Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(5): 610-618, 2022 May 28.
Article in English, Chinese | MEDLINE | ID: mdl-35753731

ABSTRACT

OBJECTIVES: The risk of arrhythmia increases in diabetic patients. However, the effects of hyperglycemia and insulin therapy on the electrophysiological properties of human cardiomyocytes remain unclear. This study is to explore the effects of high glucose and insulin on the electrophysiological properties and arrhythmias of cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs). METHODS: Immunofluorescent staining and flow cytometry were used to analyze the purity of hiPSC-CMs generated from human skin fibroblasts of a healthy donor. The hiPSC-CMs were divided into 3 group (treated with normal medium, high glucose and insulin for 4 days): a control group (NM group, containing 5 mmol/L glucose), a high glucose group (HG group, containing 15 mmol/L glucose), and a high glucose combined with insulin (HG+INS group, containing 15 mmol/L glucose+100 mg/L insulin). Electrophysiological changes of hiPSC-CMs were detected by microelectrode array (MEA) before or after treatment with glucose and insulin, including beating rate (BR), field potential duration (FPD) (similar to QT interval in ECG), FPDc (FPD corrected by BR), spike amplitude and conduction velocity (CV). Effects of sotalol on electrophysiological properties and arrhythmias of hiPSC-CMs were also evaluated. RESULTS: The expression of cardiac-specific marker of cardiac troponin T was high in the hiPSC-CMs. The purity of hiPSC-CMs was 99.06%. Compared with the NM group, BR was increased by (9.14±0.8)% in the HG group (P<0.01). After treatment with high glucose, FPD was prolonged from (460.4±9.0) ms to (587.6±23.7) ms in the HG group, while it was prolonged from (462.5±14.5) ms to (512.6±17.6) ms in the NM group. Compared with the NM group, FPD of hiPSC-CMs was prolonged by (16.8±1.4)% in the HG group (P<0.01). The FPDc of hiPSC-CMs was prolonged from (389.1±13.7) ms to (478.3±31.5) ms in the HG group, and that was prolonged from (387.7±21.6) ms to (422.6±32.9) ms in the NM group. Compared with the NM group, the FPDc of hiPSC-CMs was prolonged by (13.9±1.3)% in HG group (P<0.01). The spike amplitude and CV remained unchanged between the HG group and the NM group (P>0.05). Ten µmol/L of sotalol can induce significant arrhythmias from all wells in the HG group. After treatment with insulin and high glucose, compared with the HG group, BR was increased by (8.3±0.5)% in the HG+INS group (P<0.05). The FPD was prolonged from (463.4±9.7) ms to (532.6±12.8) ms in the HG+INS group, while it was prolonged from (460.4±9.0) ms to (587.6±23.7) ms in the HG group. Compared with the HG group, the FPD of hiPSC-CMs was shortened by (12.7±1.9)% in the HG+INS group (P<0.01). The FPDc of hiPSC-CMs was prolonged from (387.4±4.1) ms to (422.4±10.0) ms in the HG+INS group, and that was prolonged from (384.8±4.0) ms to (476.3±11.5) ms in HG group. Compared with the HG group, the FPDc of hiPSC-CMs was shortened by (14.7±1.1)% in HG group (P<0.01). After the insulin treatment, the spike amplitude of hiPSC-CMs was increased from (3.12±0.46) mV to (4.35±0.64) mV in the HG+INS group, while it was enhanced from (3.06±0.35) mV to (3.33±0.41) mV in the HG group. The spike amplitude of hiPSC-CMs was increased by (30.8±3.7)% in the HG+INS group compared with that in the HG group (P<0.05). The CV in the HG+INS group was increased from (0.23±0.08) mm/ms to (0.32±0.08) mm/ms after insulin treatment, which was increased from (0.21±0.04) mm/ms to (0.30±0.07) mm/ms in the HG group, but there was no significant difference in CV between the HG+INS group and the HG group (P>0.05). The induction experiment showed that 10 µmol/L of sotalol could prolong the FPDc of hiPSC-CMs by (78.9±11.6)% in the HG+INS group, but no arrhythmia was induced in each well. CONCLUSIONS: High glucose can induce FPD/FPDc of hiPSC-CMs prolongation and increase the risk of arrhythmia induced by drugs. Insulin can reduce the FPD/FPDc prolongation and the risk of induced arrhythmia by high glucose.These results are important to understand the electrophysiological changes of the myocardium in diabetic patients and the impact of insulin therapy on its electrophysiology. Further study on the mechanism may provide new ideas and methods for the treatment of acquired and even inherited long QT syndrome.


Subject(s)
Induced Pluripotent Stem Cells , Arrhythmias, Cardiac/metabolism , Cells, Cultured , Glucose/metabolism , Glucose/pharmacology , Humans , Induced Pluripotent Stem Cells/physiology , Insulin/pharmacology , Myocytes, Cardiac , Sotalol/adverse effects , Sotalol/metabolism
2.
BMC Cardiovasc Disord ; 20(1): 441, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032539

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

3.
BMC Cardiovasc Disord ; 20(1): 409, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32912149

ABSTRACT

BACKGROUND: Cardiac rupture (CR) is a fatal complication of ST-elevation myocardial infarction (STEMI) with its incidence markedly declined in the recent decades. However, clinical features of CR patients now and the effect of reperfusion therapy to CR remain unclear. We investigated the clinical features of CR in STEMI patients and the effect of reperfusion therapy to CR in mice. METHODS: Two studies were conducted. In clinical study, data of 1456 STEMI patients admitted to the First Hospital, Xi'an Jiaotong University during 2015.12. ~ 2018.12. were analyzed. In experimental study, 83 male C57BL/6 mice were operated to induce MI. Of them, 39 mice were permanent MI (group-1), and remaining mice received reperfusion after 1 h ischemia (21 mice, group-2) or 4 h ischemia (23 mice, group-3). All operated mice were monitored up to day-10. Animals were inspected three times daily for the incidence of death and autopsy was done for all mice found died to determine the cause of death. RESULTS: CR was diagnosed in 40 patients: free-wall rupture in 17, ventricular septal rupture in 20, and combined locations in 3 cases. CR presented in 19 patients at admission and diagnosed in another 21 patients during 1 ~ 14 days post-STEMI, giving an in-hospital incidence of 1.4%. The mortality of CR patients was high during hospitalization accounting for 39% of total in-hospital death. By multivariate logistic regression analysis, older age, peak CK-MB and peak hs-CRP were independent predictors of CR post-STEMI. In mice with non-reperfused MI, 17 animals (43.6%) died of CR that occurred during 3-6 days post-MI. In MI mice received early or delayed reperfusion, all mice survived to the end of experiment except one mouse died of acute heart failure. CONCLUSION: CR remains as a major cause of in-hospital death in STEMI patients. CR patients are characterized of being elderly, having larger infarct and more server inflammation. Experimentally, reperfusion post-MI prevented CR.


Subject(s)
Heart Rupture, Post-Infarction/etiology , ST Elevation Myocardial Infarction/complications , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Heart Rupture, Post-Infarction/diagnosis , Heart Rupture, Post-Infarction/mortality , Heart Rupture, Post-Infarction/prevention & control , Hospital Mortality , Humans , Male , Mice, Inbred C57BL , Middle Aged , Myocardial Reperfusion , Retrospective Studies , Risk Assessment , Risk Factors , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/mortality , ST Elevation Myocardial Infarction/therapy , Time Factors
5.
Exp Ther Med ; 19(2): 1388-1392, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32010313

ABSTRACT

Protective role of lncRNA HOXB-AS3 in doxorubicin (DOX)-induced cardiotoxicity and its mechanism were studied. Viability of PC and H9c2 cells treated with different doses of DOX was determined through CCK-8 assay. Relative level of HOXB-AS3 in DOX-treated cardiomyocytes was detected. Regulatory effect of HOXB-AS3 on the proliferative ability of DOX-treated cardiomyocytes was assessed. Through dual-luciferase reporter gene assay, the binding relationship between HOXB-AS3 and miRNA-875-3p was verified. Rescue experiments were conducted to explore the role of HOXB-AS3/miRNA-875-3p in influencing the proliferation of DOX-treated cardiomyocytes. The proliferative ability of cardiomyocytes was dose-dependently downregulated after DOX treatment. Relative level of HOXB-AS3 was upregulated in DOX-treated cardiomyocytes. Silence of HOXB-AS3 in cardiomyocytes undergoing DOX treatment markedly elevated their proliferative ability. miRNA-875-3p was the direct target of HOXB-AS3. Knockdown of miRNA-875-3p reversed the role of HOXB-AS3 in regulating the proliferative ability of cardiomyocytes. HOXB-AS3 protects DOX-induced suppression in the proliferation of cardiomyocytes through targeting and downregulating miRNA-875-3p.

7.
Toxicol Sci ; 167(2): 360-374, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30247688

ABSTRACT

Numerous drugs have the potential to prolong the QT interval and may cause accidental cardiac arrest (torsades de pointes [TdP]). Women are at a higher risk than men for experiencing drug-induced TdP. Due to the lack of appropriate tools, few studies have investigated whether genetic differences between men and women have any effects on drug-induced proarrhythmia. Sex hormones are believed to play a predominant role in the induction of TdP. Recently, progress in induced pluripotent stem cell (iPSC) technologies has made it possible to utilize human iPSC-derived cardiomyocytes (hiPSC-CMs) to investigate the influence of both genetics and sex hormones on cardiac ion channel gene expression and cardiomyocyte function. In this study, we investigated genetic and hormonal effects on sex differences of drug-induced QT prolongation and TdP with hiPSC-CMs from healthy male and female donors. We found that despite batch variations in beating rates and field potential durations (FPD), female-derived hiPSC-CMs showed steeper slopes of FPD to interspike interval ratios and were more sensitive to IKr blocker-induced FPD prolongation. 17ß-estradiol increased FPD and 5α-dihydrotestosterone shortened FPD, but the addition of sex hormones had limited effect on the responses of hiPSC-CMs to IKr blockades. The differential expression of KCNE1 gene and reduced repolarization reserve in female-derived hiPSC-CMs compared with male-derived hiPSC-CMs may partially explain why females are more susceptible to proarrhythmias. Human iPSC-CMs can be a useful new model to study mechanisms of sex differences in cardiomyocyte repolarization processes and aid in the prediction of drug-induced proarrhythmias in both men and women.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/etiology , Induced Pluripotent Stem Cells/drug effects , Long QT Syndrome/chemically induced , Models, Biological , Myocytes, Cardiac/drug effects , Sex Characteristics , Torsades de Pointes/chemically induced , Action Potentials/drug effects , Cells, Cultured , Drug-Related Side Effects and Adverse Reactions/metabolism , Female , Healthy Volunteers , Humans , Induced Pluripotent Stem Cells/metabolism , Ion Channels/antagonists & inhibitors , Long QT Syndrome/metabolism , Male , Myocytes, Cardiac/metabolism , Torsades de Pointes/metabolism
8.
J Cardiovasc Pharmacol ; 72(1): 11-18, 2018 07.
Article in English | MEDLINE | ID: mdl-29672418

ABSTRACT

Severe postoperative complications can affect cardiac surgery patients. Levosimendan is a novel calcium sensitizer commonly administered after cardiac surgery. However, the patient benefits are controversial. PubMed, Embase, and the Cochrane library were systematically searched for randomized controlled trials comparing levosimendan with control in adult cardiac surgery patients. Twenty-five studies (3247 patients) were included. Pooled data indicated that levosimendan reduced mortality after cardiac surgery [odds ratio (OR) 0.63, 95% confidence interval (CI): 0.47-0.84, P = 0.001]. However, this reduction was restricted to patients with low (<50%) left ventricular ejection fraction (OR 0.49, 95% CI: 0.35-0.70, P = 0.0001). It significantly reduced the incidence of postoperative acute kidney injury (OR 0.55, 95% CI: 0.41-0.74, P < 0.0001) and renal replacement therapy use (OR 0.56, 95% CI: 0.39-0.80, P = 0.002). Moreover, levosimendan significantly shortened the duration of the intensive care unit stay (weighted mean differences -0.49 day, 95% CI: -0.75 to -0.24, P = 0.0002) and mechanical ventilation use (weighted mean differences -2.30 hours, 95% CI: -3.76 to -0.84, P = 0.002). In conclusion, levosimendan reduced the mortality in patients with low left ventricular ejection fraction and decreased the incidence of acute renal injury and renal replacement therapy use. In addition, it shortened the duration of the intensive care unit stay and mechanical ventilation use.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Cardiovascular Agents/administration & dosage , Heart Diseases/surgery , Perioperative Care/methods , Simendan/administration & dosage , Acute Kidney Injury/mortality , Acute Kidney Injury/prevention & control , Cardiac Surgical Procedures/mortality , Cardiovascular Agents/adverse effects , Drug Administration Schedule , Heart Diseases/diagnosis , Heart Diseases/mortality , Heart Diseases/physiopathology , Humans , Incidence , Intensive Care Units , Length of Stay , Perioperative Care/adverse effects , Renal Replacement Therapy , Respiration, Artificial , Risk Factors , Simendan/adverse effects , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left
9.
Clin Exp Pharmacol Physiol ; 44(11): 1125-1133, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28741726

ABSTRACT

Loss of function mutations in the human ether-a-go-go-related gene (hERG) cause long QT syndrome type 2 (LQT2). Most LQT2 patients are heterozygous mutation carriers in which the mutant hERG exerts potent dominant-negative effects. 1, 3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea (NS1643) is known to enhance IKr in WT-hERG. We investigated its actions following lipofectamine-induced expression of both mutant G604S- and WT-hERG in the heterologous HEK293 expression system. Cells transfected with pcDNA3-G604S-hERG did not lead to any expression of detectable currents whether before or following NS1643 challenge. Cells transfected with both pcDNA3-WT-hERG and pcDNA3-G604S-hERG showed reduced hERG currents compared to those transfected with pcDNA3-G604S-hERG consistent with the reduced trafficking and formation of modified heteromeric WT-G604S channels reported on earlier occasions. Nevertheless, NS1643 then continued to produce concentration- and voltage-dependent increases in hERG current amplitude. It did not affect the voltage dependence of activation, recovery from inactivation and deactivation. However, NS1643 (30 µmol/L) slowed steady state inactivation and shifted the steady state half maximal activation voltage (V1/2 ) of the inactivation curve by +10 mV, and significantly increased the time constants of inactivation. Our present experimental results suggest that NS1643 significantly increases ion current and attenuates its inactivation in cells co-expressing G604S-hERG and WT-hERG. These findings raise the possibility that hERG channel activators offer potential treatment strategies for inherited LQT2.


Subject(s)
Cresols/pharmacology , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/physiopathology , Mutation , Phenylurea Compounds/pharmacology , Action Potentials/drug effects , Alleles , Female , Gene Expression , HEK293 Cells , Heterozygote , Humans , Long QT Syndrome/genetics , Long QT Syndrome/pathology , Male , Pedigree , Phenotype
10.
Toxicol Sci ; 156(1): 25-38, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28031415

ABSTRACT

Drug-induced proarrhythmia is a major safety issue in drug development. Developing sensitive in vitro assays that can predict drug-induced cardiotoxicity in humans has been a challenge of toxicology research for decades. Recently, induced pluripotent stem cell-derived human cardiomyocytes (iPSC-hCMs) have become a promising model because they largely replicate the electrophysiological behavior of human ventricular cardiomyocytes. Patient-specific iPSC-hCMs have been proposed for personalized cardiac drug selection and adverse drug response prediction; however, many procedures are involved in cardiomyocytes differentiation and purification process, which may result in large line-to-line and batch-to-batch variations. Here, we examined the purity, cardiac ion channel gene expression profile, and electrophysiological response of 3 batches of iPSC-hCMs from each of 2 major cell suppliers. We found that iPSC-hCMs from both vendors had similar purities. Most of the cardiac ion channel genes were expressed uniformly among different batches of iCells, while larger variations were found in Cor.4U cells, particularly in the expression of CACNA1C, KCND2, and KCNA5 genes, which could underlie the differences in baseline beating rate (BR) and field potential duration (FPD) measurements. Although, in general, the electrophysiological responses of different batches of cells to Na+, Ca2+, Ikr, and Iks channel blockers were similar, with Ikr blocker-induced proarrhythmia, the sensitivities were depended on baseline BR and FPD values: cells that beat slower had longer FPD and greater sensitivity to drug-induced proarrhythmia. Careful evaluation of the performance of iPSC-hCMs and methods of data analysis is warranted for shaping regulatory standards in qualifying iPSC-hCMs for drug safety testing.


Subject(s)
Calcium Channel Blockers/adverse effects , Drug Evaluation, Preclinical/methods , Gene Expression Regulation/drug effects , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/adverse effects , Toxicity Tests, Acute/methods , Voltage-Gated Sodium Channel Blockers/adverse effects , Anti-Arrhythmia Agents/pharmacology , Antioxidants/pharmacology , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Differentiation/drug effects , Cell Line , Drug Evaluation, Preclinical/economics , Electrophysiological Phenomena/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Kinetics , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Potassium Channel Blockers/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Reproducibility of Results , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Toxicity Tests, Acute/economics , Voltage-Gated Sodium Channel Blockers/pharmacology
11.
Clin Exp Pharmacol Physiol ; 43(9): 851-61, 2016 09.
Article in English | MEDLINE | ID: mdl-27199074

ABSTRACT

Mutations in human ether-a-go-go-related gene (hERG) can lead to type 2 long-QT syndrome (LQT2). The authors previously identified the hERG mutation G604S results in a loss of function and obviously decreased current amplitude and impaired channel protein trafficking when co-expressed with WT-hERG. The present study further investigates the biological and electrophysiological consequences of pharmacologic chaperones in HEK293 cells expressing G604S-hERG or co-expressing G604S-hERG and WT-hERG. It was found that a low temperature (27°C), thapsigargin, NS1643 and E-4031 fail to rescue the G604S mutation. Interestingly, only E-4031 treatment resulted in a significant increase in hERG currents in cells co-expressing G604S-hERG and WT-hERG, correspondingly more mature protein band at 155 kDa by Western blotting and an increased membrane staining by confocal microscopy. In addition, E-4031 treatment shifted the steady-state half maximal activation voltage (V1/2 ) of the inactivation curve by +8 mV in cells co-expressing G604S-hERG and WT-hERG. The present experimental results suggest that a G604S mutation is resistant to pharmacological rescue. E-4031 treatment resulted in a significant increase in hERG currents by promoting the hERG channel processing and trafficking in cells co-expressing G604S-hERG and WT-hERG.


Subject(s)
Cresols/pharmacology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/genetics , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Mutation , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Amino Acid Substitution , Gene Expression , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Temperature
12.
Nutrition ; 32(6): 645-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26837599

ABSTRACT

OBJECTIVE: It has been confirmed that adipokines are associated with atherosclerosis. Cigarette smoking was found to possibly influence adipokine secretion. However, the precise role of smoking in adipokine secretion and the underlying mechanisms are largely unknown. The aim of this study was to determine whether nicotine, the principal active ingredient of cigarettes, can influence adipokine secretion and its potential mechanism. METHODS: The present study consecutively enrolled 96 men, including 50 smokers with early atherosclerosis and 46 nonsmokers. Serum adipokines, including leptin, resistin, and visfatin, were determined with enzyme-linked immunosorbent assay in all participants. Furthermore, the effect of nicotine on secretion of these adipokines was examined in differentiated 3T3-L1 preadipocytes under the conditions of ATP-dependent potassium (KATP) channel blocked or unblocked. RESULTS: Compared with the control group, serum levels of leptin, resistin, and visfatin in smokers were significantly higher. In 3T3-L1 adipocytes, nicotine treatment significantly increased the levels of these adipokines (P = 0.014, 0.001, and 0.029, respectively). When the KATP channel was blocked, secretion of resistin and visfatin was reduced (P < 0.001), but no change was found in the leptin secretion (P = 0.522). CONCLUSIONS: Nicotine may affect the secretion of adipokines leptin, resistin, and visfatin through activation of KATP channel.


Subject(s)
KATP Channels/blood , Leptin/blood , Nicotinamide Phosphoribosyltransferase/blood , Nicotine/pharmacology , Resistin/blood , Enzyme-Linked Immunosorbent Assay , Humans , KATP Channels/drug effects , Male , Middle Aged , Nicotinamide Phosphoribosyltransferase/drug effects , Nicotine/blood , Nicotinic Agonists/blood , Nicotinic Agonists/pharmacology
13.
Mol Cell Biochem ; 382(1-2): 307-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23793583

ABSTRACT

The LMNA gene, which encodes the nuclear envelope protein lamin A/C, is considered to be the most common autosomal disease gene associated with familial dilated cardiomyopathy. To date, each mutation of the LMNA gene has been associated with a specific disease phenotype. Clinical data, family histories, and blood samples were collected from 27 biological members of a family with dilated cardiomyopathy, prominently occurring as heart failure and conduction system disease with a high incidence of sudden cardiac death in young females. Twelve exons of the LMNA gene were screened for nucleotide alterations. A novel insertion mutation (nucleotide 1526insA, amino acid T510Y) in exon nine of the LMNA gene was identified in seven subjects (7/27, 25.9%). This reveals that the LMNA gene insertion mutation (T510Y frameshift mutation) can cause dilated cardiomyopathy, conduction system disease, and sudden cardiac death without skeletal myopathy, clinically manifested with early onset, severe symptoms, and poor prognosis.


Subject(s)
Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/genetics , Death, Sudden, Cardiac , Heart Conduction System/abnormalities , Lamin Type A/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Brugada Syndrome , Cardiac Conduction System Disease , Child , Family , Female , Frameshift Mutation/genetics , Heterozygote , Humans , Lamin Type A/chemistry , Male , Molecular Sequence Data , Mutation/genetics , Pedigree , Young Adult
14.
J Cell Mol Med ; 15(2): 296-306, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19912438

ABSTRACT

Our previous works revealed that human ribosomal protein S13 (RPS13) was up-regulated in multidrug-resistant gastric cancer cells and overexpression of RPS13 could protect gastric cancer cells from drug-induced apoptosis. The present study was designed to explore the role of RPS13 in tumorigenesis and development of gastric cancer. The expression of RPS13 in gastric cancer tissues and normal gastric mucosa was evaluated by immunohistochemical staining and Western blot analysis. It was found RPS13 was expressed at a higher level in gastric cancer tissues than that in normal gastric mucosa. RPS13 was then genetically overexpressed in gastric cancer cells or knocked down by RNA interference. It was demonstrated that up-regulation of RPS13 accelerated the growth, enhanced in vitro colony forming and soft agar cologenic ability and promoted in vivo tumour formation potential of gastric cancer cells. Meanwhile, down-regulation of RPS13 in gastric cancer cells resulted in complete opposite effects. Moreover, overexpression of RPS13 could promote G1 to S phase transition whereas knocking down of RPS13 led to G1 arrest of gastric cancer cells. It was further demonstrated that RPS13 down-regulated p27(kip1) expression and CDK2 kinase activity but did not change the expression of cyclin D, cyclin E, CDK2, CDK4 and p16(INK4A). Taken together, these data indicate that RPS13 could promote the growth and cell cycle progression of gastric cancer cells at least through inhibiting p27(kip1) expression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Ribosomal Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin D/biosynthesis , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation , G1 Phase Cell Cycle Checkpoints , Gastric Mucosa/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Ribosomal Proteins/biosynthesis
15.
Int J Cardiol ; 147(1): 58-65, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-19729212

ABSTRACT

OBJECTIVE: To investigate common downstream mechanism of PGE2 and O2-sensitive voltage-dependent potassium (Kv) channels in preterm and term DA tone regulations, for suggesting respective prescriptions for preterm and term PDA. STUDY DESIGN: The expressions of Kv1.2, 1.5 and 2.1 were compared between preterm and term in rabbit and human DAs at mRNA and protein levels; DA contracting responses caused by O2, Kv channels blocker 4-AP, EP4 antagonist GW627368X, and PGE2 reduce using vessels rings and Whole-Cell Patch-Clamp were explored. RESULTS: Kv 1.2 and 2.1 expressions were developed with pregnant age in preterm DA and decreased after birth with oxygen stimulation in term DA. GW627368X led significant DA constriction and DASMC IK current decrease in preterm, which was slimier to 4-AP effects, but just slightly influenced on DA tension and DASMC IK current at term. In addition, PGE2 led great DA dilation and IK current increase of DASMC in preterm but not in term. These DA tension and IK current changes were in line with Kv channel expressions. CONCLUSION: Higher levels of PGE2 binds with GPCR EP4, which activates G-protein to couple with O2-sensitive Kv channels and to open them, leading to DA vasorelaxation in the fetus. It indicates that EP4 inhibitors, instead of PGE2 or its analogue PGE1, may be a selectable strategy for preterm PDA.


Subject(s)
Dinoprostone/physiology , Ductus Arteriosus/metabolism , Oxygen/physiology , Potassium Channels, Voltage-Gated/physiology , Receptors, Prostaglandin E, EP4 Subtype/physiology , Vasoconstriction/physiology , Animals , Ductus Arteriosus/embryology , Ductus Arteriosus/physiology , Female , Fetus , Humans , Infant, Newborn , Isoindoles/pharmacology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Pregnancy , Rabbits , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/metabolism , Signal Transduction/physiology , Sulfonamides/pharmacology , Vasoconstriction/drug effects
16.
Cell Biol Int ; 35(3): 193-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21143190

ABSTRACT

EGFP (enhanced green fluorescent protein) tagged to either the N (amino)-terminus [EGFP/hERG (human ether-a-go-go-related gene)] or C (carboxyl)-terminus (hERG/EGFP) of hERG channel is used to study mutant channel protein trafficking for several years. However, it has been reported that the process can alter hERG channel properties. The aim of the study was to determine whether EGFP tagged to N-terminus of hERG channels would alter the cellular localizations and the electrophysiological properties of hERG channels compared with untagged hERG channels. The hERG channels tagged with or without EGFP were transiently expressed in HEK (human embryonic kidney) 293 cells using a lipofectamine method. HEK 293 cells expressing pCDNA3-hERG or pEGFP-hERG were double immunolabelled with anti-hERG and anti-calnexin (an ER marker protein) followed with FITC- and TRITC (tetramethylrhodamine ß-isothiocyanate)-labelled secondary antibodies, respectively. Confocal laser scanning microscope was used to observe the cellular localization of EGFP-tagged hERG channels and untagged hERG channels. Patch-clamp technique was used to record whole cell currents. We found that the EGFP/hERG fusion protein and untagged hERG channels were both expressed not only on the cell surface membrane but also in the cytoplasm of HEK293 cells. The EGFP/hERG appeared to influence the hERG channel gating properties, including reduction of the peak tail current density, more rapid inactivation process, faster recovery from inactivation and faster deactivation kinetics compared with untagged hERG channels. Our results suggest that the EGFP/hERG channel alter the electrophysiological properties of hERG channel, but it does not seem to alter the cellular location of hERG channels. Thus, EGFP tagging to N-terminus might be used for research of subcellular location of hERG channels but not for the channel electrophysiological properties.


Subject(s)
Ether-A-Go-Go Potassium Channels/physiology , Green Fluorescent Proteins/metabolism , Calnexin/immunology , ERG1 Potassium Channel , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Microscopy, Confocal , Patch-Clamp Techniques , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
17.
Can J Cardiol ; 26(8): 417-22, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20931094

ABSTRACT

BACKGROUND: The congenital long QT syndrome is a heterogeneous genetic disease associated with delayed cardiac repolarization, prolonged QT intervals, the development of ventricular arrhythmias and sudden death. Type 2 congenital long QT syndrome (LQT2) results from KCNH2 or hERG gene mutations. hERG encodes the K(v)11.1 alpha subunit of the rapidly activating delayed rectifier K(+) current in the heart. Studies of mutant hERG channels indicate that most LQT2 missense mutations generate trafficking-deficient K(v)11.1 channels. OBJECTIVE: To identify the mechanism underlying G572R-hERG by using molecular and electrophysiological analyses. METHODS AND RESULTS: To elucidate the electrophysiological properties of the G572R-hERG mutant channels, mutant hERG subunits were heterologously expressed in HEK293 cells alone or in combination with wild-type (WT)-hERG subunits. Patch-clamp techniques were used to record currents, and double immunofluorescence protein tagging and Western blotting were performed to examine the cellular trafficking of mutant subunits. When expressed alone, G572R-hERG subunits were not present in the cell membrane and did not produce detectable currents. When coexpressed with WT-hERG subunits, G572R-hERG decreased current density and altered gating properties of the WT-hERG channel. CONCLUSION: The hERG-associated missense mutation G572R, like most LQT2 missense mutations, generates a trafficking-deficient phenotype. Furthermore, G572R-hERG causes a loss of function in hERG by a strong dominant negative effect on the WT-hERG channel.


HISTORIQUE : Le syndrome du QT long congénital est une maladie génétique hétérogène associée à un retard de la repolarisation cardiaque, à des intervalles QT prolongés, à l'apparition d'arythmies ventriculaires et à une mort subite. Le syndrome du QT long congénital de type 2 (QTL2) est causé par des mutations du gène KCNH2 ou hERG. Le gène hERG code la sous-unité alpha K11.1 du canal K+ redresseur retardé du cœur qui s'active rapidement. Des études des canaux hERG mutants indiquent que la plupart des mutations faux-sens QTL2 produisent des canaux K11.1 au trafic défectueux. OBJECTIF : Dépister les mécanismes sous-jacents du canal G572R-hERG au moyen d'analyses moléculaires et électrophysiologiques. MÉTHODOLOGIE ET RÉSULTATS : Afin d'établir les propriétés électrophysiologiques des canaux mutants G572R-hERG, les sous-unités mutantes hERG étaient exprimées de manière hétérologue dans des cellules HEK293, seules ou en combinaison avec des sous-unités hERG de type sauvage (WT). Les techniques du patch-clamp ont permis d'enregistrer les courants, tandis que le marquage des protéines par double immunofluorescence et le transfert Western ont permis d'examiner le trafic cellulaire des sous-unités mutantes. Exprimées seules, les sous-unités G572R-hERG étaient absentes de la membrane cellulaire et ne produisaient pas de courants décelables. Coexprimé avec les sous-unités WT-hERG, le canal G572R-hERG réduisait la densité du courant et modifiait les propriétés de synchronisation du canal WT-hERG. CONCLUSION : La mutation faux-sens du gène G572R associée au gène hERG, comme la plupart des mutations faux-sens QTL2, produit un phénotype au trafic défectueux. De plus, le canal G572R-hERG entraîne une perte de fonction du gène hERG par son puissant effet négatif dominant sur le canal WT-hERG.


Subject(s)
DNA/genetics , Ether-A-Go-Go Potassium Channels/genetics , Long QT Syndrome/metabolism , Mutation, Missense , Protein Transport/genetics , Blotting, Western , Cells, Cultured , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/biosynthesis , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Long QT Syndrome/congenital , Long QT Syndrome/genetics , Patch-Clamp Techniques , Prognosis
18.
J Surg Res ; 155(1): 18-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19394655

ABSTRACT

BACKGROUND: Roles that bone marrow stem cells (BMSCs) play in liver repair after liver injury and the cell therapy for liver diseases are widely accepted. However, the availability of hepatocyte-like cells from BMSCs and possible animal diseases association with culturing in fetal calf serum (FCS) are the major limitations of clinical therapy. Therefore, this study was designed to search for a new cell source for the treatment of liver injuries through investigating whether serum from radiofrequency ablation-injured rabbit livers can induce the differentiation of BMSCs into hepatocyte-like cells. METHODS: Serum was collected from rabbits 36 h after radiofrequency ablation (RFA) treatment of the liver. BMSCs were isolated from rabbit bone marrow and were cultured in the collected serum. Cellular morphology and cell cycle were observed. Hepatocyte markers of the differentiated cells were detected by immunohistochemistry. RESULTS: After induction for 7 d, spindle-shaped BMSCs turned into round cells that resembled the morphology of hepatocyte-like cells. Flow cytometry showed that the percentage of cells in the S/G2/M phase was higher in the RFA group than that in the FCS group and HGF groups. This result suggests that BMSC can transform into mature cells from stem cell phase. Albumin and CK18 were considered as typical marker of hepatocytes. Following induction for 14 d, the differentiated cells expressed immunofluorescence of FITC-labeled albumin and TRITC-labeled CK18. CONCLUSION: BMSCs treated with serum collected from radiofrequency ablation-injured livers can differentiate into hepatocyte-like cells providing a cell source to cell therapy.


Subject(s)
Blood Transfusion, Autologous , Bone Marrow Cells/cytology , Catheter Ablation , Cell Differentiation , Hepatocytes/cytology , Liver Regeneration , Animals , Cell Cycle , Flow Cytometry , Fluorescent Antibody Technique , Liver Neoplasms/therapy , Rabbits
19.
Nutrition ; 25(3): 359-63, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19056241

ABSTRACT

OBJECTIVE: This study was designed to evaluate the contribution of adenosine triphosphate-dependent potassium channels to the increase in blood pressure observed in obese rats. METHODS: The experiment was performed in male Sprague-Dawley rats. Glibenclamide-sensitive currents were measured in vascular smooth muscle cells by patch-clamp. Expressions of Kir6.1 and SUR2B were examined by reverse transcription polymerase chain reaction and western blot techniques, respectively. RESULTS: In the aortic vascular smooth muscle cells, pinacidil induced glibenclamide-sensitive currents. The current from obese rats was significantly lower (-10.55 +/- 1.63 pA/pF) compared with that from the control rats (-20.18 +/- 2.79 pA/pF). Expressions of Kir6.1 and SUR2B were downregulated in vascular smooth muscle cells of aortas from the obese rats. CONCLUSION: These findings suggest that the adenosine triphosphate-dependent potassium channel is downregulated in smooth muscle cells from the aortas of obese rats, which may contribute to the increase in blood pressure in these rats.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aorta/metabolism , Blood Pressure/physiology , Down-Regulation , Obesity/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Blood Pressure/drug effects , Blotting, Western , Glyburide/pharmacology , KATP Channels , Male , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Pinacidil/pharmacology , Potassium Channels, Inwardly Rectifying/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Drug/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sulfonylurea Receptors , Vasodilator Agents/pharmacology
20.
Acta Pharmacol Sin ; 29(10): 1167-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18817620

ABSTRACT

AIM: To determine whether ATP-sensitive potassium channels are altered in VSMC from arotas and mesenteric arteries of obese rat, and their association with obesity-triggered increase in blood pressure. METHODS: Obesity was induced by 24 weeks of high-fat diet feeding in male Sprague-Dawley rats. Control rats were fed with standard laboratory rat chow. Blood pressure and body weight of these rats were measured every 4 weeks. At the end of 24 weeks, K(ATP) channelmediated relaxation responses in the aortas and mesenteric arteries, K(ATP) channel current, and gene expression were examined, respectively. RESULTS: Blood pressure and body weight were increased in rats fed with high-fat diet. K(ATP) channelmediated relaxation responses, currents, and K(ATP) expression in VSMC of both aortas and mesenteric arteries were inhibited in these rats. CONCLUSION: Altered ATP-sensitive potassium channels in obese rats may underscore obesity-triggered increase in blood pressure.


Subject(s)
Blood Pressure/physiology , KATP Channels/physiology , Myocytes, Smooth Muscle/metabolism , Obesity/metabolism , Obesity/physiopathology , Animals , Antihypertensive Agents/therapeutic use , Aorta, Thoracic/drug effects , Body Weight/drug effects , Dietary Fats , Electrophysiology , KATP Channels/biosynthesis , Male , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Patch-Clamp Techniques , Pinacidil/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...