Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Publication year range
1.
Circ Res ; 135(3): 434-449, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38864216

ABSTRACT

BACKGROUND: Cardiac hypertrophy is an adaptive response to pressure overload aimed at maintaining cardiac function. However, prolonged hypertrophy significantly increases the risk of maladaptive cardiac remodeling and heart failure. Recent studies have implicated long noncoding RNAs in cardiac hypertrophy and cardiomyopathy, but their significance and mechanism(s) of action are not well understood. METHODS: We measured lincRNA-p21 RNA and H3K27ac levels in the hearts of dilated cardiomyopathy patients. We assessed the functional role of lincRNA-p21 in basal and surgical pressure-overload conditions using loss-of-function mice. Genome-wide transcriptome analysis revealed dysregulated genes and pathways. We labeled proteins in proximity to full-length lincRNA-p21 using a novel BioID2-based system. We immunoprecipitated lincRNA-p21-interacting proteins and performed cell fractionation, ChIP-seq (chromatin immunoprecipitation followed by sequencing), and co-immunoprecipitation to investigate molecular interactions and underlying mechanisms. We used GapmeR antisense oligonucleotides to evaluate the therapeutic potential of lincRNA-p21 inhibition in cardiac hypertrophy and associated heart failure. RESULTS: lincRNA-p21 was induced in mice and humans with cardiomyopathy. Global and cardiac-specific lincRNA-p21 knockout significantly suppressed pressure overload-induced ventricular wall thickening, stress marker elevation, and deterioration of cardiac function. Genome-wide transcriptome analysis and transcriptional network analysis revealed that lincRNA-p21 acts in trans to stimulate the NFAT/MEF2 (nuclear factor of activated T cells/myocyte enhancer factor-2) pathway. Mechanistically, lincRNA-p21 is bound to the scaffold protein KAP1 (KRAB-associated protein-1). lincRNA-p21 cardiac-specific knockout suppressed stress-induced nuclear accumulation of KAP1, and KAP1 knockdown attenuated cardiac hypertrophy and NFAT activation. KAP1 positively regulates pathological hypertrophy by physically interacting with NFATC4 to promote the overactive status of NFAT/MEF2 signaling. GapmeR antisense oligonucleotide depletion of lincRNA-p21 similarly inhibited cardiac hypertrophy and adverse remodeling, highlighting the therapeutic potential of inhibiting lincRNA-p21. CONCLUSIONS: These findings advance our understanding of the functional significance of stress-induced long noncoding RNA in cardiac hypertrophy and demonstrate the potential of lincRNA-p21 as a novel therapeutic target for cardiac hypertrophy and subsequent heart failure.


Subject(s)
Cardiomegaly , Mice, Knockout , RNA, Long Noncoding , Animals , Humans , Male , Mice , Cardiomegaly/metabolism , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cardiomegaly/pathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ventricular Remodeling
2.
Circ Res ; 128(10): 1514-1532, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33983834

ABSTRACT

Our insight into the diverse and complex nature of dilated cardiomyopathy (DCM) genetic architecture continues to evolve rapidly. The foundations of DCM genetics rest on marked locus and allelic heterogeneity. While DCM exhibits a Mendelian, monogenic architecture in some families, preliminary data from our studies and others suggests that at least 20% to 30% of DCM may have an oligogenic basis, meaning that multiple rare variants from different, unlinked loci, determine the DCM phenotype. It is also likely that low-frequency and common genetic variation contribute to DCM complexity, but neither has been examined within a rare variant context. Other types of genetic variation are also likely relevant for DCM, along with gene-by-environment interaction, now established for alcohol- and chemotherapy-related DCM. Collectively, this suggests that the genetic architecture of DCM is broader in scope and more complex than previously understood. All of this elevates the impact of DCM genetics research, as greater insight into the causes of DCM can lead to interventions to mitigate or even prevent it and thus avoid the morbid and mortal scourge of human heart failure.


Subject(s)
Alleles , Cardiomyopathy, Dilated/genetics , Genetic Loci , Genetic Variation , Cardiomyopathy, Dilated/prevention & control , Cohort Studies , Connectin/chemistry , Cross-Sectional Studies , Gene-Environment Interaction , Humans , Models, Statistical , Phenotype , Sarcomeres/chemistry
3.
Circ Res ; 127(5): 610-627, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32466726

ABSTRACT

RATIONALE: Doxorubicin-induced cardiomyopathy (DiCM) is a primary cause of heart failure and mortality in cancer patients, in which macrophage-orchestrated inflammation serves as an essential pathological mechanism. However, the specific roles of tissue-resident and monocyte-derived macrophages in DiCM remain poorly understood. OBJECTIVE: Uncovering the origins, phenotypes, and functions of proliferative cardiac resident macrophages and mechanistic insights into the self-maintenance of cardiac macrophage during DiCM progression. METHODS AND RESULTS: Mice were administrated with doxorubicin to induce cardiomyopathy. Dynamic changes of resident and monocyte-derived macrophages were examined by lineage tracing, parabiosis, and bone marrow transplantation. We found that the monocyte-derived macrophages primarily exhibited a proinflammatory phenotype that dominated the whole DiCM pathological process and impaired cardiac function. In contrast, cardiac resident macrophages were vulnerable to doxorubicin insult. The survived resident macrophages exhibited enhanced proliferation and conferred a reparative role. Global or myeloid specifically ablation of SR-A1 (class A1 scavenger receptor) inhibited proliferation of cardiac resident reparative macrophages and, therefore, exacerbated cardiomyopathy in DiCM mice. Importantly, the detrimental effect of macrophage SR-A1 deficiency was confirmed by transplantation of bone marrow. At the mechanistic level, we show that c-Myc (Avian myelocytomatosis virus oncogene cellular homolog), a key transcriptional factor for the SR-A1-P38-SIRT1 (Sirtuin 1) pathway, mediated the effect of SR-A1 in reparative macrophage proliferation in DiCM. CONCLUSIONS: The SR-A1-c-Myc axis may represent a promising target to treat DiCM through augmentation of cardiac resident reparative macrophage proliferation.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Cell Proliferation , Cell Self Renewal , Macrophages/enzymology , Myocardium/enzymology , Proto-Oncogene Proteins c-myc/metabolism , Scavenger Receptors, Class A/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cardiomyopathy, Dilated/chemically induced , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Disease Models, Animal , Doxorubicin , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Signal Transduction , Ventricular Remodeling
4.
J Recept Signal Transduct Res ; 41(5): 494-503, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33092439

