Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tingzhong Wang is active.

Publication


Featured researches published by Tingzhong Wang.


Transplant International | 2006

Intravenous transplantation of mesenchymal stem cells improves cardiac performance after acute myocardial ischemia in female rats

Wenhui Jiang; Aiqun Ma; Tingzhong Wang; Ke Han; Yu Liu; Yanmin Zhang; Xiaoge Zhao; Anping Dong; Yuan Du; Xin Huang; Jun Wang; Xinjun Lei; Zheng Xp

Mesenchymal stem cells (MSCs) are potential sources of cells for tissue repairing. However, little information is available regarding the therapeutic potency of intravenously transplanted MSCs for myocardial ischemia (MI). In the present study, MSCs were isolated from bone marrow of male rats and expanded in vitro. Three hours after ligation of left anterior descending artery, the transplanted group received an infusion of MSCs through the tail vein. At the same time, a coronary‐ligated control group was injected with culture medium. Homing of MSCs to the heart was assessed by expression of the Y chromosome sry gene using fluorescent in situ hybridization (FISH). At 1 week or 8 weeks after transplantation, sry positive cells were present in cardiac tissue in the transplanted group, but not in the hearts of control group. Cardiomyocytes, smooth muscle cells, and endothelial cells that bore sry gene were identified in transplanted group at 8 weeks after transplantation. Ultrastructural observation revealed that a large number of capillary and some immature myocytes were found to survive in the ischemia region. MSCs transplantation also decreased LVEDP pressure and −dP/dt, but increased LVSP and +dP/dt. The cardiac infarct size was significantly smaller in transplanted group than in control group. Our data suggest that intravenously transplanted MSCs improve cardiac performance and promote the regeneration of blood vessels and cardiomyocytes.


European Journal of Heart Failure | 2009

Increased late sodium currents are related to transcription of neuronal isoforms in a pressure-overload model

Yutao Xi; Geru Wu; Lin Yang; Ke Han; Yuan Du; Tingzhong Wang; Xinjun Lei; Xiaojun Bai; Aiqun Ma

The late and persistent sodium current (INa) has been identified as a target for anti‐arrhythmia drugs in patients with heart failure (HF). However, the underlying mechanism of late INa (INaL) production remains uncertain. We hypothesized that transcriptional alteration among sodium channel (NaCh) isoforms may contribute to INaL in failing cardiomyocytes.


Pflügers Archiv: European Journal of Physiology | 2007

Downregulation of neuronal sodium channel subunits Nav1.1 and Nav1.6 in the sinoatrial node from volume-overloaded heart failure rat

Yuan Du; Xin Huang; Tingzhong Wang; Ke Han; Junbo Zhang; Yutao Xi; Geru Wu; Aiqun Ma

Sodium current INa plays an important role in the pacemaker activity of the sinoatrial node (SAN). However, expression profiles of corresponding sodium channel subunits in normal SAN remain unclear. And little is known about expression alteration of sodium channel in SAN under heart failure (HF) condition. We assessed SAN function and expression of Nav1.1, Nav1.2, Nav1.3, Nav1.5, Nav1.6, and Nav1.7 in sham-operated rats and rats subjected to abdominal arteriovenous shunt (volume overload)-induced HF. Immunohistochemistry, Western blot, and quantitative real-time reverse transcriptase PCR analysis were used to quantify sodium channel subunit protein and mRNA expression in the SAN. Intrinsic heart rate declined and sinus node recovery time was prolonged in HF rats, indicating suppressed SAN pacemaker function. In rat SAN, Nav1.1 and Nav1.6 were the primary subunits, Nav1.5 and Nav1.7 were weakly expressed, and Nav1.2 and Nav1.3 were not found to be present. HF significantly decreased SAN sodium channel expression at both the protein and mRNA levels (Nav1.1 by 61 and 71%, Nav1.6 by 49 and 46%, respectively). In conclusion, Nav1.1 and Nav1.6 are the dominant subunits in rat SAN, and downregulation of Nav1.1 and Nav1.6 expression contributes to HF-induced SAN dysfunction. These findings provide additional information about molecular basis of disease-related impairment of SAN function.


Journal of Biological Chemistry | 2011

Interaction between the Cardiac Rapidly (IKr) and Slowly (IKs) Activating Delayed Rectifier Potassium Channels Revealed by Low K+-induced hERG Endocytic Degradation

Jun Guo; Tingzhong Wang; Tonghua Yang; Jianmin Xu; Wentao Li; Michael D. Fridman; John T. Fisher; Shetuan Zhang

Background: A reduction in either IKr or IKs can cause long QT syndrome. Results: Enhanced endocytic degradation of IKr decreases the expression of both IKr and IKs in the plasma membrane. Conclusion: IKr and IKs form a macrocomplex at the plasma membrane. Significance: Elucidation of IKr-IKs interaction is important for understanding the pathology of cardiac arrhythmias and designing anti-arrhythmic strategies. Cardiac repolarization is controlled by the rapidly (IKr) and slowly (IKs) activating delayed rectifier potassium channels. The human ether-a-go-go-related gene (hERG) encodes IKr, whereas KCNQ1 and KCNE1 together encode IKs. Decreases in IKr or IKs cause long QT syndrome (LQTS), a cardiac disorder with a high risk of sudden death. A reduction in extracellular K+ concentration ([K+]o) induces LQTS and selectively causes endocytic degradation of mature hERG channels from the plasma membrane. In the present study, we investigated whether IKs compensates for the reduced IKr under low K+ conditions. Our data show that when hERG and KCNQ1 were expressed separately in human embryonic kidney (HEK) cells, exposure to 0 mm K+ for 6 h completely eliminated the mature hERG channel expression but had no effect on KCNQ1. When hERG and KCNQ1 were co-expressed, KCNQ1 significantly delayed 0 mm K+-induced hERG reduction. Also, hERG degradation led to a significant reduction in KCNQ1 in 0 mm K+ conditions. An interaction between hERG and KCNQ1 was identified in hERG+KCNQ1-expressing HEK cells. Furthermore, KCNQ1 preferentially co-immunoprecipitated with mature hERG channels that are localized in the plasma membrane. Biophysical and pharmacological analyses indicate that although hERG and KCNQ1 closely interact with each other, they form distinct hERG and KCNQ1 channels. These data extend our understanding of delayed rectifier potassium channel trafficking and regulation, as well as the pathology of LQTS.


Molecular and Cellular Biochemistry | 2013

Bisoprolol reversed small conductance calcium-activated potassium channel (SK) remodeling in a volume-overload rat model

Yajuan Ni; Tingzhong Wang; Xiaozhen Zhuo; Bingxue Song; Jing Zhang; Feng Wei; Hongyuan Bai; Xuehui Wang; Dandan Yang; Li Gao; Aiqun Ma

A recent study indicated that apamin-sensitive current (IKAS, mediated by apamin-sensitive small conductance calcium-activated potassium channels subunits) density significantly increased in heart failure and led to recurrent spontaneous ventricular fibrillation. While the underlying molecular correlation with SK channels is still undetermined, we hypothesized that they are remodeled in HF and that bisoprolol could reverse the remodeling. Volume-overload models were created on male Sprague-Dawley rats by producing an abdominal arteriovenous fistula. Confocal microscopy, quantitative real-time PCR, and western blot were performed to investigate the expression of SK channels and observe the influence of β-blocker bisoprolol on the expression of SK channels IKAS, and the effect of bisoprolol on IKAS and the sensitivity of IKAS to [Ca2+]i at single isolated cells were also explored using whole-cell patch clamp techniques. SK channels were remodeled in HF rats, displaying the significant increase of SK1 and SK3 channel expression. After the treatment of HF rats with bisoprolol, the expression of SK1 and SK3 channels was significantly downregulated, and bisoprolol effectively downregulated IKAS density as well as the sensitivity of IKAS to [Ca2+]i. Our data indicated that the expression of SK1 and SK3 increased in HF. Bisoprolol effectively attenuated the change and downregulated IKAS density as well as the sensitivity of IKAS to [Ca2+]i.


Experimental Cell Research | 2011

The subpopulation of mesenchymal stem cells that differentiate toward cardiomyocytes is cardiac progenitor cells

Feng Wei; Tingzhong Wang; Juanjuan Liu; Yuan Du; Aiqun Ma

Mesenchymal stem cells (MSCs) are regarded as a promising source of cell-based therapy for heart injury. In fact, less than 30% of MSCs contribute to cardiomyocytes differentiation, and the isolation procedure and biological characteristics of this population of cells remain unknown. Here we isolate and investigate the biological characteristics of this subpopulation of MSCs. Twenty four MSC clones were randomly selected using single-cell monoclonal technology. After induced with 5-azacytidine, eight clones displayed cardiomyocyte-like morphologies, and highly (over 90%) expressed cardiac-specific markers cTnT and α-actin, and displayed transient outward K(+) current (I(to)), inwardly rectifying K(+) current (I(K1)) and delayed rectifier K(+) current (I(KDR)), which were typical of cardiomocytes. Other clones merely showed I(to) current, and the current densities were different from those of cardiomyocytes. In contrast to the other clones, before induced with 5-azacytidine, the eight clones expressed early cardiac markers GATA4 and NKX2.5, but not cTnT, α-actin, CD44 and CD90, and had no potentials for adiopogenesis, osteogenesis or chondrogenesis after induction. Our data suggest that the subgroup of MSCs that contributes to cardiomyocytes differentiation is cardiac progenitor cells. Moreover, we show the preliminary purification of this population of cells with a high potential for cardiomyocytes differentiation using single-cell monoclonal technology.


Molecular and Cellular Biochemistry | 2014

Serotonin drives the activation of pulmonary artery adventitial fibroblasts and TGF-β1/Smad3-mediated fibrotic responses through 5-HT(2A) receptors.

Chunyan Chen; Xinyuan Han; Fenling Fan; Ya Liu; Tingzhong Wang; Juanjuan Wang; Peijing Hu; Aiqun Ma; Hong-Yan Tian

Pulmonary arterial remodeling is characterized by excessive proliferation, migration, and pro-differentiation and fibrotic activation of adventitial fibroblasts in pulmonary arterial hypertension (PAH) process. Several lines of evidence indicate that serotonin (5-HT) plays a central role in the pathogenesis of pulmonary arterial remodeling. In the present study, we investigated whether 5-HT is directly involved in the functional regulation of pulmonary artery adventitial fibroblasts (PAFs). Incubation of cultured rat PAFs with 5-HT caused a dose-dependent stimulation of cell proliferation, migration activity, and a time-dependent increase of α-SMA expression, a marker of fibroblast differentiation into myofibroblasts, and adventitia fibrosis, evaluating connective tissue growth factor (CTGF) and extracellular matrix (ECM) mRNAs and proteins. These effects were attenuated by the 5-HT2A receptor antagonist, ketanserin and mimicked by the 5-HT2A receptor agonist DOI. 5-HT-induced fibroblasts phenotypic alterations and ECM accumulation were dependent on stimulation of transforming growth factor (TGF)-β1 as demonstrated using a neutralizing antibody. 5-HT also caused Smad3 phosphorylation and ketanserin diminished 5-HT-induced Smad3 activation. These results demonstrated that 5-HT can directly activate PAFs through 5-HT2A receptor and promote fibroblasts phenotypic alterations and adventitia fibrosis depending on the signaling of the TGF-β1/Smad3 pathway.


Science China-life Sciences | 2009

Proteomic analysis of mitochondria reveals a metabolic switch from fatty acid oxidation to glycolysis in the failing heart

Jun Wang; Ling Bai; Jing Li; ChaoFeng Sun; Jin Zhao; ChangCong Cui; Ke Han; Yu Liu; Xiaozhen Zhuo; Tingzhong Wang; Ping Liu; Fenling Fan; Youfei Guan; Aiqun Ma

This work characterizes the mitochondrial proteomic profile in the failing heart and elucidates the molecular basis of mitochondria in heart failure. Heart failure was induced in rats by myocardial infarction, and mitochondria were isolated from hearts by differential centrifugation. Using two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization-time of flight mass spectrometry, a system biology approach was employed to investigate differences in mitochondrial proteins between normal and failing hearts. Mass spectrometry identified 27 proteins differentially expressed that involved in energy metabolism. Among those, the up-regulated proteins included tricarboxylic acid cycle enzymes and pyruvate dehydrogenase complex subunits while the down-regulated proteins were involved in fatty acid oxidation and the OXPHOS complex. These results suggest a substantial metabolic switch from free fatty acid oxidation to glycolysis in heart failure and provide molecular evidence for alterations in the structural and functional parameters of mitochondria that may contribute to cardiac dysfunction during ischemic injury.


Journal of Geriatric Cardiology | 2012

Improvement of cardiac function and reversal of gap junction remodeling by Neuregulin-1β in volume-overloaded rats with heart failure

Xuehui Wang; Xiaozhen Zhuo; Yajuan Ni; Min Gong; Tingzhong Wang; Qun Lu; Aiqun Ma

Objective We performed experiments using Neuregulin-1β (NRG-1β) treatment to determine a mechanism for the protective role derived from its beneficial effects by remodeling gap junctions (GJs) during heart failure (HF). Methods Rat models of HF were established by aortocaval fistula. Forty-eight rats were divided randomly into the HF (HF, n = 16), NRG-1β treatment (NRG, n = 16), and sham operation (S, n = 16) group. The rats in the NRG group were administered NRG-1β (10 µg/kg per day) for 7 days via the tail vein, whereas the other groups were injected with the same doses of saline. Twelve weeks after operation, Connexin 43 (Cx43) expression in single myocytes obtained from the left ventricle was determined by immunocytochemistry. Total protein was extracted from frozen left ventricular tissues for immunoblotting assay, and the ultrastructure of myocytes was observed by transmission electron microscopy. Results Compared with the HF group, the cardiac function of rats in the NRG group was markedly improved, irregular distribution and deceased Cx43 expression were relieved. The ultrastructure of myocytes was seriously damaged in HF rats, and NRG-1β reduced these pathological damages. Conclusions Short-term NRG-1β treatment can rescue pump failure in experimental models of volume overload-induced HF, which is related to the recovery of GJs structure and the improvement of Cx43 expression.


Molecular Pharmacology | 2014

Muscarinic Receptor Activation Increases hERG Channel Expression through Phosphorylation of Ubiquitin Ligase Nedd4-2

Tingzhong Wang; Andrew Hogan-Cann; Yudi Kang; Zhi Cui; Jun Guo; Tonghua Yang; Shawn M. Lamothe; Wentao Li; Aiqun Ma; John T. Fisher; Shetuan Zhang

The human ether-à-go-go–related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel, which is important for cardiac repolarization. Reduction of hERG current due to genetic mutations or drug interferences causes long QT syndrome, leading to cardiac arrhythmias and sudden death. To date, there is no effective therapeutic method to restore or enhance hERG channel function. Using cell biology and electrophysiological methods, we found that the muscarinic receptor agonist carbachol increased the expression and function of hERG, but not ether-à-go-go or Kv1.5 channels stably expressed in human embryonic kidney cells. The carbachol-mediated increase in hERG expression was abolished by the selective M3 antagonist 4-DAMP (1,1-dimethyl-4-diphenylacetoxypiperidinium iodide) but not by the M2 antagonist AF-DX 116 (11[[2-[(diethylamino)methyl]-1-piperidinyl]-acetyl]-5,11-dihydro-6H-pyrido[2,3-b] [1,4]benzodiazepine-6-one). Treatment of cells with carbachol reduced the hERG-ubiquitin interaction and slowed the rate of hERG degradation. We previously showed that the E3 ubiquitin ligase Nedd4-2 mediates degradation of hERG channels. Here, we found that disrupting the Nedd4-2 binding domain in hERG completely eliminated the effect of carbachol on hERG channels. Carbachol treatment enhanced the phosphorylation level, but not the total level, of Nedd4-2. Blockade of the protein kinase C (PKC) pathway abolished the carbachol-induced enhancement of hERG channels. Our data suggest that muscarinic activation increases hERG channel expression by phosphorylating Nedd4-2 via the PKC pathway.

Collaboration


Dive into the Tingzhong Wang's collaboration.

Top Co-Authors

Avatar

Aiqun Ma

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Yuan Du

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Ke Han

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Dandan Yang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Ling Luo

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Yajuan Ni

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bingxue Song

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Feifei Ning

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Xiaozhen Zhuo

Xi'an Jiaotong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge