Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Tohoku J Exp Med ; 225(1): 51-7, 2011 09.
Article in English | MEDLINE | ID: mdl-21869591

ABSTRACT

Electrical remodeling at infarct border zone has been shown to contribute to the occurrence of ventricular arrhythmias after myocardial infarction (MI). Electrical remodeling is causally associated with sympathetic neural remodeling in MI. Semaphorin 3A (Sema3A), a potent neural chemorepellent for sympathetic axons, has been demonstrated to suppress sympathetic neural remodeling after MI. In the present study, we investigated whether treatment with Sema3A can ameliorate electrical remodeling at infarct border zones using a rat model of MI. Wistar rats underwent sham operation (n = 20), the ligation of left coronary artery (MI group, n = 30), MI with control adenovirus (Ad group, n = 30), and MI with Sema3A adenovirus (Sema3A group, n = 30). Eight weeks after treatment, electrophysiological properties including heart rate variability (HRV), monophasic action potential duration (MAPD) and effective refractory period (ERP) and the expression of arrhythmia-related ion channel proteins including Kv4.2, KChIP2 and Kir2.1 at the infarcted border of the left ventricle were examined. These channel proteins may be required for maintaining normal heart rhythm. Compared with the Ad group, Sema3A significantly increased HRV and shortened MAPD and ERP (all p < 0.05). The expression levels of Kv4.2, KChIP2 and Kir2.1 proteins were significantly decreased in MI group and Ad group, compared to sham control. In contrast, the expression levels of these proteins were restored in Sema3A group, which may represent the molecular basis of the Sema3A-mediated inhibition of electrical remodeling. In conclusion, Sema3A can ameliorate electrical remodeling at infarct border zones after MI.


Subject(s)
Electrophysiological Phenomena , Genetic Therapy , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Semaphorin-3A/genetics , Semaphorin-3A/therapeutic use , Ventricular Remodeling/physiology , Animals , Blood Pressure/physiology , Blotting, Western , Heart Rate/physiology , Male , Myocardial Infarction/pathology , Potassium Channels/metabolism , Rats , Rats, Wistar , Staining and Labeling
2.
Cell Biol Int ; 34(12): 1155-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20718713

ABSTRACT

Thrombin acts as a potent mitogenic factor for ECs (endothelial cells) by the release of several growth factors, including PDGF-B (platelet-derived growth factor-B). CBP (CREB-binding protein), which functions as a transcriptional coactivator, links the changes in the extracellular stimuli with alterations in gene expression. Therefore, we hypothesized that CBP could mediate thrombin-induced proliferation of ECs via PDGF-B-dependent way. Short hairpin RNA was used to down-regulate the expression of CBP in ECs. CBP and PDGF-B levels were analysed by real-time RT-PCR and Western blot. To evaluate ECs proliferation, cell cycle and DNA synthesis were analysed by flow cytometry and BrdU (bromodeoxyuridine) incorporation assay, respectively. PDGF-B was involved in the mitogenic effect of thrombin on ECs. Down-regulation of CBP attenuated ECs proliferation and inhibited cell cycle progression induced by thrombin. Silencing CBP expression also suppressed thrombin-induced PDGF-B expression in ECs. Mitogenic activity of thrombin was impaired by silencing CBP expression in ECs. This inhibitory effect was, in part, related to the inability to up-regulate PDGF-B expression in ECs. CBP could be regarded as a potential therapeutic target for vascular injury.