ABSTRACT

Down-regulated lncRNA AC061961.2 in dilated cardiomyopathy (DCM) patients was previous reported. Whether AC061961.2 has regulatory effect on DCM still need exploration. Here, we tried to investigate the effect of AC061961.2 on DCM. After DCM model rat was established through injecting Adriamycin, left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD), left ventricular ejection fraction (LVEF), and left ventricular fractional shortening (LVFS) were measured by echocardiography. Histopathological changes and apoptosis were detected by hematoxylin-eosin, Masson staining, and TUNEL. After cardiomyocytes were isolated and identified by immunofluorescence, DCM cell model was established by injecting adriamycin. After transfected with overexpressed-AC061961.2 plasmids, cell apoptosis was detected by flow cytometry. The expressions of AC061961.2, ß-catenin, Axin2, c-Myc, CRP78, CHOP, Caspase-3, Bcl-2, and Bax in cardiomyocytes and heart tissues were detected by RT-qPCR or western blot. LVEDD and LVESD were increased while LVEF and LVFS were decreased in DCM rats. The histopathological of heart tissues showed a typical sign of DCM. Apoptosis were increased in heart tissues of DCM rats. In DCM rats, the expressions of AC061961.2, ß-catenin, Axin2, c-Myc, and Bcl-2 were decreased, the expressions of CRP78, CHOP, Caspase-3, and Bax were increased. After the overexpression of AC061961.2, levels of ß-catenin, Axin2, c-Myc, and Bcl-2 were increased, while levels of CRP78, CHOP, Caspase-3, and Bax were decreased, compared with that in DCM cardiomyocytes. LncRNA AC061961.2 overexpression inhibited endoplasmic reticulum stress induced apoptosis in DCM rats and cardiomyocytes via activating Wnt/ß-catenin pathway.


Subject(s)
Apoptosis , Cardiomyopathy, Dilated/prevention & control , Endoplasmic Reticulum Stress , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Wnt1 Protein/genetics , beta Catenin/genetics
5.
FASEB J ; 34(2): 2987-3005, 2020 02.
Article in English | MEDLINE | ID: mdl-31908029

ABSTRACT

The expression of α-cardiac actin, a major constituent of the cytoskeleton of cardiomyocytes, is dramatically decreased in a mouse model of dilated cardiomyopathy triggered by inducible cardiac-specific serum response factor (Srf) gene disruption that could mimic some forms of human dilated cardiomyopathy. To investigate the consequences of the maintenance of α-cardiac actin expression in this model, we developed a new transgenic mouse based on Cre/LoxP strategy, allowing together the induction of SRF loss and a compensatory expression of α-cardiac actin. Here, we report that maintenance of α-cardiac actin within cardiomyocytes temporally preserved cytoarchitecture from adverse cardiac remodeling through a positive impact on both structural and transcriptional levels. These protective effects were accompanied in vivo by the decrease of ROS generation and protein carbonylation and the downregulation of NADPH oxidases NOX2 and NOX4. We also show that ectopic expression of α-cardiac actin protects HEK293 cells against oxidative stress induced by H2 O2 . Oxidative stress plays an important role in the development of cardiac remodeling and contributes also to the pathogenesis of heart failure. Taken together, these findings indicate that α-cardiac actin could be involved in the regulation of oxidative stress that is a leading cause of adverse remodeling during dilated cardiomyopathy development.


Subject(s)
Actins/metabolism , Cardiomyopathy, Dilated/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress , Actins/genetics , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Disease Models, Animal , Female , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism
6.
Circulation ; 139(24): 2778-2792, 2019 06 11.
Article in English | MEDLINE | ID: mdl-30922078

