1.Aconitine ameliorates cardiomyocyte hypertrophy induced by angiotensin Ⅱ.
Ning-Ning WANG ; Jia WANG ; Hong-Ling TAN ; Yu-Guang WANG ; Yue GAO ; Zeng-Chun MA
China Journal of Chinese Materia Medica 2019;44(8):1642-1647
This paper was aimed to investigate the inhibitory effect of aconitine(AC) on angiotensin Ⅱ(Ang Ⅱ)-induced H9 c2 cell hypertrophy and explore its mechanism of action. The model of hypertrophy was induced by Ang Ⅱ(1×10-6 mol·L-1),and cardiomyocytes were incubated with different concentrations of AC. Western blot was used to quantify the protein expression levels of atrial natriuretic peptide(ANP),brain natriuretic peptide(BNP),β-myosin heavy chain(β-MHC),and α-smooth muscle actin(α-SMA). Real-time quantitative PCR(qRT-PCR) was used to quantify the mRNA expression levels of cardiac hypertrophic markers ANP,BNP and β-MHC. In addition,the fluorescence intensity of the F-actin marker,an important component of myofibrils,was detected by using laser confocal microscope. AC could significantly reverse the increase of total protein content in H9 c2 cells induced by Ang Ⅱ; qRT-PCR results showed that AC could significantly inhibit the ANP,BNP and β-MHC mRNA up-regulation induced by AngⅡ. Western blot results showed that AC could significantly inhibit the ANP,BNP and β-MHC protein up-regulation induced by AngⅡ. In addition,F-actin expression induced by Ang Ⅱ could be inhibited by AC,and multiple indicators of cardiomyocyte hypertrophy induced by Ang Ⅱ could be down-regulated,indicating that AC may inhibit cardiac hypertrophy by inhibiting the expression of hypertrophic factors,providing new clues for exploring the cardiovascular protection of AC.
Aconitine
;
pharmacology
;
Actins
;
metabolism
;
Angiotensin II
;
Atrial Natriuretic Factor
;
metabolism
;
Cardiac Myosins
;
metabolism
;
Cardiomegaly
;
Cells, Cultured
;
Humans
;
Hypertrophy
;
Myocytes, Cardiac
;
drug effects
;
Myosin Heavy Chains
;
metabolism
;
Natriuretic Peptide, Brain
;
metabolism
2.Mutation and clinical relevance in a large cohort of unrelated Chinese patients with hypertrophic cardiomyopathy.
Liu JIE ; Liu WENLING ; Hu DAYI ; Zhu TIANGANG ; Liu WEN ; Ma ZHANFENG ; Yang JIE ; Xie WENLI ; Li CUILAN ; Li LEI ; Pan GUOZHONG
Chinese Journal of Cardiology 2015;43(8):682-689
OBJECTIVETo explore the genetic basis and phenotypic correlation with disease severity in a large cohort of Chinese patients with hypertrophic cardiomyopathy (HCM).
METHODSA total of 179 unrelated Chinese HCM patients admitted to our department from 2002 to 2011 were enrolled in this study. Direct gene sequencing of β-myosin heavy chain (MYH7), myosin binding protein-C ( MYBPC3), and cardiac troponin T (TNNT2) were performed and clinical data were obtained in these patients.
RESULTSA total of 34 mutations were identified in 40 patients (22.3%), 79.4% (27/34) mutations occurred only once and a possible hot spot, A26 in MYH7, was found. Distribution of mutations was 52.9% (18/34) (MYBPC3), 35.3% (12/34) ( MYH7) and 11.8% (4/34) (TNNT2) respectively. Double mutations were identified in 2.2% (4/179) patients. Genotype-positive patients were associated with an earlier symptom onset, severer left ventricular hypertrophy, a higher incidence of syncope, and were more likely to have positive family history of HCM or sudden cardiac death (SCD) , and were more likely to progress into heart failure (24.2% vs. 5.0%, P = 0.002) and at a higher risk of SCD (9.1% vs. 0, P = 0.009) during the 6.5-year follow-up. No statistical difference in any clinical parameters and outcomes was found between patients carrying MYBPC3 and MYH7 mutations. Double mutations were associated with malignant clinical progression in this cohort. Different phenotype severity could be seen in HCM patients with same genotype (e. g. MYH7-1736T, TNNT2-R92W).
CONCLUSIONMYBPC3 is the most predominant gene mutation in this HCM cohort. The presence of a sarcomere mutation in patients with HCM is associated with poor clinical outcome, although no specific genes or mutations can exactly predict the severity of clinical phenotypes.
Asian Continental Ancestry Group ; Cardiomyopathy, Hypertrophic ; Carrier Proteins ; Death, Sudden, Cardiac ; Disease Progression ; Genotype ; Humans ; Hypertrophy, Left Ventricular ; Mutation ; Phenotype ; Sarcomeres ; Troponin T ; Ventricular Myosins
3.Enhanced Cardiomyogenic Differentiation of P19 Embryonal Carcinoma Stem Cells.
Jihyun YANG ; Seok Jin KO ; Beom Suk KIM ; Hyun Seo KIM ; Sangheon PARK ; Doran HONG ; Soon Woong HONG ; Ji Hyun CHOI ; Chi Yeon PARK ; Seung Cheol CHOI ; Sun Jun HONG ; Do Sun LIM
Korean Circulation Journal 2009;39(5):198-204
BACKGROUND AND OBJECTIVES: We investigated the effects of different concentrations of serum, 5-azacytidine, and culture time on the cardiomyogenic differentiation of P19 embryonal carcinoma stem cells in the course of developing an efficient protocol for generating the cardiomyogenic lineage. MATERIALS AND METHODS: P19 cells were plated at a density of 1x10(6) cells on 10-cm bacterial dishes for 96 hours in the presence of 1% dimethyl sulfoxide to form embryoid bodies. The embryoid bodies were cultured in medium with 2% or 10% fetal bovine serum for an additional 10 or 15 consecutive days in the presence of 0, 1, or 3 microM 5-azacytidine. RESULTS: Quantitative real-time polymerase chain reaction (PCR) analysis showed that the messenger ribonucleic acid (mRNA) expression of cardiac muscle-specific genes, such as GATA4, alpha-actin, alpha-myosin heavy chain, and cardiac troponin T, were significantly higher in the 15-day culture groups than in the 10-day culture groups. Furthermore, the cardiac muscle-specific genes were expressed more in the high-serum groups compared to the low-serum groups regardless of the culture time. Cardiomyogenic differentiation of the P19 cells was most effective in 1 microM 5-azacytidine regardless of the serum concentrations. In addition, the stimulation effects of 5-azacytidine on cardiomyogenic differentiation were more significant under low-serum culture conditions compared to high-serum culture conditions. Cardiomyogenic differentiation of P19 cells was further confirmed by immunostaining with cardiac muscle-specific antibodies. CONCLUSION:Taken together, these results demonstrated that cardiomyogenic differentiation of P19 cells was enhanced by a combination of different experimental factors.
Actins
;
Antibodies
;
Azacitidine
;
Carcinoma, Embryonal
;
Cell Differentiation
;
Dimethyl Sulfoxide
;
Embryoid Bodies
;
Embryonal Carcinoma Stem Cells
;
Myocytes, Cardiac
;
Real-Time Polymerase Chain Reaction
;
RNA
;
Safrole
;
Troponin T
;
Ventricular Myosins
4.Effects of Ischemic Preconditioning on the Left Ventricular Function, Myosin Isozyme, Myofibrillar ATPase Activity and Ultrastructure of the Rabbit Heart.
Dae joong KIM ; Yeon Jung YOON ; Ho dirk KIM ; Bong jin RAH
Korean Circulation Journal 1995;25(3):665-675
BACKGROUND: Single or multiple episodes of brief period of ischemia and reperfusion(ischemic preconditioning, IP) have been shown to limit infarct size after a subsequent longer period of ischemia. A considerable number of possible mechanisms has been proposed, however, controversies still remain. Accordingly, we evaluated the effect of four cycles of 5 minutes ischemia and 5 minutes reperfusion(IP) followed by subsequent 30 minutes ischemia(ISCH) and 60 minutes reperfusion using isolated Langendorff-Perfused rabbit hearts. Methods and RESULTS: After a 50-minute recovery phase, parameters of the left ventricular function(LVF) including left ventricular developed pressure(LVDP), contractility and the heart rate were recorded, and ultrastructure was examined. Myosin ATPase activity was determined by measurement of inorganic phosphorus and isozymes of the myosin heavy chain were examined by polyacrylamide gel electrophoresis containing pyrophosphate buffer. The ISCH hearts showed severe to irreversible change of the cardiac myocytes homogenously in contrast to the IP hearts in which changes were not homogenous and irreversible injury was only focal. However, parameters of the LVF were not significantly different between the IP and the ISHC hearts during reperfusion. Myosin ATPase activities were also not significantly different(0.67+/-0.123 micromol/mg protein/h in the IP hearts, 0.56+/-0.172 micromol/mg protein/h in the ISCH hearts, and 0.76+/-0.239 micromol/mg protein/h in the control hearts). Band patterns of the myofibrillar proteins, separated by sodium ddodecyl sulfate-polyacrylamide gel electrophoresis, revealed no differences between the IP, ISCH and the control hearts. Myosin heavy chains in the IP and the ISCH hearts were separated into 3 isozymes, V1,V2and V3in pyrophosphate gel electrophoresis in contrast that the control hearts revealed two isozymes, V1and V2. However, there were no differences in the protein composition and electrophoretic motility between the IP and the ISCH hearts. CONCLUSION: These results indicate that IP could not attenuate the changes in LVF, myosin ATPase activity and myosin isozymes on reperfusion, however, it could attenuate the ultrastructural changes of the cardiac myocytes.
Adenosine Triphosphatases*
;
Electrophoresis
;
Electrophoresis, Polyacrylamide Gel
;
Heart Rate
;
Heart*
;
Ischemia
;
Ischemic Preconditioning*
;
Isoenzymes
;
Myocytes, Cardiac
;
Myosin Heavy Chains
;
Myosins*
;
Phosphorus
;
Reperfusion
;
Sodium
;
Ventricular Function, Left*
5.Pro731Ser mutation in the β-myosin heavy chain and hypertrophic cardiomyopathy in a Chinese pedigree.
Xintao ZHAO ; Yajie WU ; Yi CHEN ; Xinxing FENG ; Ying SONG ; Yilu WANG ; Yubao ZOU ; Jizheng WANG ; Yibing SHAO ; Rutai HUI ; Lei SONG ; Xu WANG
Chinese Journal of Cardiology 2014;42(7):571-576
OBJECTIVETo identify the casual mutation of a Chinese pedigree with hypertrophic cardiomyopathy (HCM), and to analyze the genotype-phenotype relationship.
METHODSThe coding exons of 26 reported disease genes were sequenced by targeted resequencing in the proband and the identified mutation were detected with bi-directional Sanger sequencing in all family members and 307 healthy controls. The genotype-phenotype correlation was analyzed in the family.
RESULTSA missense mutation (c.2191C > T, p. Pro731Ser) in the 20th exon of MYH7 gene was identified. This mutation was absent in 307 healthy controls and predicted to be pathogenic by PolyPhen-HCM. Totally 13 family members carried this mutation, including 10 patients with HCM and 3 asymptomatic mutation carriers. The proband manifested severe congestive heart failure and 8 patients expressed various clinical manifestations of heart failure, including dyspnea, palpitations, chest pain, amaurosis or syncope. Five patients were diagnosed as HCM at the age of 16 or younger. One family member suffered sudden cardiac death.
CONCLUSIONSThe Pro731Ser of MYH7 gene mutation is a causal and malignant mutation linked with familiar HCM.
Adolescent ; Asian Continental Ancestry Group ; Base Sequence ; Cardiomyopathy, Hypertrophic ; ethnology ; genetics ; Death, Sudden, Cardiac ; Exons ; Humans ; Mutation, Missense ; Myosin Heavy Chains ; genetics ; Pedigree ; Phenotype ; Research Design ; Ventricular Myosins
6.Analysis of phenotype and MYH7 gene variant in a family of patients with hypertrophic cardiomyopathy.
Xueli ZHAO ; Bo WANG ; Xiaoli ZHU ; Qianli YANG ; Ying LIU ; Hong SHAO ; Lei ZUO ; Yun LUO ; Yue WANG ; Liwen LIU
Chinese Journal of Medical Genetics 2022;39(8):873-876
OBJECTIVE:
To analyze the clinical phenotype and MYH7 gene variant in a Chinese pedigree affected with hypertrophic cardiomyopathy (HCM).
METHODS:
The proband was screened for variant of 96 cardiomyopathy-associated genes by exonic amplification and high-throughput sequencing. Candidate variant was verified by Sanger sequencing among 300 healthy controls as well as family members of the proband. Co-segregation analysis of genotypes and clinical phenotypes was carried out for the pedigree. Clustal X software was used to analyze the sequence conservation of the variant among various species, and its pathogenicity was predicted by using bioinformatics software.
RESULTS:
6 out of 12 members from this pedigree were found to harbor heterozygous c.4124A>G (p.Tyr1375Cys) variant of the MYH7 gene, among whom five were diagnosed with HCM. The remaining one had failed to meet the diagnostic criteria for HCM, but had abnormal ECG. The same variant was not found in the 300 healthy controls. Amino acid sequence analysis showed that the variant is located in a highly conserved region, and bioinformatics analysis predicted that this variant may affect protein function and has a deleterious effect. Based on the American College of Medical Genetics and Genomics (ACMG) guidelines, the variant was predicted to be likely pathogenic (PM2+ PP1_Moderate+PP3+PP5).
CONCLUSION
The c.4124A>G (p.Tyr1375Cys) variant of the MYH7 gene probably underlay the pathogenesis in this pedigree. Above finding has important value for the early diagnosis of patients with HCM.
Cardiac Myosins/genetics*
;
Cardiomyopathy, Hypertrophic/genetics*
;
Genotype
;
Humans
;
Mutation
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype
7.The role of three-dimensional speckle tracking imaging derived parameters on predicting outcome of hypertrophic cardiomyopathy patients with MYH7 mutations.
Jie ZHAO ; Jing WANG ; Li Wen LIU ; Yu ZHENG ; Bo WANG ; Wen Xia LI ; Fan YANG ; Nan KANG ; Lei ZUO
Chinese Journal of Cardiology 2020;48(4):287-293
Objective: To evaluate the cardiac functional changes in hypertrophic cardiomyopathy(HCM) patients with β-myosin heavy chain gene (MYH7) mutations by three-dimensional (3D) speckle tracking imaging(3D-STI) and conventional echocardiography modalities, and then to explore the potential predictors of adverse cardiovascular events in these patients. Methods: A consecutive series of 192 HCM patients admitted in our center from October 2014 to October 2016 were genetically screened to identify MYH7 mutations in this retrospective study. A total of 43 HCM patients with MYH7 mutations were enrolled. The patients were divided into events group(n=13) and no event group(n=30) according to the presence or absence of adverse cardiovascular events(primary and secondary endpoints). All patients were followed up to January 2019 after comprehensive evaluation of 3D-STI, two-dimensional and Doppler echocardiography. The adverse cardiovascular events were recorded. Results: The median follow up time was 1 012 (812, 1 330) days. During follow-up, 13 patients (30.2%) reached endpoints: 6 cases of the primary endpoints(2 cases of sudden cardiac death(SCD), 3 cases of survival after defibrillation, and 1 case of appropriate implantable cardioverter-defibrillator(ICD) discharge); 7 cases of the second endpoints(5 cases of heart failure hospitalization, 1 case of syncope and cardioversion due to supraventricular tachycardia, and 1 case of end-stage HCM). Patients with adverse cardiovascular events had higher prevalence of syncope and risk of SCD, enlarged left atrial volume index(LAVI) and reduced 3D left ventricular global longitudinal train (3D-GLS), as compared to those without adverse events(all P<0.05). The multivariate Cox regression analysis showed that reduced 3D-GLS(HR=0.814, 95%CI 0.663-0.999, P=0.049) was an independent predictor for adverse cardiovascular events. The cutoff value of 3D-GLS≤13.67% was linked with significantly increased risk of adverse cardiovascular events in this patient cohort(AUC=0.753, 95%CI 0.558-0.948, sensitivity 86%, specificity 69%, P<0.05). The Kaplan-Meier analysis indicated that the patients with the 3D-GLS≤ 13.67% faced higher risk of death than those with 3D-GLS>13.67%. Conclusion: 3D-GLS is useful on predicting adverse cardiovascular events in HCM patients with MYH7 mutations.
Cardiac Myosins/genetics*
;
Cardiomyopathy, Hypertrophic/genetics*
;
Echocardiography
;
Humans
;
Mutation
;
Myosin Heavy Chains/genetics*
;
Predictive Value of Tests
;
Retrospective Studies
;
Risk Factors
8.Recent research on childhood hypertrophic cardiomyopathy caused by MYH7 gene mutations.
Kui ZHENG ; Lu LIU ; Ying-Qian ZHANG
Chinese Journal of Contemporary Pediatrics 2023;25(4):425-430
Hypertrophic cardiomyopathy (HCM) is the most common monogenic inherited myocardial disease in children, and mutations in sarcomere genes (such as MYH7 and MYBPC3) are the most common genetic etiology of HCM, among which mutations in the MYH7 gene are the most common and account for 30%-50%. MYH7 gene mutations have the characteristics of being affected by environmental factors, coexisting with multiple genetic variations, and age-dependent penetrance, which leads to different or overlapping clinical phenotypes in children, including various cardiomyopathies and skeletal myopathies. At present, the pathogenesis, course, and prognosis of HCM caused by MYH7 gene mutations in children remain unclear. This article summarizes the possible pathogenesis, clinical phenotype, and treatment of HCM caused by MYH7 gene mutations, in order to facilitate the accurate prognostic evaluation and individualized management and treatment of the children with this disorder.
Child
;
Humans
;
Cardiomyopathy, Hypertrophic/therapy*
;
Phenotype
;
Troponin T/genetics*
;
Mutation
;
Carrier Proteins/genetics*
;
Myosin Heavy Chains/genetics*
;
Cardiac Myosins/genetics*
9.Clinical phenotype and genetic analysis of patients with left ventricular noncompaction caused by the biallelic mutation of MYBPC3 and MYH7.
Ya Hui ZHANG ; Xiao Yan LI ; Bang Rong SONG ; Yue Li WANG ; Jun Rui ZHANG ; Yan Long REN
Chinese Journal of Cardiology 2023;51(11):1160-1165
Objective: To explore the relationship between pathogenic gene, mutation and phenotype of left ventricular noncompaction (LVNC) patients and their family members. Methods: The subjects were the proband with LVNC and her family members. The medical history including electrocardiogram, echocardiography and cardiac magnetic resonance examination of the proband and family members were collected. Whole exome sequencing of the proband was performed, bioinformatics analysis focused on the genes related to hereditary cardiomyopathy. Candidate pathogenic sites were validated by Sanger sequencing. The clinical interpretation of sequence variants were classified according to American College of Medical Genetics and Genomics (ACMG) guidelines. Results: The proband carried a heterozygous variation of the MYBPC3 gene c.C2827T and the MYH7 gene c.G2221C. The proband's sister carried heterozygous variation of MYBPC3 gene c.C2827T. According to the ACMG guidelines, the variant was determined to be pathogenic. Conclusion: The missense variant of MYBPC3 gene c.C2827T and MYH7 gene c.G2221C are identified from the proband with LVNC and her family member, which provides a genetic basis for clinical diagnosis and genetic counseling of the patients and the family members with LVNC.
Female
;
Humans
;
Cardiac Myosins/genetics*
;
Heart Defects, Congenital
;
Mutation
;
Mutation, Missense
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype
10.Clinical phenotype and genetic analysis of patients with left ventricular noncompaction caused by the biallelic mutation of MYBPC3 and MYH7.
Ya Hui ZHANG ; Xiao Yan LI ; Bang Rong SONG ; Yue Li WANG ; Jun Rui ZHANG ; Yan Long REN
Chinese Journal of Cardiology 2023;51(11):1160-1165
Objective: To explore the relationship between pathogenic gene, mutation and phenotype of left ventricular noncompaction (LVNC) patients and their family members. Methods: The subjects were the proband with LVNC and her family members. The medical history including electrocardiogram, echocardiography and cardiac magnetic resonance examination of the proband and family members were collected. Whole exome sequencing of the proband was performed, bioinformatics analysis focused on the genes related to hereditary cardiomyopathy. Candidate pathogenic sites were validated by Sanger sequencing. The clinical interpretation of sequence variants were classified according to American College of Medical Genetics and Genomics (ACMG) guidelines. Results: The proband carried a heterozygous variation of the MYBPC3 gene c.C2827T and the MYH7 gene c.G2221C. The proband's sister carried heterozygous variation of MYBPC3 gene c.C2827T. According to the ACMG guidelines, the variant was determined to be pathogenic. Conclusion: The missense variant of MYBPC3 gene c.C2827T and MYH7 gene c.G2221C are identified from the proband with LVNC and her family member, which provides a genetic basis for clinical diagnosis and genetic counseling of the patients and the family members with LVNC.
Female
;
Humans
;
Cardiac Myosins/genetics*
;
Heart Defects, Congenital
;
Mutation
;
Mutation, Missense
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype