Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Sci Rep ; 13(1): 16122, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37752166

ABSTRACT

Although systolic function characteristically shows gradual impairment in pressure overload (PO)-evoked left ventricular (LV) hypertrophy (LVH), rapid progression to congestive heart failure (HF) occurs in distinct cases. The molecular mechanisms for the differences in maladaptation are unknown. Here, we examined microRNA (miRNA) expression and miRNA-driven posttranscriptional gene regulation in the two forms of PO-induced LVH (with/without systolic HF). PO was induced by aortic banding (AB) in male Sprague-Dawley rats. Sham-operated animals were controls. The majority of AB animals demonstrated concentric LVH and slightly decreased systolic function (termed as ABLVH). In contrast, in some AB rats severely reduced ejection fraction, LV dilatation and increased lung weight-to-tibial length ratio was noted (referred to as ABHF). Global LV miRNA sequencing revealed fifty differentially regulated miRNAs in ABHF compared to ABLVH. Network theoretical miRNA-target analysis predicted more than three thousand genes with miRNA-driven dysregulation between the two groups. Seventeen genes with high node strength value were selected for target validation, of which five (Fmr1, Zfpm2, Wasl, Ets1, Atg16l1) showed decreased mRNA expression in ABHF by PCR. PO-evoked systolic HF is associated with unique miRNA alterations, which negatively regulate the mRNA expression of Fmr1, Zfmp2, Wasl, Ets1 and Atg16l1.


Subject(s)
Heart Failure, Systolic , MicroRNAs , Male , Rats , Animals , Heart Failure, Systolic/genetics , Rats, Sprague-Dawley , Gene Expression Regulation , Hypertrophy, Left Ventricular , MicroRNAs/genetics , RNA, Messenger , Weight Gain , Fragile X Mental Retardation Protein
2.
Sci Rep ; 12(1): 85, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997065

ABSTRACT

Suture-based transverse aortic constriction (TAC) in mice is one of the most frequently used experimental models for cardiac pressure overload-induced heart failure. However, the incidence of heart failure in the conventional TAC depends on the operator's skill. To optimize and simplify this method, we proposed O-ring-induced transverse aortic constriction (OTAC) in mice. C57BL/6J mice were subjected to OTAC, in which an o-ring was applied to the transverse aorta (between the brachiocephalic artery and the left common carotid artery) and tied with a triple knot. We used different inner diameters of o-rings were 0.50 and 0.45 mm. Pressure overload by OTAC promoted left ventricular (LV) hypertrophy. OTAC also increased lung weight, indicating severe pulmonary congestion. Echocardiographic findings revealed that both OTAC groups developed LV hypertrophy within one week after the procedure and gradually reduced LV fractional shortening. In addition, significant elevations in gene expression related to heart failure, LV hypertrophy, and LV fibrosis were observed in the LV of OTAC mice. We demonstrated the OTAC method, which is a simple and effective cardiac pressure overload method in mice. This method will efficiently help us understand heart failure (HF) mechanisms with reduced LV ejection fraction (HFrEF) and cardiac hypertrophy.


Subject(s)
Aorta/surgery , Heart Failure, Systolic/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Stroke Volume , Ventricular Function, Left , Ventricular Remodeling , Animals , Aorta/physiopathology , Constriction , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Heart Failure, Systolic/etiology , Heart Failure, Systolic/genetics , Heart Failure, Systolic/metabolism , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Male , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology
3.
Cardiovasc Res ; 118(1): 53-64, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33620071

ABSTRACT

It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggests that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. This review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signalling, vascular inflammation and senescence will be discussed.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Coronary Vessels/drug effects , Genetic Therapy , Heart Failure, Diastolic/therapy , Heart Failure, Systolic/therapy , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Vaccines/therapeutic use , Angiogenesis Inducing Agents/adverse effects , Animals , Coronary Circulation/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Genetic Therapy/adverse effects , Heart Failure, Diastolic/genetics , Heart Failure, Diastolic/metabolism , Heart Failure, Diastolic/physiopathology , Heart Failure, Systolic/genetics , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Microcirculation/drug effects , Microvessels/metabolism , Microvessels/physiopathology , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Recovery of Function , Vaccines/adverse effects , Ventricular Function, Left/drug effects
4.
Eur Heart J ; 42(20): 2000-2011, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33677556