ABSTRACT

BACKGROUND: Enhancers are genomic regulatory elements conferring spatiotemporal and signal-dependent control of gene expression. Recent evidence suggests that enhancers can generate noncoding enhancer RNAs, but their (patho)biological functions remain largely elusive. METHODS: We performed chromatin immunoprecipitation-coupled sequencing of histone marks combined with RNA sequencing of left ventricular biopsies from experimental and genetic mouse models of human cardiac hypertrophy to identify transcripts revealing enhancer localization, conservation with the human genome, and hypoxia-inducible factor 1α dependence. The most promising candidate, hypoxia-inducible enhancer RNA ( HERNA)1, was further examined by investigating its capacity to modulate neighboring coding gene expression by binding to their gene promoters by using chromatin isolation by RNA purification and λN-BoxB tethering-based reporter assays. The role of HERNA1 and its neighboring genes for pathological stress-induced growth and contractile dysfunction, and the therapeutic potential of HERNA1 inhibition was studied in gapmer-mediated loss-of-function studies in vitro using human induced pluripotent stem cell-derived cardiomyocytes and various in vivo models of human pathological cardiac hypertrophy. RESULTS: HERNA1 is robustly induced on pathological stress. Production of HERNA1 is initiated by direct hypoxia-inducible factor 1α binding to a hypoxia-response element in the histoneH3-lysine27acetylation marks-enriched promoter of the enhancer and confers hypoxia responsiveness to nearby genes including synaptotagmin XVII, a member of the family of membrane-trafficking and Ca2+-sensing proteins and SMG1, encoding a phosphatidylinositol 3-kinase-related kinase. Consequently, a substrate of SMG1, ATP-dependent RNA helicase upframeshift 1, is hyperphoshorylated in a HERNA1- and SMG1-dependent manner. In vitro and in vivo inactivation of SMG1 and SYT17 revealed overlapping and distinct roles in modulating cardiac hypertrophy. Finally, in vivo administration of antisense oligonucleotides targeting HERNA1 protected mice from stress-induced pathological hypertrophy. The inhibition of HERNA1 postdisease development reversed left ventricular growth and dysfunction, resulting in increased overall survival. CONCLUSIONS: HERNA1 is a novel heart-specific noncoding RNA with key regulatory functions in modulating the growth, metabolic, and contractile gene program in disease, and reveals a molecular target amenable to therapeutic exploitation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/prevention & control , Cardiomyopathy, Hypertrophic/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocytes, Cardiac/metabolism , Oligonucleotides, Antisense/administration & dosage , RNA, Untranslated/metabolism , Animals , Binding Sites , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Case-Control Studies , Disease Models, Animal , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Promoter Regions, Genetic , RNA, Untranslated/genetics , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
7.
Microb Pathog ; 144: 104169, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32205210

ABSTRACT

Viral myocarditis (VMC) is a type of inflammation affecting myocardial cells caused by viral infection and has been an important cause of dilated cardiomyopathy (DCM) worldwide. Type B3 coxsackievirus (CVB3), a non-enveloped positive-strand RNA virus of the Enterovirus genus, is one of most common agent of viral myocarditis. Till now, effective treatments for VMC are lacking due to lack of drugs or vaccine. Lithium chloride (LiCl) is applied in the clinical management of manic depressive disorders. Accumulating evidence have demonstrated that LiCl, also as an effective antiviral drug, exhibited antiviral effects for specific viruses. However, there are few reports of evaluating LiCl's antiviral effect in mice model. Here, we investigated the inhibitory influence of LiCl on the CVB3 replication in vitro and in vivo and the development of CVB3-induced VMC. We found that LiCl significantly suppressed CVB3 replication in HeLa via inhibiting virus-induced cell apoptosis. Moreover, LiCl treatment in vivo obviously inhibited virus replication within the myocardium and alleviated CVB3-induced acute myocarditis. Collectively, our data demonstrated that LiCl inhibited CVB3 replication and negatively regulated virus-triggered inflammatory responses. Our finding further expands the antiviral targets of LiCl and provides an alternative agent for viral myocarditis.


Subject(s)
Antiviral Agents/pharmacology , Cardiomyopathy, Dilated/drug therapy , Coxsackievirus Infections/drug therapy , Enterovirus B, Human/drug effects , Lithium Chloride/pharmacology , Myocarditis/drug therapy , Animals , Apoptosis/drug effects , Cardiomyopathy, Dilated/prevention & control , Cardiomyopathy, Dilated/virology , Cell Line , Coxsackievirus Infections/prevention & control , Coxsackievirus Infections/virology , Disease Models, Animal , Drug Repositioning , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Myocarditis/prevention & control , Myocarditis/virology , Myocardium/pathology , Virus Replication/drug effects
8.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751757

ABSTRACT

Nearly one in three people in the U.S. will develop heart failure (HF), characterized by fluid retention (edema) in the lungs and elsewhere. This leads to difficult breathing, deterioration of physical capacity, restriction of normal activities and death. There is little data about the safety and effects of sexual interactions in patients with HF. We tested whether a lack of sexual interactions affected pathophysiological outcomes in a pre-clinical mouse model of dilated cardiomyopathy that recapitulates the progressive stages of human HF. Male mice were randomly given access to, or deprived from, sexual interactions with female mice, which were confirmed by videography and generation of offspring. Cohousing with access to sexual interactions markedly prolonged survival, while cohousing without access to sexual activity did not. Sexual interactions improved systolic function, reduced HF-associated edema, altered transcription of heart contractile protein genes and decreased plasma testosterone levels. To determine whether testosterone levels contributed to survival, testosterone levels were experimentally reduced. Reduction of testosterone levels significantly prolonged survival. Taken together, in mice with dilated cardiomyopathy, sexual activity altered cardiac contractile gene transcription, improved systolic function, reduced edema and prolonged survival which may be in part due to lower testosterone levels.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Coitus/physiology , Heart Failure/prevention & control , Sexual Behavior/physiology , Animals , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Female , Heart Failure/physiopathology , Humans , Male , Mice , Myocardial Contraction , Survival/physiology
9.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344926

ABSTRACT

AIMS: Coxsackievirus B3 (CVB3) is known to be an important cause of myocarditis and dilated cardiomyopathy. Enterovirus-2C (E2C) is a viral RNA helicase. It inhibits host protein synthesis. Based on these facts, we hypothesize that the inhibition of 2C may suppress virus replication and prevent enterovirus-mediated cardiomyopathy. METHODS AND RESULTS: We generated a chemically modified enterovirus-2C inhibitor (E2CI). From the in vitro assay, E2CI was showed strong antiviral effects. For in vivo testing, mice were treated with E2CI intraperitoneally injected daily for three consecutive days at a dose of 8mg/kg per day, after CVB3 post-infection (p.i) (CVB3 + E2CI, n = 33). For the infected controls (CVB3 only, n = 35), mice were injected with PBS (phosphate buffered saline) in a DBA/2 strain to establish chronic myocarditis. The four-week survival rate of E2CI-treated mice was significantly higher than that of controls (92% vs. 71%; p < 0.05). Virus titers and myocardial damage were significantly reduced in the E2CI treated group. In addition, echocardiography indicated that E2CI administration dramatically maintained mouse heart function compared to control at day 28 p.i chronic stage (LVIDD, 3.1 ± 0.08 vs. 3.9 ± 0.09, p < 0.01; LVDS, 2.0 ± 0.07 vs. 2.5 ± 0.07, p < 0.001; FS, 34.8 ± 1.6% vs. 28.5 ± 1.5%; EF, 67. 9 ± 2.9% vs. 54.7 ± 4.7%, p < 0.05; CVB3 + E2CI, n = 6 vs. CVB3, n = 4). Moreover, E2CI is effectively worked in human iPS (induced pluripotent stem cell) derived cardiomyocytes. CONCLUSION: Enterovirus-2C inhibitor (E2CI) was significantly reduced viral replication, chronic myocardium damage, and CVB3-induced mortality in DBA/2 mice. These results suggested that E2CI is a novel therapeutic agent for the treatment of enterovirus-mediated diseases.


Subject(s)
Antiviral Agents/pharmacology , Coxsackievirus Infections/drug therapy , Enterovirus B, Human/enzymology , Induced Pluripotent Stem Cells/drug effects , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , RNA Helicases/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/therapeutic use , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/prevention & control , Chronic Disease , Coxsackievirus Infections/complications , Enterovirus B, Human/drug effects , Enterovirus B, Human/physiology , HeLa Cells , Humans , Induced Pluripotent Stem Cells/virology , Luciferases, Renilla/analysis , Male , Mice , Mice, Inbred DBA , Myocarditis/etiology , Myocarditis/virology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/virology , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Oxazoles/pharmacology , Oxazoles/therapeutic use , Recombinant Fusion Proteins/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/prevention & control , Virus Replication/drug effects
10.
Basic Res Cardiol ; 113(3): 19, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29564567

ABSTRACT

Previous studies have suggested the involvement of CD4 + T lymphocytes in cardiac remodelling. T-bet can direct Th1 lineage commitment. This study aimed to investigate the functional significance of T-bet in cardiac remodelling induced by pressure overload using T-bet global knockout rats. Increased T-bet levels were observed in rodent and human hypertrophied hearts. T-bet deficiency resulted in a less severe hypertrophic phenotype in rats. CD4 + T-lymphocyte reconstitution in T-bet-/- rats resulted in aggravated cardiac remodelling. T-cell homing molecule expression and cytokine secretion were altered in T-bet-deficient rat hearts. Administration of exogenous interferon-γ (IFN-γ) offset T-bet deficiency-mediated cardioprotection. Cardiomyocytes cultured in T-bet-/- CD4 + T-cell-conditioned media showed a reduced hypertrophic response after hypertrophic stimuli, which was abolished by an IFN-γ-neutralizing antibody. Taken together, our findings show that T-bet deficiency attenuates pressure overload-induced cardiac remodelling in rats. Specifically, targeting T-bet in T cells may be of great importance for the treatment of pathological cardiac remodelling and heart failure.


Subject(s)
Cardiomegaly/metabolism , Cardiomyopathy, Dilated/metabolism , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/deficiency , Th1 Cells/metabolism , Ventricular Remodeling , Adoptive Transfer , Animals , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Chemotaxis, Leukocyte , Cytokines/immunology , Cytokines/metabolism , Gene Knockdown Techniques , Genotype , Humans , Interferon-gamma/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Paracrine Communication , Phenotype , Rats, Sprague-Dawley , Rats, Transgenic , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/transplantation , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics
11.
Nutr Metab Cardiovasc Dis ; 28(11): 1188-1195, 2018 11.
Article in English | MEDLINE | ID: mdl-30143409

ABSTRACT

BACKGROUND AND AIMS: Several studies propose that (-)-epicatechin, a flavonol present in high concentration in the cocoa, has cardioprotective effects. This study aimed to evaluate the impact of (-)-epicatechin on the development of dilated cardiomyopathy in a δ sarcoglycan null mouse model. METHODS AND RESULTS: δ Sarcoglycan null mice were treated for 15 days with (-)-epicatechin. Histological and morphometric analysis of the hearts treated mutant mice showed significant reduction of the vasoconstrictions in the coronary arteries as well as fewer areas with fibrosis and a reduction in the loss of the ventricular wall. On the contrary, it was observed a thickening of this region. By Western blot analysis, it was shown, and increment in the phosphorylation level of eNOS and PI3K/AKT/mTOR/p70S6K proteins in the heart of the (-)-epicatechin treated animals. On the other hand, we observed a significantly decreased level of the atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) heart failure markers. CONCLUSION: All the results indicate that (-)-epicatechin has the potential to prevent the development of dilated cardiomyopathy of genetic origin and encourages the use of this flavonol as a pharmacological therapy for dilated cardiomyopathy and heart failure diseases.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Catechin/pharmacology , Myocytes, Cardiac/drug effects , Sarcoglycans/deficiency , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Coronary Vessels/drug effects , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Disease Models, Animal , Fibrosis , Male , Mice, Knockout , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sarcoglycans/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Vasoconstriction/drug effects
12.
Circ Res ; 116(5): e28-39, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25605649

