1.The myocardial protective effect of propofol on rats with experimental myocardial infarction and its mechanism.
Ming-Xiao ZHANG ; Qing-Xin TIAN ; Jian-Long LIU
Acta Physiologica Sinica 2021;73(6):878-884
The aim of the present study was to investigate the protective effect of propofol on the experimental myocardial infarction in rats. The myocardial infarction model was established by ligating the anterior descending branch of left coronary artery in rats. Model rats were treated with propofol. Cardiac function was evaluated by echocardiography. Cardiac hemodynamic changes were detected by multiconductor biorecorder. Pathological changes in the infarcted myocardia were detected by HE staining. The expression levels of cardiac hypertrophy marker genes and fibrosis marker proteins were analyzed by real-time quantitative PCR and Western blot. The results showed that, compared with the sham surgery group, the model group exhibited larger infarct size (> 40%), impaired heart function, and significantly increased left ventricular end-diastolic pressure (LVEDP). Propofol reduced cardiac function impairment and decreased LVEDP in the model group. Propofol significantly reduced lung weight/body weight ratio, heart weight/body weight ratio, left ventricular weight/body weight ratio and left atrial weight/body weight ratio in the model group. Furthermore, after myocardial infarction, the administration of propofol significantly improved the diastolic strain rate, down-regulated the mRNA expression levels of myocardial hypertrophy markers, atrial natriuretic peptide and β-myosin heavy chain, and reversed the up-regulation of matrix metalloproteinase 2 (MMP2), MMP9 and tissue inhibitor of metalloproteinase-2 (TIMP-2) induced by myocardial infarction. These results suggest propofol can reduce adverse ventricular remodeling, cardiac dysfunction, myocardial hypertrophy and fibrosis after myocardial infarction, and has protective effect against the experimental myocardial infarction induced by coronary artery ligation in rats.
Animals
;
Cardiotonic Agents/pharmacology*
;
Matrix Metalloproteinase 2
;
Matrix Metalloproteinase 9
;
Myocardial Infarction/drug therapy*
;
Myocardium
;
Propofol/pharmacology*
;
Rats
;
Tissue Inhibitor of Metalloproteinase-2/genetics*
;
Ventricular Remodeling
2.Study on protective effect of vanillic acid from Astragalus membranaceus on hypertensive cardiac remodeling based on network pharmacology screen.
Bo-Yang WANG ; Tian-Long LIU ; Jing LIU ; Ming-Jie ZHANG ; Jian-Jun SUN ; Xiao-Lei LIU ; Rui-Lian MA
China Journal of Chinese Materia Medica 2020;45(2):367-373
To identify and verify the active ingredients from Astragalus membranaceus on hypertensive cardiac remodeling based on network pharmacology and heart RNA-sequencing data. The monomers of A. membranaceus and their intervention target database were established by using network pharmacology. The genes associated to cardiac remodeling were then screened by analyzing cardiac RNA-sequencing data. An overlap between genes related to cardiac remodeling and targets of ingredients form A. membranaceus was collected to obtain monomers with protective effect on hypertensive cardiac remodeling. Angiotensin Ⅱ(AngⅡ)-induced mouse cardiac remodeling model was used to validate the protective effect of active ingredients from A. membranaceus on hypertensive cardiac remodeling. Finally, a total of 81 monomers and 1 197 targets were enrolled in our database. Mouse RNA-sequencing data showed that 983 genes were significantly up-regulated and 465 genes were down-regulation in myocardial tissues of the cardiac remodeling mice as compared with blank group mice, respectively. Ninety-two genes were found via overlapping between genes related to cardiac remodeling and targets, involving 59 monomers from A. membranaceus. Further research found that vanillic acid(VA) could intervene 27 genes associated with hypertensive cardiac remodeling, ranking top 1. Meanwhile, VA could significantly inhibit AngⅡ-induced increase in ratio of heart weight to body weight and heart weight to tibial length, ANP and BNP mRNA levels in myocardial tissues, myocardial tissue damage, cardiac fibrosis level and cardiac hypertrophy level in vivo. Those results showed that network pharmacology screen-based VA has protective effect on AngⅡ-induced cardiac remodeling.
Angiotensin II
;
Animals
;
Astragalus propinquus/chemistry*
;
Heart
;
Hypertension/genetics*
;
Mice
;
Protective Agents/pharmacology*
;
Vanillic Acid/pharmacology*
;
Ventricular Remodeling/genetics*
3.Wild-type p53-induced Phosphatase 1 Deficiency Exacerbates Myocardial Infarction-induced Ischemic Injury.
Ke-Mei LIU ; Hai-Hong ZHANG ; Ya-Nan WANG ; Lian-Mei WANG ; Hong-Yu CHEN ; Cai-Feng LONG ; Lian-Feng ZHANG ; Hong-Bing ZHANG ; Hong-Bing YAN
Chinese Medical Journal 2017;130(11):1333-1341
BACKGROUNDMyocardial infarction (MI) is a major disease burden. Wild-type p53-induced phosphatase 1 (Wip1) has been studied extensively in the context of cancer and the regulation of different types of stem cells, but the role of Wip1 in cardiac adaptation to MI is unknown. We investigated the significance of Wip1 in a mouse model of MI.
METHODSThe study began in June 2014 and was completed in July 2016. We compared Wip1-knockout (Wip1-KO) mice and wild-type (WT) mice to determine changes in cardiac function and survival in response to MI. The heart weight/body weight (HW/BW) ratio and cardiac function were measured before MI. Mouse MI was established by ligating the left anterior descending (LAD) coronary artery under 1.5% isoflurane anesthesia. After MI, survival of the mice was observed for 4 weeks. Cardiac function was examined by echocardiography. The HW/BW ratio was analyzed, and cardiac hypertrophy was measured by wheat germ agglutinin staining. Hematoxylin and eosin (H&E) staining was used to determine the infarct size. Gene expression of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) was assessed by quantitative real-time polymerase chain reaction (qPCR), and the levels of signal transducers and activators of transcription 3 (stat3) and phosphor-stat3 (p-stat3) were also analyzed by Western blotting. Kaplan-Meier survival analysis, log-rank test, unpaired t-test, and one-way analysis of variance (ANOVA) were used for statistical analyses.
RESULTSWip1-KO mice had a marginally increased HW/BW ratio and slightly impaired cardiac function before LAD ligation. After MI, Wip1-deficient mice exhibited increased mortality (57.14% vs. 29.17%; n = 24 [WT], n = 35 [Wip1-KO], P< 0.05), increased cardiac hypertrophy (HW/BW ratio: 7 days: 7.25 ± 0.36 vs. 5.84 ± 0.18, n = 10, P< 0.01, and 4 weeks: 6.05 ± 0.17 vs. 5.87 ± 0.24, n = 10, P > 0.05; cross-sectional area: 7 days: 311.80 ± 8.29 vs. 268.90 ± 11.15, n = 6, P< 0.05, and 4 weeks: 308.80 ± 11.26 vs. 317.00 ± 13.55, n = 6, P > 0.05), and reduced cardiac function (ejection fraction: 7 days: 29.37 ± 1.38 vs. 34.72 ± 1.81, P< 0.05, and 4 weeks: 19.06 ± 2.07 vs. 26.37 ± 2.95, P< 0.05; fractional shortening: 7 days: 13.72 ± 0.71 vs. 16.50 ± 0.94, P< 0.05, and 4 weeks: 8.79 ± 1.00 vs. 12.48 ± 1.48, P< 0.05; n = 10 [WT], n = 15 [Wip1-KO]). H&E staining revealed a larger infarct size in Wip1-KO mice than in WT mice (34.79% ± 2.44% vs. 19.55% ± 1.48%, n = 6, P< 0.01). The expression of IL-6 and p-stat3 was downregulated in Wip1-KO mice (IL-6: 1.71 ± 0.27 vs. 4.46 ± 0.79, n = 6, P< 0.01; and p-stat3/stat3: 1.15 ± 0.15 vs. 1.97 ± 0.23, n = 6, P< 0.05).
CONCLUSIONThe results suggest that Wip1 could protect the heart from MI-induced ischemic injury.
Animals ; Echocardiography ; Interleukin-1beta ; metabolism ; Interleukin-6 ; metabolism ; Male ; Mice ; Mice, Knockout ; Myocardial Infarction ; genetics ; metabolism ; pathology ; Myocytes, Cardiac ; metabolism ; Protein Phosphatase 2C ; deficiency ; genetics ; metabolism ; Real-Time Polymerase Chain Reaction ; Tumor Necrosis Factor-alpha ; metabolism ; Ventricular Remodeling
4.Impact of the beta-1 adrenergic receptor polymorphism on tolerability and efficacy of bisoprolol therapy in Korean heart failure patients: association between beta adrenergic receptor polymorphism and bisoprolol therapy in heart failure (ABBA) study.
Hae Young LEE ; Wook Jin CHUNG ; Hui Kyung JEON ; Hong Seog SEO ; Dong Ju CHOI ; Eun Seok JEON ; Jae Joong KIM ; Joon Han SHIN ; Seok Min KANG ; Sung Cil LIM ; Sang Hong BAEK
The Korean Journal of Internal Medicine 2016;31(2):277-287
BACKGROUND/AIMS: We evaluated the association between coding region variants of adrenergic receptor genes and therapeutic effect in patients with congestive heart failure (CHF). METHODS: One hundred patients with stable CHF (left ventricular ejection fraction [LVEF] < 45%) were enrolled. Enrolled patients started 1.25 mg bisoprolol treatment once daily, then up-titrated to the maximally tolerable dose, at which they were treated for 1 year. RESULTS: Genotypic analysis was carried out, but the results were blinded to the investigators throughout the study period. At position 389 of the beta-1 adrenergic receptor gene (ADRB1), the observed minor Gly allele frequency (Gly389Arg + Gly389Gly) was 0.21, and no deviation from Hardy-Weinberg equilibrium was observed in the genotypic distribution of Arg389Gly (p = 0.75). Heart rate was reduced from 80.8 +/- 14.3 to 70.0 +/- 15.0 beats per minute (p < 0.0001). There was no significant difference in final heart rate across genotypes. However, the Arg389Arg genotype group required significantly more bisoprolol compared to the Gly389X (Gly389Arg + Gly389Gly) group (5.26 +/- 2.62 mg vs. 3.96 +/- 2.05 mg, p = 0.022). There were no significant differences in LVEF changes or remodeling between two groups. Also, changes in exercise capacity and brain natriuretic peptide level were not significant. However, interestingly, there was a two-fold higher rate of readmission (21.2% vs. 10.0%, p = 0.162) and one CHF-related death in the Arg389Arg group. CONCLUSIONS: The ADRB1 Gly389X genotype showed greater response to bisoprolol than the Arg389Arg genotype, suggesting the potential of individually tailoring beta-blocker therapy according to genotype.
Adrenergic beta-1 Receptor Antagonists/adverse effects/*therapeutic use
;
Adult
;
Aged
;
Bisoprolol/adverse effects/*therapeutic use
;
Female
;
Gene Frequency
;
Genotype
;
Heart Failure/diagnosis/*drug therapy/*genetics/physiopathology
;
Heart Rate/drug effects
;
Humans
;
Male
;
Maximum Tolerated Dose
;
Middle Aged
;
Pharmacogenomic Testing
;
Phenotype
;
*Polymorphism, Genetic
;
Precision Medicine
;
Receptors, Adrenergic, beta-1/*drug effects/*genetics
;
Republic of Korea
;
Stroke Volume/drug effects
;
Time Factors
;
Treatment Outcome
;
Ventricular Function, Left/drug effects
;
Ventricular Remodeling/drug effects
5.Calpain mediated pulmonary vascular remodeling in hypoxia induced pulmonary hypertension.
Weifang ZHANG ; Tiantian ZHU ; Aizhen XIONG ; Xiaoyue GE ; Ruilai XU ; Shegui LU ; Changping HU
Journal of Central South University(Medical Sciences) 2016;41(9):929-936
OBJECTIVE:
To explore the role of calpain in pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension and the underlying mechanisms.
METHODS:
Sprague-Dawley rats were randomly divided into the hypoxia group and the normoxia control group. Right ventricular systolic pressure (RVSP) and mean pulmonary artery pressure (mPAP) were monitored by a method with right external jugular vein cannula. Right ventricular hypertrophy index was presented as the ratio of right ventricular weight to left ventricular weight (left ventricle plus septum weight). Levels of calpain-1, -2 and -4 mRNA in pulmonary artery were determined by real-time PCR. Levels of calpain-1, -2 and -4 protein were determined by Western blot. Primary rat pulmonary arterial smooth muscle cells (PASMCs) were divided into 4 groups: a normoxia control group, a normoxia+MDL28170 group, a hypoxia group and a hypoxia+MDL28170 group. Cell proliferation was detected by MTS and flow cytometry. Levels of Ki-67 and proliferating cell nuclear antigen (PCNA) mRNA were determined by real-time PCR.
RESULTS:
RVSP, mPAP and right ventricular remodeling index were significantly elevated in the hypoxia group compared to those in the normoxia group. In the hypoxia group, pulmonary vascular remodeling was significantly developed, accompanied by up-regulation of calpain-1, -2 and -4. MDL28170 significantly inhibited hypoxia-induced proliferation of PASMCs concomitant with the suppression of Ki-67 and PCNA mRNA expression.
CONCLUSION
Calpain mediates vascular remodeling via promoting proliferation of PASMCs in hypoxia-induced pulmonary hypertension.
Animals
;
Calpain
;
genetics
;
physiology
;
Cell Proliferation
;
Dipeptides
;
physiology
;
Hypertension, Pulmonary
;
chemically induced
;
genetics
;
physiopathology
;
Hypertrophy, Right Ventricular
;
Hypoxia
;
Ki-67 Antigen
;
drug effects
;
Myocytes, Smooth Muscle
;
physiology
;
Proliferating Cell Nuclear Antigen
;
drug effects
;
Pulmonary Artery
;
Rats
;
Rats, Sprague-Dawley
;
Real-Time Polymerase Chain Reaction
;
Up-Regulation
;
Vascular Remodeling
;
genetics
;
physiology
6.Hepatocyte growth factor improves right ventricular remodeling in pulmonary arterial hypertensive rats via decreasing neurohormonal activation and inhibiting apoptosis.
Xiaolin WANG ; Yongjin WANG ; Shuling RONG ; Hongbiao MA ; Qing MA ; Junqing ZHAO
Chinese Medical Journal 2014;127(10):1924-1930
BACKGROUNDHepatocyte growth factor (HGF) inhibits the development of pulmonary artery hypertension (PAH) by reducing pulmonary artery pressure and right ventricle (RV) hypertrophy. However, whether HGF can prevent RV remodeling via inhibiting apoptosis in RV cardiomyocytes and decreasing neurohormonal activation remains unknown.
METHODSThe PAH and subsequent RV remodeling in rats were induced by subcutaneous injection of monocrotaline (MCT). The PAH rats were transfected with adenovirus carrying HGF (Ad-HGF) via intratracheal instillation. Three weeks after transfection, the hemodynamics indexes were measured, serum levels for angiotonin II (ANG II) and brain natriuretic peptide (BNP) were determined by ELISA. Histological analysis was used to assess the RV hypertrophy and fibrosis. The cardiomyocyte apoptosis in RV was assayed by TUNEL staining. The mRNA expression of BNP, angiotensin-converting enzyme (ACE), Bax and Bcl-2 in RV was determined by reverse transcriptase polymerase chain reaction (RT-PCR), the protein expression of transforming growth factor (TGF)-β1 and tumor necrosis factor (TNF)-α in RV was determined by Western blotting.
RESULTSHGF treatment significantly decreased the mean PAH, RV systolic pressure, serum ANG II and BNP levels. HGF treatment also significantly decreased the RV hypertrophy, collagen deposition, and the number of apoptotic cardiomyocytes. Moreover, HGF treatmemt significantly decreased the expression of BNP, ACE, Bax, TGF-β1, and TNF-α, while it significantly increased the expression of Bcl-2.
CONCLUSIONSGene transfer of HGF decreases MCT-induced PAH and improves RV remodeling. This effect is mediated not only by improving the hemodynamics but also by decreasing neurohormonal activation and inhibiting cardiomyocytes apoptosis. HGF gene treatment may be an effective strategy for improving RV remodeling in MCT-induced PAH.
Animals ; Apoptosis ; genetics ; physiology ; Hepatocyte Growth Factor ; genetics ; physiology ; therapeutic use ; Humans ; Hypertension, Pulmonary ; metabolism ; therapy ; Male ; Rats ; Rats, Sprague-Dawley ; Ventricular Remodeling ; genetics ; physiology
7.Atorvastatin reduces myocardial fibrosis in a rat model with post-myocardial infarction heart failure by increasing the matrix metalloproteinase-2/tissue matrix metalloproteinase inhibitor-2 ratio.
Zhe AN ; Guang YANG ; Yu-quan HE ; Ning DONG ; Li-li GE ; Shu-mei LI ; Wen-qi ZHANG
Chinese Medical Journal 2013;126(11):2149-2156
BACKGROUNDThe cholesterol-lowering statin drugs have some non-lipid-lowering effects, such as inhibiting myocardial remodeling. However, the underlying mechanism is still unclear.
METHODSThe left anterior descending coronary artery was ligated to establish a rat model of heart failure, and the rats were divided into a sham operation (SO) group, myocardial infarction model (MI) group, and MI-atorvastatin group. Changes in hemodynamic parameters were recorded after the final drug administration. Histological diagnosis was made by reviewing hematoxylin and eosin (HE) stained tissue. Real-time quantitative polymerase chain reaction (PCR) was performed to determine the expressions of type I and type III collagen, matrix metalloproteinase-2 (MMP-2), and tissue matrix metalloproteinase inhibitor-2 (TIMP-2). Further, primary rat cardiac fibroblasts were cultured and the MTT assay was performed to determine the effect of atorvastatin on cardiac fibroblast proliferation.
RESULTSThe model of heart failure was established and the results of HE staining and Masson's trichrome staining revealed that the rats in the heart failure group showed obvious hyperplasia of fibrotic tissue, which was significantly reduced in the atorvastatin group. Real-time quantitative PCR showed that the MI group showed a significantly increased expression of type I and type III collagen, MMP-2, and TIMP-2, but a significantly reduced MMP-2/TIMP-2 ratio. Compared with the MI group, the atorvastatin group showed significantly reduced expression of type I and III collagen, unchanged expression of MMP-2, significantly reduced expression of TIMP-2, and an increased MMP-2/TIMP-2 ratio. We further found that atorvastatin significantly inhibited the Ang II-induced fibroblast proliferation and the expression of type I and type III collagen in cardiac fibroblasts while increasing the MMP-2/TIMP-2 ratio.
CONCLUSIONSThese data suggest that atorvastatin can inhibit cardiac fibroblast proliferation and enhance collagen degradation by increasing the MMP-2/TIMP-2 ratio, thereby inhibiting the formation of myocardial fibrosis in rats with heart failure after myocardial infarction.
Animals ; Atorvastatin Calcium ; Collagen ; biosynthesis ; Disease Models, Animal ; Female ; Fibrosis ; Heart Failure ; drug therapy ; pathology ; Heptanoic Acids ; pharmacology ; therapeutic use ; Hydroxymethylglutaryl-CoA Reductase Inhibitors ; pharmacology ; Matrix Metalloproteinase 2 ; genetics ; Myocardial Infarction ; complications ; Myocardium ; pathology ; Pyrroles ; pharmacology ; therapeutic use ; Rats ; Rats, Wistar ; Tissue Inhibitor of Metalloproteinase-2 ; genetics ; Ventricular Remodeling ; drug effects
8.Lipopolysaccharide induced activin A-follistatin imbalance affects cardiac fibrosis.
Wen-Qi ZHANG ; Chun-Yan YANG ; Shu-Mei LI ; Miao LIU ; Mei DING ; Guo-Hui LIU ; Ping YANG
Chinese Medical Journal 2012;125(12):2205-2212
BACKGROUNDInflammation plays a pivotal role in cardiac remodeling, especially in myocardial fibrosis. Abnormal growth of cardiac fibroblasts is critically involved in the pathophysiology of cardiac hypertrophy/remodeling. Previous study has demonstrated that many inflammation stimulating factors trigger transforming growth factor-β (TGF-β) induction and reactive myocardial fibrosis. Activin A (ACT A) is a member of TGF-β superfamily, and follistatin (FS) is an activin-binding protein, i.e. an antagonist of ACT A. Our previous studies have shown that ACT A-FS imbalance occurs in rats with heart failure (HF), and overexpression of ACT A can lead to ventricular remodeling, and resultant HF. Low expression of FS after myocardial infarction further exacerbated HF. The pathogenic change resulting from overexpression of ACT A is consistent with that of overexpression of angiotensin II (AngII). Ventricular remodeling includes cardiocyte remodeling and myocardial interstitial collagen deposition and fibrosis. Therefore, the present study was designed to investigate the effects of inflammatory factors on the ACT A-FS and the secretions of cardiac fibroblasts in order to explore in depth the mechanism of myocardial fibrosis.
METHODSA rat model with HF was established, and the results showed that there was a greater degree of cardiac fibrosis in HF rats. In addition, we found that there was an imbalance of the ACT A/FS system in HF rats, which was characterized by increased levels of ACT A. Further, primary rat cardiac fibroblasts were cultured and the MTT assay was performed to determine the effect of the inflammatory factor-bacterial endotoxin lipopolysaccharide (LPS) on cardiac fibroblast proliferation.
RESULTSThe results showed that LPS can stimulate the cardiac fibroblasts to proliferate in a dose-dependent manner. Cellular immunohistochemical staining showed that the rat cardiac fibroblasts themselves could express ACT A and FS proteins, and stimulation by LPS could apparently promote the cultured primary rat cardiac fibroblasts to secrete ACT A, but inhibit the secretion of FS. The results also showed that ACT A promoted, in a dose-dependent manner, the proliferation of the cultured primary rat cardiac fibroblasts, and the expression of collagen types I and III. Moreover, ACT A promoted, in a dose dependent manner, the cardiac fibroblasts to secrete nitric oxide (NO), and unregulated the expression of inducible nitric oxide synthase (iNOS) mRNA.
CONCLUSIONSThese results suggest that the inflammatory mediator LPS can promote ACT A-FS imbalance in cardiac fibroblasts, mainly overexpression of ACT A. Overexpression of ACT A promotes the proliferation and the secretion of collagens in cardiac fibroblasts through autocrine/paracrine stimulation of NO, and is involved in the pathological process of myocardial fibrosis.
Activins ; genetics ; metabolism ; Animals ; Cell Proliferation ; Cells, Cultured ; Enzyme-Linked Immunosorbent Assay ; Female ; Fibroblasts ; cytology ; drug effects ; Follistatin ; genetics ; metabolism ; Immunohistochemistry ; Lipopolysaccharides ; pharmacology ; Myocardium ; cytology ; Nitric Oxide ; metabolism ; Rats ; Rats, Wistar ; Real-Time Polymerase Chain Reaction ; Ventricular Remodeling ; drug effects
9.AKT-modified autologous intracoronary mesenchymal stem cells prevent remodeling and repair in swine infarcted myocardium.
Yun-Sheng YU ; Zhen-Ya SHEN ; Wen-Xue YE ; Hao-Yue HUANG ; Fei HUA ; Yi-Huan CHEN ; Ke CHEN ; Wei-Jie LAO ; Li TAO
Chinese Medical Journal 2010;123(13):1702-1708
BACKGROUNDTransplantation of adult bone marrow-derived mesenchymal stem cells (MSCs) has been proposed as a strategy for cardiac repair following myocardial damage. However cell transplantation strategies to replace lost myocardium are limited by the inability to deliver large numbers of cells that resist peritransplantation graft cell death. Accordingly, we set out to isolate and expand adult swine bone marrow-derived MSCs, and to engineer these cells to overexpress AKT1 (protein kinase B), to test the hypothesis that AKT1-engineered MSCs are more resistant to apoptosis and can enhance cardiac repair after transplantation into the ischemic swine heart.
METHODSThe CDS (regulation domain of AKT1) AKT1-cDNA fragment was amplified, and MSCs were transfected following synthesis with a pCDH1-AKT1 shuttling plasmid. Western blotting analysis and real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed. Myocardial infarction (MI) models were constructed in Meishan pigs, and cardiac function was evaluated by magnetic resonance imaging (MRI) measurements and echocardiography 4 weeks later. All pigs were assigned to four groups: control (A), DMEM (B), MSC (C), and AKT-transfected (D). MSCs were transfected with the AKT1 gene, and autologous BrdU-labeled stem cells (1 x 10(7)/5 ml) were injected into left anterior descending coronary atery (LAD) of the infarct heart in groups C and D. In group B, DMEM was injected using the same approach. In group A, there was no injection following LAD occlusion. After 4 weeks, cardiac function and regional perfusion measurements were repeated by MRI and echocardiography, and histological characteristics of the hearts were assessed. Connecxin-43 (CX-43), BrdU, and von Willebrand factor (VWF) immunoreactivity was tested using enzyme linked immunosorbent assay (ELISA). Vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1) were analyzed at the same time.
RESULTSAKT1-cDNA was cloned into pCDH1-MCS1-EF1-copGFP and the sequence was confirmed. AKT mRNA expression was detected at 24 hours after transfection. AKT1 expression in MSCs remained strong after 2 weeks, according to real-time RT-PCR and Western blotting. Prior to cell implantation, end-diastolic left ventricular dimension (EDLVd) increased and stroke volume (SV) decreased in the MI hearts. MRI scans revealed significantly improved cardiac function following implantation, and implanted MSCs prevented thinning and expanding in the infarct region, as well as improved contraction and increased perfusion in all groups compared to control hearts. The left ventricular chamber size was smaller in cell-transplanted hearts than in control hearts. Moreover, group D exhibited significant improvement. The expression of CX-43, BrdU, and VWF could be found in the immunohistochemical pathological sections of group C and group D. The level of VEGF reached a high level 1 week after implanting the MSCs, but the level of TGF-beta1 decreased gradually.
CONCLUSIONSThe AKT1-expressing lentiviral vector resulted in stable over-expression of AKT1 in MSCs. MSC engraftment in host myocardium improved cardiac function by attenuating contractile dysfunction and pathological thinning of the infracted left ventricular wall, which likely resulted from myocardial regeneration and angiogenesis.
Animals ; Blotting, Western ; Disease Models, Animal ; Electrophoresis ; Genetic Vectors ; genetics ; Lentivirus ; genetics ; Mesenchymal Stem Cell Transplantation ; Mesenchymal Stromal Cells ; metabolism ; Myocardial Infarction ; metabolism ; therapy ; Proto-Oncogene Proteins c-akt ; genetics ; metabolism ; Reverse Transcriptase Polymerase Chain Reaction ; Swine ; Ventricular Remodeling ; physiology
10.Effect of adenovirus-mediated stromal cell-derived factor-alpha gene transfer on ventricular remodeling in rats with myocardial infarction.
Jian-ye YANG ; Ying-chun ZHANG ; Jun-ming TANG ; Qing-bao AN ; Ling-yun GUO ; Xia KONG ; Yong-zhang HUANG ; Fei ZHENG ; Jia-ning WANG
Journal of Southern Medical University 2010;30(1):38-42
OBJECTIVETo explore the effect of adenovirus-mediated human stromal cell-derived factor-1alpha (hSDF-1alpha) on ventricular remodeling in rats with myocardial infarction.
METHODSA recombinant adenoviral plasmid containing hSDF-1alpha cDNA was constructed using homologous recombination in bacteria and the recombinant adenovirus particles expressing hSDF-1alpha (AdV-SDF-1) were prepared. In rat models of myocardial infarction induced by left anterior descending artery occlusion, 1x10(10) PFU AdV-SDF-1 or PFU AdV-LacZ were injected at multiple sites into the infarcted myocardium 1 h after the operation, using 200 l cell-free PBS as the control. Four weeks after the injection, the cardiac function of the rats was analyzed, and the heart tissues were taken after the measurement of hemodynamics. On serial frozen sections, histological observation and morphometric measurement were carried out using a microscopic image analysis system, and the expression of hSDF-1alpha was detected by immunocytochemistry.
RESULTSFour weeks after AdV-SDF-1 injection, the myocardium in the infracted area showed significantly higher expression rates of hSDF-1alpha. The injection resulted in a obvious reduction in the infarct size and collagen content and a marked increase in the left ventricle wall, and the rats showed improved cardiac functions.
CONCLUSIONSDF-1alpha can improve the cardiac structure and function in rats with myocardial infarction by inhibiting collagen synthesis and deposition in the infarcted area.
Adenoviridae ; genetics ; metabolism ; Animals ; Chemokine CXCL12 ; administration & dosage ; biosynthesis ; genetics ; Female ; Gene Transfer Techniques ; Genetic Vectors ; genetics ; Male ; Myocardial Infarction ; physiopathology ; therapy ; Rats ; Rats, Sprague-Dawley ; Recombinant Proteins ; administration & dosage ; biosynthesis ; genetics ; Transfection ; Ventricular Remodeling ; drug effects

Result Analysis
Print
Save
E-mail