ABSTRACT

AIMS: Our objective was to better understand the genetic bases of dilated cardiomyopathy (DCM), a leading cause of systolic heart failure. METHODS AND RESULTS: We conducted the largest genome-wide association study performed so far in DCM, with 2719 cases and 4440 controls in the discovery population. We identified and replicated two new DCM-associated loci on chromosome 3p25.1 [lead single-nucleotide polymorphism (SNP) rs62232870, P = 8.7 × 10-11 and 7.7 × 10-4 in the discovery and replication steps, respectively] and chromosome 22q11.23 (lead SNP rs7284877, P = 3.3 × 10-8 and 1.4 × 10-3 in the discovery and replication steps, respectively), while confirming two previously identified DCM loci on chromosomes 10 and 1, BAG3 and HSPB7. A genetic risk score constructed from the number of risk alleles at these four DCM loci revealed a 3-fold increased risk of DCM for individuals with 8 risk alleles compared to individuals with 5 risk alleles (median of the referral population). In silico annotation and functional 4C-sequencing analyses on iPSC-derived cardiomyocytes identify SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine transporter whose involvement in myocardial dysfunction and DCM is supported by numerous observations in humans and animals. At the 22q11.23 locus, in silico and data mining annotations, and to a lesser extent functional analysis, strongly suggest SMARCB1 as the candidate culprit gene. CONCLUSION: This study provides a better understanding of the genetic architecture of DCM and sheds light on novel biological pathways underlying heart failure.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure, Systolic , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Cardiomyopathy, Dilated/genetics , Chromosomes , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Heart Failure, Systolic/genetics , Humans , Polymorphism, Single Nucleotide/genetics
5.
Gene ; 726: 144136, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31629817

ABSTRACT

Chronic systolic heart failure (CSHF) was a complex syndrome. Recently, vagus nerve stimulation (VNS), a novel treatment method, has emerged for the treatment of CSHF. therefore the aim of this study was to explore the possible mechanism of VNS treatment alleviating CSHF in rats. Firstly, we found after VNS treatment for 72 h, the level of B-type natriuretic peptide in VNS group was lower than that in CSHF group. In addition, VNS treatment induced the elevated left ventricular ejection fraction level, reduced left ventricular end diastolic volume and left ventricular end systolic volume level in VNS group, suggesting a mitigation of CSHF by VNS. Then we found the level of miR-183-3p in CSHF group was much lower than that in VNS group by High-throughput sequencing. The further results indicated that Bcl-2 interacting protein 3 like (BNIP3L) was identified as the target gene of miR-183-3p, and the expression of BNIP3L was notably reduced in rats of VNS group compared with CSHF group. Moreover, the down-regulated expression of miR-183-3p increased BNIP3L-mediated autophagy in rats of CSHF group compared with VNS group. Further mechanism findings demonstrated that up-regulation of miR-183-3p reduced the expression of BNIP3L, while down-regulation of miR-183-3p facilitated the expression of BNIP3L in H9c2 cells. miR-183-3p could also regulate autophagy by targeting BNIP3L in vitro, which was manifested by overexpression of miR-183-3p to inhibit BNIP3L-mediated autophagy. Our data demonstrated that VNS treatment benefited CSHF via the up-regulation of miRNA-183-3p, which reduced the BNIP3L-mediated autophagy, providing a new therapeutic direction for CSHF.


Subject(s)
Autophagy/genetics , Heart Failure, Systolic/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Up-Regulation/genetics , Animals , Down-Regulation/genetics , Male , Rats , Rats, Wistar , Stroke Volume/genetics , Transcriptional Activation/genetics , Vagus Nerve Stimulation/methods , Ventricular Function, Left/genetics
6.
Int J Cardiol ; 303: 80-85, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31757654

ABSTRACT

BACKGROUND: Circulating microRNAs (miRs) have been proposed as potential diagnostic biomarkers in heart failure. Studies investigating the prognostic value of circulating miRs in patients with chronic systolic heart failure (HFrEF) are scarce. The aim of this study was to investigate the prognostic value of circulating miRs in patients with HFrEF. METHODS AND RESULTS: A pathway-focused microRNA array was performed in derivation case-control cohort of 40 patients with HFrEF who died during the follow-up (cases) and 36 survivors (controls). MicroRNA expression profiling revealed significant differential expression of miR-122, miR-126 and miR-423 between cases and controls. In a validation cohort, circulating levels of these 3 miRs were assessed using qPCR in 234 patients with HFrEF. Primary study endpoints were all-cause and cardiovascular mortality. During a median follow-up time of 3.2 years, 76 patients (32.5%) died. Only miR-122 and miR-423 were independent predictors of the primary endpoint with respective hazard ratios per increase of one standard deviation (HR per 1-SD) of 1.14 (95% CI: 1.02-1.29, p = 0.021) and 1.24 (95% CI: 1.09-1.41, p = 0.001). Adding miR-122 to multivariable model including clinical risk factors and NT-proBNP improved net reclassification index (NRI) by 40.4% (p = 0.004), whereas miR-423 improved NRI by 35.3% (p = 0.012). Adding miR-122, but not miR-423, to the same model improved Harrell's C index from 0.78 (95% CI: 0.73-0.83) to 0.81 (95% CI: 0.76-0.86, p = 0.030). CONCLUSION: Circulating miR-122 as a biomarker is predicting all-cause and cardiovascular mortality and improved risk stratification of HFrEF patients. Thus, miR-122 might be a new biomarker for risk assessment in HFrEF.


Subject(s)
Heart Failure, Systolic/genetics , Liver/metabolism , MicroRNAs/genetics , Stroke Volume/physiology , Aged , Biomarkers/blood , Female , Follow-Up Studies , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Male , MicroRNAs/blood , Middle Aged , Prognosis , Risk Factors
7.
Genome Med ; 9(1): 20, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28228157

ABSTRACT

Heart failure is a major health burden, affecting 40 million people globally. One of the main causes of systolic heart failure is dilated cardiomyopathy (DCM), the leading global indication for heart transplantation. Our understanding of the genetic basis of both DCM and systolic heart failure has improved in recent years with the application of next-generation sequencing and genome-wide association studies (GWAS). This has enabled rapid sequencing at scale, leading to the discovery of many novel rare variants in DCM and of common variants in both systolic heart failure and DCM. Identifying rare and common genetic variants contributing to systolic heart failure has been challenging given its diverse and multiple etiologies. DCM, however, although rarer, is a reasonably specific and well-defined condition, leading to the identification of many rare genetic variants. Truncating variants in titin represent the single largest genetic cause of DCM. Here, we review the progress and challenges in the detection of rare and common variants in DCM and systolic heart failure, and the particular challenges in accurate and informed variant interpretation, and in understanding the effects of these variants. We also discuss how our increasing genetic knowledge is changing clinical management. Harnessing genetic data and translating it to improve risk stratification and the development of novel therapeutics represents a major challenge and unmet critical need for patients with heart failure and their families.


Subject(s)
Cardiomyopathy, Dilated/genetics , Genetic Predisposition to Disease , Genetic Variation , Heart Failure, Systolic/genetics , Animals , Cardiomyopathy, Dilated/diagnosis , Disease Management , Genomics , Heart Failure, Systolic/diagnosis , Humans
8.
Sci Rep ; 7: 39335, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045017

ABSTRACT

Adhesive intercellular connections at cardiomyocyte intercalated disks (IDs) support contractile force and maintain structural integrity of the heart muscle. Disturbances of the proteins at IDs deteriorate cardiac function and morphology. An adaptor protein afadin, one of the components of adherens junctions, is expressed ubiquitously including IDs. At present, the precise role of afadin in cardiac physiology or disease is unknown. To explore this, we generated conditional knockout (cKO) mice with cardiomyocyte-targeted deletion of afadin. Afadin cKO mice were born according to the expected Mendelian ratio and have no detectable changes in cardiac phenotype. On the other hand, chronic pressure overload induced by transverse aortic constriction (TAC) caused systolic dysfunction, enhanced fibrogenesis and apoptosis in afadin cKO mice. Afadin deletion increased macrophage infiltration and monocyte chemoattractant protein-1 expression, and suppressed transforming growth factor (TGF) ß receptor signaling early after TAC procedure. Afadin also associated with TGFß receptor I at IDs. Pharmacological antagonist of TGFß receptor I (SB431542) augmented mononuclear infiltration and fibrosis in the hearts of TAC-operated control mice. In conclusion, afadin is a critical molecule for cardiac protection against chronic pressure overload. The beneficial effects are likely to be a result from modulation of TGFß receptor signaling pathways by afadin.


Subject(s)
Aorta/physiology , Heart Failure, Systolic/genetics , Intercellular Junctions/metabolism , Macrophages/immunology , Microfilament Proteins/metabolism , Myocardium/pathology , Myocytes, Cardiac/physiology , Animals , Apoptosis/genetics , Cell Movement , Chemokine CCL2/metabolism , Constriction, Pathologic , Fibrosis , Mice , Mice, Knockout , Microfilament Proteins/genetics , Myocardial Contraction , Signal Transduction , Transforming Growth Factor beta/metabolism
9.
Eur J Heart Fail ; 19(3): 314-323, 2017 03.
Article in English | MEDLINE | ID: mdl-27990720

ABSTRACT

AIMS: Psychopathologies may occur in heart failure (HF) and can be associated with adverse outcomes. Amongst neuropeptide S receptor gene functional sequence variants, the T-allele [asparagine(107)isoleucine, NPSR1 rs324981] has been identified as a risk factor for increased anxiety/overinterpretation of bodily symptoms. We investigated all-cause death and re-hospitalization (composite primary endpoint, CPEP) and healthcare utilization in patients hospitalized for decompensated systolic HF with the TT vs. the AT/AA genotype. METHODS AND RESULTS: Participants in the Interdisciplinary Network Heart Failure programme were eligible if consenting to genetic testing (n = 924) and randomization to usual care (UC, n = 464) or nurse-co-ordinated disease management (DM, n = 460). Follow-up was 180 days (100% complete). Compared with AT/AA carriers (n = 726), TT genotype carriers (n = 198) had more CPEP events [47% vs. 39%, hazard ratio (HR) 1.27, 95% confidence interval (CI) 1.01-1.61, P = 0.044] and were more frequently re-hospitalized (43% vs. 35%, HR 1.31, 95% CI 1.02-1.67, P = 0.033); mortality rate was similar in both groups (HR 1.11, 95% CI 0.68-1.81, P = 0.664). In subjects undergoing DM, CPEP and re-hospitalization occurred more often in TT (51% and 47%) than in AT/AA carriers (36% and 33%; HR 2.14, 95% CI 1.44-3.19, and HR 2.29, 95% CI 1.52-3.44, genotype/treatment interaction both P = 0.007). Furthermore, TT genotype carriers undergoing DM visited cardiologists and other specialists more often than AT/AA carriers (P = 0.009 and P = 0.005). With UC, event rates did not differ between genotype subgroups. CONCLUSION: We identified a psychogenetic determinant of clinical outcomes and healthcare utilization after acute HF, which was modulated by the type of care. Future investigations need to clarify whether NPSR1 genotyping might further enhance the concept of 'personalized' medicine in HF. TRIAL REGISTRATION: ISRCTN23325295.


Subject(s)
Heart Failure, Systolic/genetics , Hospitalization/statistics & numerical data , Mortality , Receptors, G-Protein-Coupled/genetics , Aged , Aged, 80 and over , Cause of Death , Disease Management , Female , Genotype , Health Services/statistics & numerical data , Heart Failure, Systolic/mortality , Humans , Male , Middle Aged , Practice Patterns, Nurses' , Prognosis , Proportional Hazards Models , Randomized Controlled Trials as Topic , Risk Factors
10.
BMC Endocr Disord ; 15: 36, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26215257

ABSTRACT

BACKGROUND: The pathogenesis of diabetic cardiomyopathy (DCM) involves the enhanced activation of peroxisome proliferator activating receptor (PPAR) transcription factors, including the most prominent isoform in the heart, PPARα. In cancer cells and adipocytes, post-translational modification of PPARs have been identified, including ligand-dependent degradation of PPARs by specific ubiquitin ligases. However, the regulation of PPARs in cardiomyocytes and heart have not previously been identified. We recently identified that muscle ring finger-1 (MuRF1) and MuRF2 differentially inhibit PPAR activities by mono-ubiquitination, leading to the hypothesis that MuRF3 may regulate PPAR activity in vivo to regulate DCM. METHODS: MuRF3-/- mice were challenged with 26 weeks 60% high fat diet to induce insulin resistance and DCM. Conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARß, and PPARγ1 activities were assayed. RESULTS: MuRF3-/- mice exhibited a premature systolic heart failure by 6 weeks high fat diet (vs. 12 weeks in MuRF3+/+). MuRF3-/- mice weighed significantly less than sibling-matched wildtype mice after 26 weeks HFD. These differences may be largely due to resistance to fat accumulation, as MRI analysis revealed MuRF3-/- mice had significantly less fat mass, but not lean body mass. In vitro ubiquitination assays identified MuRF3 mono-ubiquitinated PPARα and PPARγ1, but not PPARß. CONCLUSIONS: These findings suggest that MuRF3 helps stabilize cardiac PPARα and PPARγ1 in vivo to support resistance to the development of DCM. MuRF3 also plays an unexpected role in regulating fat storage despite being found only in striated muscle.


Subject(s)
Diabetic Cardiomyopathies/genetics , Diet, High-Fat/adverse effects , Heart Failure, Systolic/genetics , Muscle Proteins/genetics , Myocytes, Cardiac/metabolism , Adipose Tissue , Animals , Body Composition , Body Weight , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Heart Failure, Systolic/etiology , Heart Failure, Systolic/metabolism , In Vitro Techniques , Mice , Mice, Knockout , Muscle Proteins/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Ubiquitination
11.
PLoS One ; 10(5): e0128105, 2015.
Article in English | MEDLINE | ID: mdl-26023924

ABSTRACT

GATA4 and GATA6 are central cardiac transcriptional regulators. The postnatal, stage-specific function of the cardiac transcription factors GATA4 and GATA6 have not been evaluated. In part, this is because current Cre-loxP approaches to cardiac gene inactivation require time consuming and costly breeding of Cre-expressing and "floxed" mouse lines, often with limited control of the extent or timing of gene inactivation. We investigated the stage-specific functions of GATA4 and GATA6 in the postnatal heart by using adeno-associated virus serotype 9 to control the timing and extent of gene inactivation by Cre. Systemic delivery of recombinant, adeno-associated virus 9 (AAV9) expressing Cre from the cardiac specific Tnnt2 promoter was well tolerated and selectively and efficiently recombined floxed target genes in cardiomyocytes. AAV9:Tnnt2-Cre efficiently inactivated Gata4 and Gata6. Neonatal Gata4/6 inactivation caused severe, rapidly lethal systolic heart failure. In contrast, Gata4/6 inactivation in adult heart caused only mild systolic dysfunction but severe diastolic dysfunction. Reducing the dose of AAV9:Tnnt2-Cre generated mosaics in which scattered cardiomyocytes lacked Gata4/6. This mosaic knockout revealed that Gata4/6 are required cell autonomously for physiological cardiomyocyte growth. Our results define novel roles of GATA4 and GATA6 in the neonatal and adult heart. Furthermore, our data demonstrate that evaluation of gene function hinges on controlling the timing and extent of gene inactivation. AAV9:Tnnt2-Cre is a powerful tool for controlling these parameters.


Subject(s)
Dependovirus/genetics , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/metabolism , Heart/growth & development , Integrases/genetics , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Genetic Vectors , Heart/physiology , Heart Failure, Systolic/genetics , Integrases/administration & dosage , Mice, Transgenic
12.
Nitric Oxide ; 47: 91-6, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25917853

ABSTRACT

BACKGROUND: The endothelial nitric oxide synthase (eNOS) gene single nucleotide polymorphism G894T is associated with thrombotic vascular diseases. However, its functional significance is controversial and data are scarce concerning its influence in heart failure (HF). METHODS: We studied 215 patients with chronic systolic HF. DNA was analyzed for eNOS gene G894T polymorphism using PCR and DNA sequencing. Evaluation of clinical characteristics and analysis of factors associated with 2-year mortality were performed for the homozygous G-allele G894T variant (GG), relative to the TT and GT variants. RESULTS: The genotype distributions of eNOS G894T alleles were: GG 135 patients (63%) and TT/GT 80 (37%). Two-year mortality was significantly higher in the GG variant (48%) than the combined TT/GT group (32%). The usage of nitrates was associated with increased 2-year mortality (HR 2.0, 95% CI 1.28-3.17; p = 0.003), which was most significant in the GG group treated with nitrates (73.5%) in comparison to the TT/GT group not treated with nitrates (34%); HR 2.75, 95% CI 1.57-4.79, P < 0.001. CONCLUSIONS: Homozygosity for the G allele of the eNOS G894T polymorphism was associated with worse survival in systolic HF patients, especially in those treated with nitrates. ENOS polymorphism may result in different mechanistic interactions in HF than in thrombotic vascular diseases, suggesting that overexpression of NO may be associated with deleterious effects in systolic HF.


Subject(s)
Heart Failure, Systolic/diagnosis , Heart Failure, Systolic/genetics , Nitric Oxide Synthase Type III/genetics , Polymorphism, Genetic/genetics , Aged , Female , Heart Failure, Systolic/enzymology , Humans , Male , Middle Aged , Nitric Oxide Synthase Type III/metabolism , Prognosis
14.
J Mol Cell Cardiol ; 82: 174-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25784084

ABSTRACT

Differential DNA methylation exists in the epigenome of end-stage failing human hearts but whether it contributes to disease progression is presently unknown. Here, we report that cardiac specific deletion of Dnmt3b, the predominant DNA methyltransferase in adult mouse hearts, leads to an accelerated progression to severe systolic insufficiency and myocardial thinning without a preceding hypertrophic response. This was accompanied by widespread myocardial interstitial fibrosis and myo-sarcomeric disarray. By targeted candidate gene quantitative RT-PCR, we discovered an over-activity of cryptic splice sites in the sarcomeric gene Myh7, resulting in a transcript with 8 exons missing. Moreover, a region of differential methylation overlies the splice site locus in the hearts of the cardiac-specific conditional knockout (CKO) mice. Although abundant and complex forms of alternative splice variants have been reported in diseased hearts and the contribution of each remains to be understood in further detail, our results demonstrate for the first time that a link may exist between alternative splicing and the cardiac epigenome. In particular, this gives the novel evidence whereby the loss of an epigenome modifier promotes the development and progression of heart disease.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Alternative Splicing , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Disease Models, Animal , Fibrosis , Gene Deletion , Heart Failure/pathology , Heart Failure/physiopathology , Heart Failure, Systolic/genetics , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/pathology , Heart Failure, Systolic/physiopathology , Humans , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/genetics , Organ Specificity/genetics , Protein Aggregates , Proteolysis , Sarcomeres/genetics , Sarcomeres/metabolism , Sarcomeres/pathology , Ubiquitin/metabolism , DNA Methyltransferase 3B
15.
J Transl Med ; 13: 44, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25638254

ABSTRACT

BACKGROUND: G-protein receptor kinase 5 (GRK5) Gln41 > Leu and ß1-adrenergic receptor (ADRB1) Arg389 > Gly polymorphisms presented the different distribution of genotype frequencies between Caucasian American and African American, and produced the difference in ß-blocker treatment effect among them with systolic heart failure (SHF). OBJECTIVE: This study sought to identify the distributed characteristics of these variant genotypes in Chinese population, and influence of GRK5 and ADRB1 polymorphisms on SHF morbidity and ß-blocker treatment effect in patients with SHF. METHODS: This study was based on cross-sectional survey data. 1794 and 1718 subjects' ADRB1 and GRK5 gene sequencing (sanger method) data were achieved respectively. Blood samples collection, clinical laboratory detection, electrocardiogram and echocardiography examinations were performed. Medication usage was confirmed at in-hospital visits or the questionnaire by personal interview. RESULTS: GRK5 Leu41Leu genotype was not found in our Chinese population. In non-SHF population, allele frequencies of GRK5 Gln41 and Leu41 were 2782 (0.992) and 22 (0.008) (Hardy-Weinberg equilibrium test χ(2) = 0.088, P = 0.767), and allele frequencies of ADRB1 Arg389 and Gly389 were 2127 (0.715) and 849 (0.285) (χ(2) = 0.272, P = 0.602). In SHF patients, allele frequencies of Gln41 and Leu41 were 446 (0.991) and 4 (0.009) (χ(2) = 0.018, P = 0.893), and allele frequencies of Arg389 and Gly389 were 331 (0.726) and 125 (0.274) (χ(2) = 1.892, P = 0.169). Further in logistic regression model, these ADRB1 and GRK5 variants were not significantly independently associated with the risk of SHF morbidity. Those carrying genotype ADRB1 Gly389Gly did not reduce significantly the risk of SHF morbidity after ß-blocker therapy. CONCLUSIONS: GRK5 Leu41Leu genotype was not found in our Chinese population, neither ADRB1 nor GRK5 variants presented independently associated with the risk of SHF morbidity, most ADRB1 and GRK5 polymorphisms did decrease significantly the risk of SHF morbidity after ß-blocker therapy except for those carrying genotype ADRB1 Gly389Gly.


Subject(s)
G-Protein-Coupled Receptor Kinase 5/genetics , Heart Failure, Systolic/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Adrenergic, beta-1/genetics , Adrenergic beta-Antagonists/therapeutic use , Base Sequence , Heart Failure, Systolic/drug therapy , Humans , Models, Genetic , Molecular Sequence Data , Multivariate Analysis , Sequence Analysis, DNA
16.
Kardiologiia ; 55(9): 25-30, 2015.
Article in Russian | MEDLINE | ID: mdl-26922818

ABSTRACT

AIM: to elucidate association between polymorphic markers of interleukin-6 (Il-6) and tumor necrosis factor (TNF) genes and unfavorable outcomes in patients with chronic heart failure (CHF). MATERIAL AND METHODS: We determined levels of TNF and Il-6 and genotypes of polymorphic markers G(-238)A of TNF gene (rs361525) and G(--174)C of IL-6 gene (rs1800795) in 151 patients (mean age 64.5 years) hospitalized because of decompensation of systolic CHF (left ventricular ejection fraction ≤ 40%) after stabilization of their state. Unfavorable outcomes were registered during follow-up for 2 years. RESULTS: Mean levels of NT-proBNP, Il-6, and TNF were 2481.1 ± 199.86 fmol/ml, 21.8 7.46 rg/ml, and 10.07 ± 0.65 rg/ml, respectively. 138 (94.4%), 13 (8.6%) and 0 patients were carriers of genotypes GG, AG, and AA of polymorphic marker G(-238)A of TNF gene, respectively; 54 (35.8%), 69 (45.7%), and 28 (18.5%) patients carried genotypes GG, GC, and CC of polymorphic marker G(-174)C gene IL-6, respectively. There was no association between Il-6, TNF levels and carriage of either of genotypes as well as unfavorable clinical course of CHF. Mean survival time before repetitive episode of CHF decompensation (including lethal one) was significantly shorter among carriers of A allele compared with carriers of G allele of polymorphic marker G(-238)A of TNF gene (243 ± 97.7 and 947 ± 78 days, respectively, p = 0.018). Mean time before all cause death was also shorter in carriers of A compared with carriers of G allele (289 ± 122.9 and 1039 ± 73.3 days, respectively, p = 0.03). The studied polymorphism of IL-6 gene had no prognostic value. CONCLUSION: We obtained data on association between carriage of A allele of polymorphic marker G(-238)A of TNF gene and unfavorable prognosis in patients with CHF and inpraired left ventricular systolic function.


Subject(s)
DNA/genetics , Heart Failure, Systolic/genetics , Polymorphism, Genetic , Tumor Necrosis Factor-alpha/genetics , Alleles , Female , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/mortality , Humans , Male , Middle Aged , Prognosis , Risk Factors , Russia/epidemiology , Survival Rate/trends , Tumor Necrosis Factor-alpha/metabolism
17.
J Hypertens ; 33(1): 3-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25380161

ABSTRACT

Heart failure accounts for a significant portion of heart diseases. Molecular mechanisms gradually emerge that participate in pathways leading to left ventricular dysfunction in common systolic heart failure (SHF) and diastolic heart failure (DHF). A human genome-wide association study (GWAS) identified two markers for SHF and no GWAS on DHF has been documented. However, genetic analyses in rat models of SHF and DHF have begun to unravel the genetic components known as quantitative trait loci (QTLs) initiating systolic and diastolic function. A QTL for systolic function was detected and the gene responsible for it is identified to be that encoding the soluble epoxide hydrolase. Diastolic function is determined by multiple QTLs and the Ccl2/monocyte chemotactic protein gene is the strongest candidate. An amelioration on diastolic dysfunction is merely transient from changing such a single QTL accompanied by a blood pressure reduction. A long-term protection can be achieved only via combining alleles of several QTLs. Thus, distinct genes in synergy are involved in physiological mechanisms durably ameliorating or reversing diastolic dysfunction. These data lay the foundation for identifying causal genes responsible for individual diastolic function QTLs and the essential combination of them to attain a permanent protection against diastolic dysfunction, and consequently will facilitate the elucidation of pathophysiological mechanisms underlying hypertensive diastolic dysfunction. Novel pathways triggering systolic and diastolic dysfunction have emerged that will likely provide new diagnostic tools, innovative therapeutic targets and strategies in reducing, curing and even reversing SHF and DHF.


Subject(s)
Heart Failure, Diastolic/genetics , Heart Failure, Systolic/genetics , Animals , Chemokine CCL2/genetics , Disease Models, Animal , Epoxide Hydrolases/genetics , Genetic Markers/genetics , Genome-Wide Association Study , Heart Failure, Diastolic/complications , Heart Failure, Systolic/complications , Humans , Quantitative Trait Loci , Rats , Ventricular Dysfunction, Left/etiology
18.
BMC Med Genet ; 15: 140, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25528061

ABSTRACT

BACKGROUND: Recent genome-wide association studies have identified multiple loci that are associated with an increased risk of developing coronary artery disease (CAD). The impact of these loci on the disease severity and prognosis of ischemic heart failure due to CAD is currently unknown. METHODS: We undertook association analysis of 7 single nucleotide polymorphism (rs599839, rs17465637, rs2972147, rs6922269, rs1333049, rs501120, and rs17228212) at 7 well established CAD risk loci (1p13.3, 1q41, 2q36.3, 6q25.1, 9p21.3, 10q11.21, and 15q22.33, respectively) in 3,320 subjects diagnosed with systolic heart failure of ischemic aetiology and participating in the COntrolled ROsuvastatin multiNAtional Trial in Heart Failure (CORONA) trial. The primary outcome was the composite of time to first event of cardiovascular death, non-fatal myocardial infarction and non-fatal stroke, secondary outcomes included mortality and hospitalization due to worsening heart failure. RESULTS: None of the 7 loci were significantly associated with the primary composite endpoint of the CORONA trial (death from cardiovascular cases, nonfatal myocardial infarction, and nonfatal stroke). However, the 1p13.3 locus (rs599839) showed evidence for association with all-cause mortality (after adjustment for covariates; HR 0.74, 95%CI [0.61 to 0.90]; P = 0.0025) and we confirmed the 1p13.3 locus (rs599839) to be associated with lipid parameters (total cholesterol (P = 1.1x10(-4)), low-density lipoprotein levels (P = 3.5 × 10(-7)) and apolipoprotein B (P = 2.2 × 10(-10))). CONCLUSION: Genetic variants strongly associated with CAD risk are not associated with the severity and outcome of ischemic heart failure. The observed association of the 1p13.3 locus with all-cause mortality requires confirmation in further studies.


Subject(s)
Coronary Artery Disease/genetics , Heart Failure, Systolic/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Clinical Trials as Topic , Coronary Artery Disease/complications , Coronary Artery Disease/mortality , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Prognosis
19.
J Cardiovasc Pharmacol Ther ; 19(5): 439-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24500905

ABSTRACT

INTRODUCTION: Vitamin D deficiency has been associated with the development of myocardial hypertrophy and inflammation. These findings suggest that vitamin D status and vitamin D receptor (VDR) genomics may play a role in myocardial fibrosis. The aim of this pilot study was to determine the association between vitamin D levels, VDR polymorphisms, and biomarkers of left ventricular remodeling and hemodynamics. METHODS: In a cross-sectional pilot study, patients with ejection fraction (EF) <40% (and New York Heart Association ≥ II) undergoing right heart catheterization were included in the study. Blood was collected for determination of 25-hydroxyvitamin D level (antibody competitive immunoassay), VDR genotypes (BsmI, ApaI, TaqI, and FokI), and biomarkers (N-terminal propeptide of collagen type III [PIIINP], matrix metalloproteinase 2, and galectin 3). The vitamin D genotypes were determined through the use of pyrosequencing. RESULTS: A total of 30 patients with a mean EF of 17% ± 8% were enrolled. There was a significant association between the BsmI C allele, ApaI G allele, and TaqI A allele, which formed a haplotype block (CGA) for analysis. There were no differences in baseline parameters between patients with the VDR haplotype block (n = 20) and those without (n = 10). Individual genotypes were not associated with any biomarker or hemodynamics. Patients with the CGA haplotype demonstrated significantly higher log PIIINP values (1.74 ± 0.32 mcg/mL vs 1.36 ± 0.31 mcg/mL, P = .0041). When evaluating vitamin D levels below and above the median level (19 ng/mL), there was no significant difference between these 2 groups in regard to biomarker levels for left ventricular remodeling. CONCLUSION: This study has shown that a biomarker for collagen type III synthesis, PIIINP, was associated with the CGA haplotype of BsmI, ApaI, and TaqI single nucleotide polymorphisms on the VDR. These findings suggest that VDR genetics may play a role in myocardial fibrosis in patients with systolic heart failure.


Subject(s)
Biomarkers/blood , Heart Failure, Systolic/genetics , Receptors, Calcitriol/genetics , Adult , Aged , Cross-Sectional Studies , Female , Fibrosis , Genotype , Haplotypes , Heart Failure, Systolic/blood , Heart Failure, Systolic/physiopathology , Hemodynamics , Humans , Male , Middle Aged , Pilot Projects , Receptors, Calcitriol/blood , Vitamin D/blood , Vitamin D/genetics , Vitamin D Deficiency
20.
Am J Cardiol ; 113(6): 976-81, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24462065

ABSTRACT

Atrial fibrillation (AF) is associated with poor prognosis in patients with heart failure (HF). Although platelets play an important role in rendering a prothrombotic state in AF, the exact mechanism by which the effect is mediated is still debated. MicroRNAs (miRNAs), which have been shown to be involved in a variety of cardiovascular conditions, are abundant in platelets and in a cell-free form in the circulation. In the present study, we performed a genome-wide screen for miRNA expression in platelets of patients with systolic HF and in controls without cardiac disease, in pursuit of specific miRNAs that are associated with the presence of AF. MiRNA expression was measured in platelets from 50 patients with systolic HF and 50 controls, of which, samples from 41 patients with HF and 35 controls were used in the final analysis because of a quality control process. MiR-150 expression was 3.2-fold lower (p = 0.0003) in platelets of patients with HF with AF relative to those without AF. A similar effect was seen in serum samples from the same patients, in which miR-150 levels were 1.5-fold lower (p = 0.004) in patients with HF with AF. Furthermore, the serum levels of miR-150 were correlated to platelet levels in patients with AF (r = 0.65, p = 0.0087). In conclusion, miR-150 expression levels in platelets of patients with systolic HF with AF are significantly reduced and correlated to the cell-free circulating levels of this miRNA.


Subject(s)
Atrial Fibrillation/genetics , Blood Platelets/metabolism , Gene Expression Regulation , Heart Failure, Systolic/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Aged , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Electrocardiography, Ambulatory , Female , Follow-Up Studies , Heart Failure, Systolic/blood , Heart Failure, Systolic/etiology , Humans , Male , MicroRNAs/biosynthesis , Middle Aged , Polymerase Chain Reaction , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...