Subject(s)
CREB-Binding Protein/genetics , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Proto-Oncogene Proteins c-sis/physiology , Thrombin/pharmacology , Animals , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/metabolism , CREB-Binding Protein/physiology , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Rats
3.
Zhonghua Yi Xue Za Zhi ; 88(24): 1707-10, 2008 Jun 24.
Article in Zh | MEDLINE | ID: mdl-19024544

ABSTRACT

OBJECTIVE: To investigate the effects of sympathetic nerve stimulation (SNS) on connexin43 (Cx43) and ventricular arrhythmias during acute myocardial ischemia (MI). METHODS: Ninety five Wistar rats were randomly divided into four groups: MI group (n=25), undergoing: ligation of the anterior descending coronary; MII-SNS group (n=25); undergoing electric stimulation of sympathetic nerve since the beginning of ligation of the anterior descending coronary and lasting till 30 min after the ligation, sympathetic nerve stimulation preconditioning + myocardial ischemia (pSNS-MI) group (n=25), undergoing electric stimulation of sympathetic nerve since the beginning of ligation of the anterior descending coronary that ended just after the ligation; and sham operation (SO) group (n=20), without coronary ligation. Ventricular arrhythmias were monitored by electrocardiography. Western blotting and RT-PCR were used to detect the protein and mRNA expression of Cx43 respectively. Immunofluorescence analysis was used to observe the changes of Cx43 protein distribution. RESULTS: One and 3 rate died due to ventricular fibrillation in the MI group and MI-SS group respectively. The incidence of ventricular tachycardia (VT)/VF within 30-minute after ligation in the MI-SNS group was 80.0%, significant higher than that of the MI group (52.0%, P < 0.05). The incidence of VT/VF within 30-minute after ligation of the pSNS-MI group was 20.0%, significantly lower than that of the MI-SNS group (P < 0.05). 30 minutes after the ligation, the percentage of phosphorylated Cx43 of the pSNS-MI and MI-SNS groups were 71.2% +/- 7.0% and 73.4% +/- 6.7% respectively, both significantly higher than that of the MI group (46.7% +/- 6.3%) (both P < 0.05). The total contents of Cx43 of the MI and pSNS-MI groups were 1.29 +/- 0.14 and 1.25 +/- 0.13 respectively, both similar to that of the SO group [(1.30 +/- 0.10), both P > 0.05], while the total Cr43 content of the MI-SNS group was 0.73 +/- 0. 12, significantly lower than that of the SO group [(1.30 +/- 0.10), P < 0.05]. The Cx43 mRNA levels of the 3 experimental groups were all significantly lower than that of the SO group (all P < 0.05). Immunofluorescence analysis confirmed that ischemia and sympathetic nerve stimulation induced the changes of connexin43 distribution and sympathetic nerve stimulation preconditioning inhibited the changes of connexin43 distribution induced by ischemia. CONCLUSION: SNS promotes ventricular arrhythmias by promoting Cx43 degradation, and sympathetic nerve stimulation preconditioning inhibits ventricular arrhythmias by preventing Cx43 dephosphorylation.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Connexin 43/metabolism , Myocardial Ischemia/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Blotting, Western , Connexin 43/genetics , Electric Stimulation/methods , Fluorescent Antibody Technique , Male , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
4.
Auton Neurosci ; 214: 19-23, 2018 11.
Article in English | MEDLINE | ID: mdl-30100240

ABSTRACT

BACKGROUND: Electrical remodeling at infarct border zone (IBZ) has been shown to contribute to the occurrence of ventricular arrhythmias after myocardial infarction (MI). Sema3A has been demonstrated to reduce the inducibility of ventricular arrhythmias. Neuropilin 1 (NRP1) is the receptor of Sema3A. In the present study, we investigated whether treatment with NRP1 can ameliorate electrical remodeling at IBZ after MI. METHODS AND RESULTS: Wistar rats underwent sham operation (n = 20), the ligation of left coronary artery (MI group, n = 30), MI with control adenovirus (Ad group, n = 30), and MI with NRP1 adenovirus (NRP1 group, n = 30). Eight weeks after treatment, electrophysiological properties including heart rate variability (HRV), monophasic action potential duration (MAPD), effective refractory period (ERP) and the inducibility of ventricular arrhythmias and the expression of arrhythmia-related ion channel proteins including Kv4.2, Kv4.3, KChIP2 and Kir2.1 at the IBZ of the left ventricle were examined. Compared with the MI or Ad group, NRP1 significantly increased HRV and shortened MAPD and ERP (all P < 0.05). Inducibility of VT by electrophysiological study was significantly lower in the NRP1 group than in the MI or Ad group (P < 0.05). The expression levels of Kv4.2, Kv4.3, KChIP2 and Kir2.1 proteins were significantly decreased in MI group and Ad group. In contrast, the expression levels of these proteins were restored in NRP1 group, which may represent the molecular basis of the NRP1-mediated inhibition of electrical remodeling. CONCLUSIONS: NRP1 can ameliorate electrical remodeling at IBZ after MI.


Subject(s)
Atrial Remodeling/drug effects , Myocardial Infarction/complications , Myocardial Infarction/therapy , Neuropilin-1/therapeutic use , Analysis of Variance , Animals , Disease Models, Animal , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/pathology , Hemodynamics , Kv Channel-Interacting Proteins/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Rats , Rats, Wistar , Transduction, Genetic
5.
Zhonghua Xin Xue Guan Bing Za Zhi ; 35(9): 844-7, 2007 Sep.
Article in Zh | MEDLINE | ID: mdl-18070481

ABSTRACT

OBJECTIVE: To investigate the effects of endothelial progenitor cells (EPCs) transfusion on reendothelialization and neointima proliferation in balloon-denuded rabbit carotid arteries. METHOD: Bone marrow-derived rabbit mononuclear cells (MNCs) were cultured in endothelial basal medium to form EPCs. The cell makers were assayed by immunocytochemistry. The rabbit right carotid artery was injured with 2.5 FPTCA balloon catheter and the EPCs (5 ml) were transfused into the injured carotid artery at a rate of 15 ml/h. The rabbits were killed at 7 days or 14 days post operation. Reendothelialization area and ratio of intima/media (I/M) in injured artery were measured. RESULT: EPCs transfusion significantly increased the percentage of endothelialization at 7 days (50.923% +/- 2.476% vs. 28.647% +/- 2.241%) and at 14 days (82.609% +/- 2.611% vs. 49.800% +/- 2.660%) compared to control group (all P < 0.05). I/M ratio was significantly lower in EPCs transfusion group than that in control group (0.378 +/- 0.029 vs. 0.898 +/- 0.038, P < 0.05) 14 days after operation. The labeled EPCs could be detected by immunohistochemistry in the injured vessel wall. CONCLUSION: Intra-arterial transfusion of EPCs can effectively accelerate reendothelialization and reduce neointima formation in balloon-denuded rabbit carotid arteries.


Subject(s)
Angioplasty/methods , Carotid Artery Injuries/therapy , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Female , Male , Rabbits , Regeneration , Stem Cells/cytology
6.
Mol Cell Biochem ; 315(1-2): 123-30, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18496732

ABSTRACT

Thrombin is a potent mitogen for vascular smooth muscle cells (VSMCs). CBP has been regarded as a potential therapeutic target on the basis of its ability to affect cell growth. Therefore we hypothesized that CBP mediates thrombin-induced proliferation of VSMCs. We constructed recombinant adenoviral vector that expresses four short hairpin RNA (shRNA) targeting rat CBP mRNA (CBP-shRNA/Ad). VSMCs were infected with CBP-shRNA/Ad and treated with thrombin. CBP level were analyzed by quantitative real-time PCR and Western blot. To evaluate VSMC proliferation, the cell cycle and DNA synthesis were analyzed by flow cytometry and (3)H-thymidine incorporation, respectively. CBP-shRNA/Ad infection inhibited thrombin-induced CBP expression in a dose-dependent manner concomitant with a decrease in the percentage of cells in the S phase and in DNA synthesis. These findings suggest that CBP plays a pivotal role in the S phase progression of VSMCs.


Subject(s)
CREB-Binding Protein/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Thrombin/pharmacology , Adenoviridae , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , DNA/biosynthesis , RNA, Small Interfering/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL