 2017.
Article in English | MEDLINE | ID: mdl-28420000

ABSTRACT

BACKGROUND/AIMS: Flavonol (-)-epicatechin (EPI) is present in high amounts in cocoa and tea products, and has been shown to exert beneficial effects on the cardiovascular system. However, the precise mechanism of EPI on cardiomyocyte hypertrophy has not yet been determined. In this study, we examined whether EPI could inhibit cardiac hypertrophy. METHODS: We utilised cultured neonatal mouse cardiomyocytes and mice for immunofluorescence, immunochemistry, qRT-PCR, and western blot analyses. RESULTS: 1µM EPI significantly inhibited 1µM angiotensin II (Ang II)-induced increase of cardiomyocyte size, as well as the mRNA and protein levels of ANP, BNP and ß-MHC in vitro. The effects of EPI were accompanied with an up-regulation of SP1 and SIRT1, and were abolished by SP1 inhibition. Up-regulation of SP1 could block Ang II-induced increase in cardiomyocyte size, as well as the mRNA and protein levels of ANP, BNP and ß-MHC, and increase the protein levels of SIRT1 in vitro. Moreover, 1 mg/kg body weight/day EPI significantly inhibited mouse cardiac hypertrophy induced by Ang II, which could be eliminated by SP1 inhibition in vivo. CONCLUSION: Our data indicated that EPI inhibited AngII-induced cardiac hypertrophy by activating the SP1/SIRT1 signaling pathway.


Subject(s)
Angiotensin II/adverse effects , Cardiomegaly , Catechin/pharmacology , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Sp1 Transcription Factor/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomegaly/pathology , Mice , Myocytes, Cardiac/pathology
5.
Drug Des Devel Ther ; 9: 3695-704, 2015.
Article in English | MEDLINE | ID: mdl-26229434

ABSTRACT

The hERG gene encodes the pore-forming α-subunit of the rapidly activating delayed rectifier potassium channel (I Kr), which is important for cardiac repolarization. Reduction of I hERG due to genetic mutations or drug interferences causes long QT syndrome, leading to life-threatening cardiac arrhythmias (torsades de pointes) or sudden death. Probucol is a cholesterol-lowering drug that could reduce hERG current by decreasing plasma membrane hERG protein expression and eventually cause long QT syndrome. Here, we investigated the mechanisms of probucol effects on I hERG and hERG-channel expression. Our data demonstrated that probucol reduces SGK1 expression, known as SGK isoform, in a concentration-dependent manner, resulting in downregulation of phosphorylated E3 ubiquitin ligase Nedd4-2 expression, but not the total level of Nedd4-2. As a result, the hERG protein reduces, due to the enhanced ubiquitination level. On the contrary, carbachol could enhance the phosphorylation level of Nedd4-2 as an alternative to SGK1, and thus rescue the ubiquitin-mediated degradation of hERG channels caused by probucol. These discoveries provide a novel mechanism of probucol-induced hERG-channel deficiency, and imply that carbachol or its analog may serve as potential therapeutic compounds for the handling of probucol cardiotoxicity.


Subject(s)
Anticholesteremic Agents/toxicity , Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/chemically induced , Probucol/toxicity , Anticholesteremic Agents/administration & dosage , Carbachol/pharmacology , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Endosomal Sorting Complexes Required for Transport/metabolism , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Nedd4 Ubiquitin Protein Ligases , Phosphorylation/drug effects , Probucol/administration & dosage , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
Cell Physiol Biochem ; 37(1): 284-96, 2015.
Article in English | MEDLINE | ID: mdl-26303164

ABSTRACT

BACKGROUND/AIMS: Abnormal QT prolongation is the most prominent cardiac electrical disturbance in patients with diabetes mellitus (DM). It is well known that the human ether-ago-go-related gene (hERG) controls the rapid delayed rectifier K+ current (IKr) in cardiac cells. The expression of the hERG channel is severely down-regulated in diabetic hearts, and this down-regulation is a critical contributor to the slowing of repolarization and QT prolongation. However, the intracellular mechanisms underlying the diabetes-induced hERG deficiency remain unknown. METHODS: The expression of the hERG channel was assessed via western blot analysis, and the hERG current was detected with a patch-clamp technique. RESULTS: The results of our study revealed that the expression of the hERG protein and the hERG current were substantially decreased in high-glucose-treated hERG-HEK cells. Moreover, we demonstrated that the high-glucose-mediated damage to the hERG channel depended on the down-regulation of protein levels but not the alteration of channel kinetics. These discoveries indicated that high glucose likely disrupted hERG channel trafficking. From the western blot and immunoprecipitation analyses, we found that high glucose induced trafficking inhibition through an effect on the expression of Hsp90 and its interaction with hERG. Furthermore, the high-glucose-induced inhibition of hERG channel trafficking could activate the unfolded protein response (UPR) by up-regulating the expression levels of activating transcription factor-6 (ATF-6) and the ER chaperone protein calnexin. In addition, we demonstrated that 100 nM insulin up-regulated the expression of the hERG channel and rescued the hERG channel repression caused by high glucose. CONCLUSION: The results of our study provide the first evidence of a high-glucose-induced hERG channel deficiency resulting from the inhibition of channel trafficking. Furthermore, insulin promotes the expression of the hERG channel and ameliorates the high-glucose-induced inhibition of the hERG channel.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Glucose/metabolism , Protein Transport/physiology , Action Potentials/physiology , Arrhythmias, Cardiac/metabolism , Brugada Syndrome , Cardiac Conduction System Disease , Cell Line , Down-Regulation/physiology , Gene Expression/physiology , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Heart Conduction System/abnormalities , Heart Conduction System/metabolism , Humans , Insulin/metabolism , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques/methods , Up-Regulation/physiology
7.
Basic Clin Pharmacol Toxicol ; 116(6): 516-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25395240

ABSTRACT

Arsenic trioxide (As2O3) is used to treat acute pro-myelocytic leukaemia. However, the cardiotoxicity of long QT syndrome restricts its clinical application. Previous studies showed that As2O3 can damage the hERG current via disturbing its trafficking to cellular membrane. Consistent with these findings, in this study, we reported that As2O3 inhibited hERG channel at both protein and mRNA levels and damaged hERG current but did not affect channel kinetics. Further, we demonstrated that As2O3 up-regulated miR-21 and miR-23a expression in hERG-HEK293 cells and neonatal cardiomyocytes. In addition, knock-down of miR-21 by its specific antisense molecules AMO-21 was able to rescue Sp1 and hERG inhibition caused by As2O3. Consistently, phosphorylation of NF-κB, the upstream regulatory factor of miR-21, was significantly up-regulated by As2O3 . This finding revealed that regulation of the NF-κB-miR-21-Sp1 signalling pathway is a novel mechanism for As2O3-induced hERG inhibition. Meanwhile, the expression of Hsp90 and hERG was rescued by transfection with AMO-23a. And the hERG channel inhibition induced by As2O3 was rescued after being transfected with AMO-23a, which may be a molecular mechanism for the role of As2O3 in hERG trafficking deficiency. In brief, our study revealed that miR-21 and miR-23a are involved in As2O3-induced hERG deficiency at transcriptional and transportational levels. This discovery may provide a novel mechanism of As2O3-induced hERG channel deficiency, and these miRNAs may serve as potential therapeutic targets for the handling of As2O3 cardiotoxicity.


Subject(s)
Ether-A-Go-Go Potassium Channels/deficiency , MicroRNAs/biosynthesis , Oxides/toxicity , Potassium Channel Blockers/toxicity , Animals , Animals, Newborn , Arsenic Trioxide , Arsenicals , Ether-A-Go-Go Potassium Channels/drug effects , In Vitro Techniques , Kinetics , MicroRNAs/drug effects , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Transfection , Up-Regulation/drug effects
8.
J Pharmacol Sci ; 123(4): 318-27, 2013.
Article in English | MEDLINE | ID: mdl-24225402

ABSTRACT

We investigated the effects of Ginkgo biloba extract (GBE) and ginkgolide (GLD) on human ether-a-go-go-related gene (hERG)-encoded K(+) channels and its underlying mechanisms in the hERG-HEK293 cell line by determining GBE- and GLD-induced changes in action potential duration (APD), L-type calcium currents (ICa-L), and the intracellular calcium concentration ([Ca(2+)]i) in guinea-pig ventricular myocytes. hERG currents, APD and ICa-L were recorded using the whole-cell patch clamp technique, the [Ca(2+)]i was examined by an immunofluorescence experiment. In the present study, we found that a low concentration of GBE (0.005 mg/ml) increased hERG currents, but the high concentration of GBE (from 0.05 to 0.25 mg/ml) reduced hERG currents. GLD reduced hERG currents in a concentration-dependent manner (from 0.005 to 0.25 mg/ml). Both GBE and GLD altered kinetics of the hERG channel. GBE accelerated the activation of hERG channels without changing the inactivation curve, but reduced the time constant of inactivation; GLD did not shift the activation or the inactivation curve, but only reduced the time constant of inactivation. Both GBE and GLD shortened the APD, inhibited the ICa-L currents, and decreased the [Ca(2+)]i in isolated guinea-pig ventricular myocytes. The results indicate that GBE and GLD can prevent ischemic arrhythmias and have an antiarrhythmic effect potential via inhibition of IKr and ICa-L currents.


Subject(s)
Anti-Arrhythmia Agents , Ether-A-Go-Go Potassium Channels/genetics , Ginkgo biloba , Ginkgolides/pharmacology , Plant Extracts/pharmacology , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/prevention & control , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Guinea Pigs , HEK293 Cells , Heart Ventricles , Humans , Molecular Targeted Therapy , Myocytes, Cardiac/metabolism , Phytotherapy
9.
Cell Physiol Biochem ; 29(3-4): 431-42, 2012.
Article in English | MEDLINE | ID: mdl-22508050

ABSTRACT

Liensinine and neferine, a kind of isoquinoline alkaloid, can antagonize the ventricular arrhythmias. The human ether-a-go-go-related gene (hERG) is involved in repolarization of cardiac action potential. We investigated the effects of liensinine and neferine on the biophysical properties of hERG channel and the underlying structure-activity relationships. The effects of liensinine and neferine were examined on the hERG channels in the stable transfected HEK293 cells using a whole-cell patch clamp technique, western blot analysis and immunofluorescence experiment. The pharmacokinetics and tissue distribution determination of liensinine and neferine in rats were determined by a validated RP-HPLC method. Liensinine and neferine induced decrease of current amplitude in dose-dependent. Liensinine reduced hERG tail current from 70.3±6.3 pA/pF in control group to 56.7±2.8 pA/pF in the 1 µM group, 53.0±2.3 pA/pF (3 µM) and 17.8±0.7 pA/pF (30 µM); the corresponding current densities of neferine-treated cells were 41.9±3.1 pA/pF, 32.3±3.1 pA/pF and 16.2±0.6 pA/pF, respectively. Neferine had binding affinity for the open and inactivated state of hERG channel, liensinine only bound to the open state. The inhibitory effects of liensinine and neferine on hERG current were attenuated in the F656V or Y652A mutant channels. Neferine distributed more quickly than liensinine in rats, which was found to be in higher concentration than liensinine. Both liensinine and neferine had no effect on the generation and expression of hERG channels. In conclusion, neferine is a more potent blocker of hERG channels than liensinine at low concentration (<10 µM), which may be due to higher hydrophobic nature of neferine compared with liensinine. Neferine may be safety even for long-term treatment as an antiarrhythmic drug.


Subject(s)
Benzylisoquinolines/pharmacology , Ether-A-Go-Go Potassium Channels/drug effects , Isoquinolines/pharmacology , Phenols/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/pharmacology , Benzylisoquinolines/pharmacokinetics , Binding Sites , Cell Membrane/metabolism , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Isoquinolines/pharmacokinetics , Membrane Potentials , Patch-Clamp Techniques , Phenols/pharmacokinetics , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/pharmacokinetics , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Time Factors , Tissue Distribution , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...