ABSTRACT

RATIONALE: Sustained activation of Gαq transgenic (Gq) signaling during pressure overload causes cardiac hypertrophy that ultimately progresses to dilated cardiomyopathy. The molecular events that drive hypertrophy decompensation are incompletely understood. Ca(2+)/calmodulin-dependent protein kinase II δ (CaMKIIδ) is activated downstream of Gq, and overexpression of Gq and CaMKIIδ recapitulates hypertrophy decompensation. OBJECTIVE: To determine whether CaMKIIδ contributes to hypertrophy decompensation provoked by Gq. METHODS AND RESULTS: Compared with Gq mice, compound Gq/CaMKIIδ knockout mice developed a similar degree of cardiac hypertrophy but exhibited significantly improved left ventricular function, less cardiac fibrosis and cardiomyocyte apoptosis, and fewer ventricular arrhythmias. Markers of oxidative stress were elevated in mitochondria from Gq versus wild-type mice and respiratory rates were lower; these changes in mitochondrial function were restored by CaMKIIδ deletion. Gq-mediated increases in mitochondrial oxidative stress, compromised membrane potential, and cell death were recapitulated in neonatal rat ventricular myocytes infected with constitutively active Gq and attenuated by CaMKII inhibition. Deep RNA sequencing revealed altered expression of 41 mitochondrial genes in Gq hearts, with normalization of ≈40% of these genes by CaMKIIδ deletion. Uncoupling protein 3 was markedly downregulated in Gq or by Gq expression in neonatal rat ventricular myocytes and reversed by CaMKIIδ deletion or inhibition, as was peroxisome proliferator-activated receptor α. The protective effects of CaMKIIδ inhibition on reactive oxygen species generation and cell death were abrogated by knock down of uncoupling protein 3. Conversely, restoration of uncoupling protein 3 expression attenuated reactive oxygen species generation and cell death induced by CaMKIIδ. Our in vivo studies further demonstrated that pressure overload induced decreases in peroxisome proliferator-activated receptor α and uncoupling protein 3, increases in mitochondrial protein oxidation, and hypertrophy decompensation, which were attenuated by CaMKIIδ deletion. CONCLUSIONS: Mitochondrial gene reprogramming induced by CaMKIIδ emerges as an important mechanism contributing to mitotoxicity in decompensating hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cardiomegaly/enzymology , Cardiomyopathy, Dilated/etiology , Heart Failure/etiology , Mitochondria, Heart/physiology , Acetylcysteine/pharmacology , Animals , Apoptosis , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Disease Progression , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Gene Expression Profiling , Heart Failure/physiopathology , Ion Channels/biosynthesis , Ion Channels/genetics , Ion Channels/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Myocytes, Cardiac/metabolism , Oxidative Stress , PPAR alpha/biosynthesis , PPAR alpha/genetics , Point Mutation , Pressure , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species , Sequence Analysis, RNA , Sulfonamides/pharmacology , Transfection , Uncoupling Protein 3
13.
Echocardiography ; 34(5): 709-715, 2017 May.
Article in English | MEDLINE | ID: mdl-28332315

ABSTRACT

BACKGROUNDS: It's still unclear if different patterns of intraventricular flow dynamics may be detected in patients nonresponders to cardiac resynchronization therapy (CRT) as compared to responders ones. Aim of this study was to evaluate the characteristics of left ventricular (LV) flow dynamics 6-months after CRT to identify Echo-particle imaging velocity (PIV) patterns were more frequently detected in nonresponders patients. METHODS: Thirty-two patients with dilated cardiomyopathy, undergoing CRT, were enrolled in this study. All patients underwent 2D and 3D echo and fluid dynamics assessment 6 months after CRT, during active CRT (CRT-ON) and during a temporarily discontinued state (CRT-OFF). LV volumes systolic and diastolic volumes (LVESV and LVEDV), ejection fraction (LVEF), global longitudinal strain (GLS), systolic dyssynchrony index (SDI), and several geometrical and functional Echo-PIV-derived parameters were calculated. Patients were divided in two groups: "responders" to CRT (decrease in LVESV>15% 6 months after CRT) and "nonresponders." RESULTS: During CRT-OFF, LVEF, GLS were lower, while SDI and LVESV were higher in nonresponders group (P=.030, P=.051, P=.035, and P=.025, respectively). Energy dissipation, vortex area, and vorticity fluctuation were higher in "nonresponders" patients during CRT-OFF (P=.038, P=.054, and P=.035, respectively). During CRT-ON, energy dissipation, vortex area, and vorticity fluctuation further increase in nonresponders patients (P=.020, P=.038, and P=.030, respectively) with a concomitant worsening of SDI (P=.045). CONCLUSION: Our data show a significant worsening in flow-derived parameters in CRT "nonresponders" patients as compared with responders. Further larger longitudinal studies are necessary to assess whether these more chaotic intraventricular flow-patterns may contribute to a persistent adverse remodeling observed in this subset of patients.


Subject(s)
Blood Flow Velocity , Cardiac Resynchronization Therapy/methods , Cardiomyopathy, Dilated/prevention & control , Cardiomyopathy, Dilated/physiopathology , Stroke Volume , Ventricular Dysfunction, Left/prevention & control , Ventricular Dysfunction, Left/physiopathology , Aged , Cardiomyopathy, Dilated/diagnostic imaging , Energy Transfer , Female , Humans , Male , Reproducibility of Results , Sensitivity and Specificity , Treatment Failure , Treatment Outcome , Ventricular Dysfunction, Left/diagnostic imaging
14.
Herz ; 42(7): 669-676, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27832288

ABSTRACT

BACKGROUND: This study evaluated whether subclinical myocardial dysfunction occurs in first-degree relatives of patients with idiopathic dilated cardiomyopathy (IDCM), using strain echocardiographic imaging, before apparent left ventricular (LV) failure is observed. PATIENTS AND METHODS: The study comprised 77 subjects aged 16-63 years who had first-degree relatives with a previous or new diagnosis of IDCM. LV myocardial deformation parameters of the first-degree relatives with normal LVEF (≥55%) values, as assessed using 2D echocardiography, were evaluated. The findings of the first-degree relatives were compared with an age- and sex-matched control group (n = 86). RESULTS: No difference in terms of age, gender, and body surface area was detected between first-degree relatives and controls. First-degree relatives of IDCM patients had significantly lower LVEF (62.04 ± 5.8% vs. 65.65 ± 6.3%, p < 0.001) and FS values (39.4 ± 6.6 vs. 41.45 ± 5.5, p = 0.03) compared with the controls. Assessment of LV deformation parameters revealed that LV global longitudinal strain (-17.34 ± 2.19% vs. -19.21 ± 2.16%, p < 0.001) and strain rate (0.94 ± 0.14 s-1 vs. 1.03 ± 0.14 s-1, p < 0.001), radial strain (34.47 ± 9.14% vs 42.79 ± 11.91%, p < 0.001) and strain rate (1.6 ± 0.38 s-1 vs. 1.75 ± 0.29 s-1, p = 0.006), circumferential strain (-6.07 ± 2.83% vs. -18.29 ± 3.39%, p < 0.001) and strain rate (1.09 ± 0.24 s-1 vs. 1.2 ± 0.25 s-1, p = 0.004), and torsion (10.07 ± 5.18o/cm vs. 12.42 ± 5.78o/cm, p = 0.009) were significantly reduced in first-degree relatives compared with controls. CONCLUSION: LV deformation parameters are impaired in first-degree relatives of patients with IDCM. Screening of this population using standard 2D echocardiography and strain imaging may provide early detection of those with subclinical myocardial dysfunction.


Subject(s)
Cardiomyopathy, Dilated/genetics , Echocardiography/methods , Genetic Testing , Ventricular Dysfunction, Left/genetics , Adolescent , Adult , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/prevention & control , Female , Heart Ventricles/diagnostic imaging , Humans , Male , Middle Aged , Reference Values , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/prevention & control , Young Adult
15.
Clin Sci (Lond) ; 130(4): 289-99, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26566650

ABSTRACT

Increase in the apoptotic molecule Fas ligand (FasL) in serum and cardiomyocytes has been shown to be associated with progressive dilated cardiomyopathy (DCM) and congestive heart failure (CHF) in humans. However, the underlying mechanism(s) of FasL-related deterioration of heart function remain obscure. The aim of the present study is to determine roles of myocardial FasL in the activation of alternative pathways such as extracellular-signal-regulated kinase 1/2 (ERK1/2), inflammation or fibrosis and to identify effective treatments of progressive DCM and advanced CHF. Transgenic mice with cardiomyocyte-specific overexpression of FasL were investigated and treated with an ERK1/2 inhibitor (U-0126), losartan (los), prednisolone (pred) or placebo. Morpho-histological and molecular studies were subsequently performed. FasL mice showed significantly higher mortality compared with wild-type (WT) littermates due to DCM and advanced CHF. Prominent perivascular and interstitial fibrosis, increased interleukin secretion and diffuse CD3-positive cell infiltration were evident in FasL hearts. Up-regulation of the short form of Fas-associated death domain (FADD)-like interleukin 1ß-converting enzyme (FLICE) inhibitory protein (s-FLIP), RIP (receptor-interacting protein) and ERK1/2 and down-regulation of transforming growth factor beta 1 (TGFß1) and nuclear factor-κB (NF-κB) was determined in the myocardium, whereas expression of ERK1/2, periostin (Postn) and osteopontin increased in cardiac fibroblasts. U-0126 and los increased CHF survival by 75% compared with pred and placebo groups. U-0126 had both anti-fibrotic and anti-apoptotic effects, whereas los reduced fibrosis only. Myocardial FasL expression in mice activates differential robust fibrotic, apoptotic and inflammatory responses via ERK1/2 in cardiomyocytes and cardiac fibroblasts inducing DCM and CHF. Blocking the ERK1/2 pathway prevented progression of FasL-induced DCM and CHF by reducing fibrosis, inflammation and apoptosis in the myocardium.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Fas Ligand Protein/metabolism , Heart Failure/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/enzymology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Disease Progression , Enzyme Activation , Fas Ligand Protein/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Genetic Predisposition to Disease , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Heart Failure/prevention & control , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors
16.
Circ Res ; 114(2): 257-65, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24192653

ABSTRACT

RATIONALE: Dysfunctional Parkin-mediated mitophagic culling of senescent or damaged mitochondria is a major pathological process underlying Parkinson disease and a potential genetic mechanism of cardiomyopathy. Despite epidemiological associations between Parkinson disease and heart failure, the role of Parkin and mitophagic quality control in maintaining normal cardiac homeostasis is poorly understood. OBJECTIVE: We used germline mutants and cardiac-specific RNA interference to interrogate Parkin regulation of cardiomyocyte mitochondria and examine functional crosstalk between mitophagy and mitochondrial dynamics in Drosophila heart tubes. METHODS AND RESULTS: Transcriptional profiling of Parkin knockout mouse hearts revealed compensatory upregulation of multiple related E3 ubiquitin ligases. Because Drosophila lack most of these redundant genes, we examined heart tubes of parkin knockout flies and observed accumulation of enlarged hollow donut mitochondria with dilated cardiomyopathy, which could be rescued by cardiomyocyte-specific Parkin expression. Identical abnormalities were induced by cardiomyocyte-specific Parkin suppression using 2 different inhibitory RNAs. Parkin-deficient cardiomyocyte mitochondria exhibited dysmorphology, depolarization, and reactive oxygen species generation without calcium cycling abnormalities, pointing to a primary mitochondrial defect. Suppressing cardiomyocyte mitochondrial fusion in Parkin-deficient fly heart tubes completely prevented the cardiomyopathy and corrected mitochondrial dysfunction without normalizing mitochondrial dysmorphology, demonstrating a central role for mitochondrial fusion in the cardiomyopathy provoked by impaired mitophagy. CONCLUSIONS: Parkin deficiency and resulting mitophagic disruption produces cardiomyopathy in part by contamination of the cardiomyocyte mitochondrial pool through fusion between improperly retained dysfunctional/senescent and normal mitochondria. Limiting mitochondrial contagion by inhibiting organelle fusion shows promise for minimizing organ dysfunction produced by defective mitophagic signaling.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Drosophila Proteins/deficiency , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Dynamics , Mitophagy , Myocytes, Cardiac/enzymology , Ubiquitin-Protein Ligases/deficiency , Animals , Calcium Signaling , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Gene Expression Profiling , Genotype , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/pathology , Mitochondria, Heart/pathology , Mutation , Myocytes, Cardiac/pathology , Phenotype , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Ubiquitin-Protein Ligases/genetics
17.
Pacing Clin Electrophysiol ; 39(8): 848-57, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27198580

ABSTRACT

BACKGROUND: Clinical guidelines on implantable cardioverter defibrillator (ICD) therapy changed significantly in the last decades with potential inherent effects on therapy efficacy. We aimed to study therapy rates in time and the association between therapies and mortality. METHODS: All patients receiving an ICD, primary and secondary prevention, were included in a single-center retrospective registry. Information on first appropriate and inappropriate therapies was documented. Dates of implant were divided in P1: 1996-2001, P2: 2002-2008, and P3: 2009-2014. RESULTS: A total of 727 patients, 84.9% male-66.4% ischemic cardiomyopathy (ICM)-56% primary prevention-mean follow-up 5.2 ± 4.1 years, were included. There was a shift from secondary to primary prevention indications, from ischemic to non-ICM, and from single chamber to cardiac resynchronization therapy defibrillator devices. The annual 1- and 3-year appropriate shock (AS) rate declined from 29.4% and 15.1% in P1, over 13.3% and 9.2% in P2 to 7.8% and 5.7% in P3 (log-rank P < 0.001), while inappropriate shock (IAS) rates remained unchanged (log-rank P = 0.635). After multivariate regression analysis a higher age at implant, lower left ventricular ejection fraction, history of stroke, diabetes mellitus, intake of loop diuretics or digitalis, higher creatinine, and longer QTc were independent predictors of mortality. CONCLUSION: These changes in clinical practice with a shift to primary prevention and rise in non-ICM implants caused a significant decrease in AS incidence, while IAS remained stable. Receiving AS or IAS was not an independent predictor of mortality in our real-life cohort.


Subject(s)
Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/prevention & control , Defibrillators, Implantable/statistics & numerical data , Myocardial Ischemia/mortality , Myocardial Ischemia/prevention & control , Prosthesis Implantation/mortality , Age Distribution , Aged , Belgium/epidemiology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Practice Patterns, Physicians'/statistics & numerical data , Practice Patterns, Physicians'/trends , Prevalence , Prosthesis Implantation/statistics & numerical data , Prosthesis Implantation/trends , Retrospective Studies , Risk Factors , Survival Rate , Time Factors
18.
Environ Toxicol ; 30(10): 1216-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24706507

ABSTRACT

Exposure to tobacco smoke has epidemiologically been linked to the occurrence of cardiovascular disease among nonsmokers but the associated molecular events are not well elucidated yet. When Sprague Dawley rats were exposed to second-hand tobacco cigarette smoke twice a day for a 30 days period at an exposure rate of 10 cigarettes/30 min, they showed adverse effects including reduced left ventricle weight, increased cardiac damages, deteriorated cardiac features, and cardiac fibrosis. Exposure to second-hand smoking (SHS) increased the molecular markers of cardiac fibrosis such as urokinase plasminogen activator and matrix metallopeptidases. The modulations in the protein levels were led by the activation of extracellular signal-regulated kinases (ERK1/2), the transcription factor-specificity protein 1 (SP1), and the fibrogenic master switch-connective for epithelial-mesenchymal transition tissue growth factor there by indicating their effective role in SHS-induced myocardial infraction. Dilong, an edible earthworm extract used in Chinese medicine and its bioactive fibrinolytic enzyme product-lumbrokinase, when administered in rats, restricted the SHS exposure induced cardiac fibrosis and provided cardio-protection. The results show that lumbrokinase and dilong administration can efficiently prevent epidemiological incidence of cardiac disease among SHS-exposed nonsmokers.


Subject(s)
Endopeptidases/pharmacology , Heart/drug effects , Myocardium/pathology , Oligochaeta/enzymology , Tobacco Smoke Pollution/adverse effects , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/prevention & control , Endopeptidases/isolation & purification , Fibrosis , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism
19.
Circulation ; 127(23): 2285-94, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23671208

ABSTRACT

BACKGROUND: Activation of innate pattern-recognition receptors promotes CD4+ T-cell-mediated autoimmune myocarditis and subsequent inflammatory cardiomyopathy. Mechanisms that counterregulate exaggerated heart-specific autoimmunity are poorly understood. METHODS AND RESULTS: Experimental autoimmune myocarditis was induced in BALB/c mice by immunization with α-myosin heavy chain peptide and complete Freund's adjuvant. Together with interferon-γ, heat-killed Mycobacterium tuberculosis, an essential component of complete Freund's adjuvant, converted CD11b(hi)CD11c(-) monocytes into tumor necrosis factor-α- and nitric oxide synthase 2-producing dendritic cells (TipDCs). Heat-killed M. tuberculosis stimulated production of nitric oxide synthase 2 via Toll-like receptor 2-mediated nuclear factor-κB activation. TipDCs limited antigen-specific T-cell expansion through nitric oxide synthase 2-dependent nitric oxide production. Moreover, they promoted nitric oxide synthase 2 production in hematopoietic and stromal cells in a paracrine manner. Consequently, nitric oxide synthase 2 production by both radiosensitive hematopoietic and radioresistant stromal cells prevented exacerbation of autoimmune myocarditis in vivo. CONCLUSIONS: Innate Toll-like receptor 2 stimulation promotes formation of regulatory TipDCs, which confine autoreactive T-cell responses in experimental autoimmune myocarditis via nitric oxide. Therefore, activation of innate pattern-recognition receptors is critical not only for disease induction but also for counterregulatory mechanisms, protecting the heart from exaggerated autoimmunity.


Subject(s)
Autoimmune Diseases/physiopathology , Dendritic Cells/metabolism , Immune Tolerance/physiology , Interferon-gamma/physiology , Myocarditis/physiopathology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/biosynthesis , Signal Transduction , T-Lymphocytes, Helper-Inducer/pathology , Toll-Like Receptor 2/physiology , Animals , Autoimmune Diseases/immunology , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/prevention & control , Cell Differentiation/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/immunology , Enzyme Induction/drug effects , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/radiation effects , Immune Tolerance/immunology , Mice , Mice, Inbred Strains , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Mycobacterium tuberculosis/immunology , Myocarditis/immunology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Paracrine Communication , Peptide Fragments/immunology , Peptide Fragments/toxicity , Radiation Chimera , Radiation Tolerance , Stromal Cells/enzymology , Stromal Cells/radiation effects , T-Lymphocytes, Helper-Inducer/immunology , Ventricular Myosins/immunology , Ventricular Myosins/toxicity
20.
Am J Physiol Heart Circ Physiol ; 307(6): H922-32, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25038143

ABSTRACT

We have previously reported that ectopic trypsin in the myocardium triggers acute myocarditis after influenza A virus (IAV) infection. As myocarditis is a common precursor to dilated cardiomyopathy (DCM), the aim of the present study was to investigate the influence of trypsin on the progression of DCM after IAV infection. IAV-infected mice treated with saline or trypsin inhibitor were euthanized on days 0, 9, 20, 40 and 60 postinfection. Trypsin expression colocalized with myocardial inflammatory loci and IAV-induced myocarditis peaked on day 9 postinfection and alleviated by day 20 but persisted until day 60 postinfection, even though replication of IAV was not detected from day 20 postinfection. Similar time courses were observed for the activation of pro-matrix metalloproteinase (pro-MMP)-9 and expression of the proinflammatory cytokines IL-6, IL-1ß, and TNF-α. Degradation of collagen type I, proliferation of ventricular interstitial collagen, and expression of collagen type I and III mRNA increased significantly during acute and chronic phases; collagen type III mRNA increased more significantly than collagen type I mRNA. Cardiac function progressively deteriorated with progressive left ventricular dilation. The trypsin inhibitor aprotinin suppressed pro-MMP-9 activation and cytokine release, alleviated myocardial inflammation, and restored collagen metabolism during acute and chronic phases of myocarditis. This effectively prevented ventricular dilation and improved cardiac function. These results suggest that ectopic trypsin in the myocardium promoted DCM through chronic activation of pro-MMP-9, persistent induction of cytokines, and mediation of collagen remodeling. Pharmacological inhibition of trypsin activity might be a promising approach for the prevention of viral cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Influenza A Virus, H1N1 Subtype/pathogenicity , Myocarditis/prevention & control , Myocardium/enzymology , Orthomyxoviridae Infections/complications , Trypsin/metabolism , Animals , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/virology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Disease Models, Animal , Disease Progression , Enzyme Precursors/metabolism , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/prevention & control , Hypertrophy, Left Ventricular/virology , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/physiopathology , Myocarditis/virology , Orthomyxoviridae Infections/virology , RNA, Messenger/metabolism , Time Factors , Trypsin Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/prevention & control , Ventricular Dysfunction, Left/virology , Ventricular Function, Left , Ventricular Remodeling , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL