Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Nat Commun ; 13(1): 81, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013172

ABSTRACT

Despite the high prevalence of ischemic heart diseases worldwide, no antibody-based treatment currently exists. Starting from the evidence that a specific isoform of the Bone Morphogenetic Protein 1 (BMP1.3) is particularly elevated in both patients and animal models of myocardial infarction, here we assess whether its inhibition by a specific monoclonal antibody reduces cardiac fibrosis. We find that this treatment reduces collagen deposition and cross-linking, paralleled by enhanced cardiomyocyte survival, both in vivo and in primary cultures of cardiac cells. Mechanistically, we show that the anti-BMP1.3 monoclonal antibody inhibits Transforming Growth Factor ß pathway, thus reducing myofibroblast activation and inducing cardioprotection through BMP5. Collectively, these data support the therapeutic use of anti-BMP1.3 antibodies to prevent cardiomyocyte apoptosis, reduce collagen deposition and preserve cardiac function after ischemia.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Morphogenetic Protein 1/genetics , Cardiotonic Agents/pharmacology , Cicatrix/genetics , Endomyocardial Fibrosis/genetics , Myocardial Infarction/genetics , Animals , Bone Morphogenetic Protein 1/antagonists & inhibitors , Bone Morphogenetic Protein 1/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 5/genetics , Bone Morphogenetic Protein 5/metabolism , Case-Control Studies , Cell Survival/drug effects , Cicatrix/etiology , Cicatrix/metabolism , Cicatrix/prevention & control , Disease Models, Animal , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/prevention & control , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Troponin T/genetics , Troponin T/metabolism
2.
Nat Commun ; 13(1): 149, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013221

ABSTRACT

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Subject(s)
Cachexia/genetics , Endomyocardial Fibrosis/genetics , Heart Failure/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Transcription Factors/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Aged , Aged, 80 and over , Animals , Cachexia/metabolism , Cachexia/physiopathology , Cachexia/prevention & control , Case-Control Studies , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/physiopathology , Endomyocardial Fibrosis/prevention & control , Female , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/deficiency , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency
3.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768917

ABSTRACT

Aging is the major risk factor for cardiovascular disease, which is the leading cause of mortality worldwide among aging populations. Cisd2 is a prolongevity gene that mediates lifespan in mammals. Previously, our investigations revealed that a persistently high level of Cisd2 expression in mice is able to prevent age-associated cardiac dysfunction. This study was designed to apply a genetic approach that induces cardiac-specific Cisd2 overexpression (Cisd2 icOE) at a late-life stage, namely a time point immediately preceding the onset of old age, and evaluate the translational potential of this approach. Several discoveries are pinpointed. Firstly, Cisd2 is downregulated in the aging heart. This decrease in Cisd2 leads to cardiac dysfunction and impairs electromechanical performance. Intriguingly, Cisd2 icOE prevents an exacerbation of age-associated electromechanical dysfunction. Secondly, Cisd2 icOE ameliorates cardiac fibrosis and improves the integrity of the intercalated discs, thereby reversing various structural abnormalities. Finally, Cisd2 icOE reverses the transcriptomic profile of the aging heart, changing it from an older-age pattern to a younger pattern. Intriguingly, Cisd2 icOE modulates a number of aging-related pathways, namely the sirtuin signaling, autophagy, and senescence pathways, to bring about rejuvenation of the heart as it enters old age. Our findings highlight Cisd2 as a novel molecular target for developing therapies targeting cardiac aging.


Subject(s)
Aging/genetics , Autophagy-Related Proteins/genetics , Endomyocardial Fibrosis/genetics , Heart/physiology , Longevity/genetics , Nerve Tissue Proteins/genetics , Rejuvenation/physiology , Animals , Autophagy/genetics , Autophagy-Related Proteins/biosynthesis , Cellular Senescence/genetics , Endomyocardial Fibrosis/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Sirtuins/metabolism , Transcriptome/genetics
4.
Cells ; 10(7)2021 07 06.
Article in English | MEDLINE | ID: mdl-34359886

ABSTRACT

Cardiac fibrosis is the excess deposition of extracellular matrix (ECM), such as collagen. Myofibroblasts are major players in the production of collagen, and are differentiated primarily from resident fibroblasts. Collagen can compensate for the dead cells produced by injury. The appropriate production of collagen is beneficial for preserving the structural integrity of the heart, and protects the heart from cardiac rupture. However, excessive deposition of collagen causes cardiac dysfunction. Recent studies have demonstrated that myofibroblasts can change their phenotypes. In addition, myofibroblasts are found to have functions other than ECM production. Myofibroblasts have macrophage-like functions, in which they engulf dead cells and secrete anti-inflammatory cytokines. Research into fibroblasts has been delayed due to the lack of selective markers for the identification of fibroblasts. In recent years, it has become possible to genetically label fibroblasts and perform sequencing at single-cell levels. Based on new technologies, the origins of fibroblasts and myofibroblasts, time-dependent changes in fibroblast states after injury, and fibroblast heterogeneity have been demonstrated. In this paper, recent advances in fibroblast and myofibroblast research are reviewed.


Subject(s)
Endomyocardial Fibrosis/pathology , Extracellular Matrix/pathology , Fibroblasts/pathology , Macrophages/pathology , Myofibroblasts/pathology , Animals , Cardiotonic Agents/therapeutic use , Cell Differentiation , Cell Lineage , Collagen/genetics , Collagen/metabolism , Cytokines/genetics , Cytokines/metabolism , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/classification , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Macrophages/drug effects , Macrophages/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Signal Transduction
5.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34302205

ABSTRACT

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Subject(s)
Angiotensin II/pharmacology , Antifibrotic Agents/pharmacology , Bone Morphogenetic Protein 4/genetics , Fibroblasts/drug effects , MAP Kinase Kinase 4/genetics , Vinca Alkaloids/pharmacology , Animals , Animals, Newborn , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Collagen/genetics , Collagen/metabolism , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Phosphorylation/drug effects , Primary Cell Culture , Rats , Rats, Wistar , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Am J Physiol Cell Physiol ; 319(5): C781-C796, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32845719

ABSTRACT

Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.


Subject(s)
Cardiomyopathy, Dilated/genetics , Endomyocardial Fibrosis/genetics , Mucocutaneous Lymph Node Syndrome/genetics , Myocardial Infarction/genetics , Myocarditis/genetics , Tenascin/genetics , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Gene Expression Regulation , Humans , Inflammation , Mice , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Tenascin/metabolism , Ventricular Remodeling/genetics , Wound Healing/genetics
7.
Mol Biochem Parasitol ; 238: 111283, 2020 07.
Article in English | MEDLINE | ID: mdl-32564978

ABSTRACT

Posaconazole (POS) is an inhibitor of ergosterol biosynthesis in clinical use for treating invasive fungal infections. POS has potent and selective anti-Trypanosoma cruzi activity and has been evaluated as a possible treatment for Chagas disease. Microtissues are a 3D culture system that has been shown to reproduce better tissue architecture and functionality than cell cultures in monolayer (2D). It has been used to evaluate chemotropic response as in vitro disease models. We previously developed an in vitro model that reproduces aspects of cardiac fibrosis observed in Chagas cardiomyopathy, using microtissues formed by primary cardiac cells infected by the T. cruzi, here called T. cruzi fibrotic cardiac microtissue (TCFCM). We also showed that the treatment of TCFCM with a TGF-ß pathway inhibitor reduces fibrosis. Here, we aimed to evaluate the effect of POS in TCFCM, observing parasite load and molecules involved in fibrosis. To choose the concentration of POS to be used in TCFCM we first performed experiments in a monolayer of primary cardiac cell cultures and, based on the results, TCFCM was treated with 5 nM of POS for 96 h, starting at 144 h post-infection. Our previous studies showed that at this time the TCFCM had established fibrosis, resulting from T. cruzi infection. Treatment with POS of TCFCM reduced 50 % of parasite load as observed by real-time PCR and reduced markedly the fibrosis as observed by western blot and immunofluorescence, associated with a strong reduction in the expression of fibronectin and laminin (45 % and 54 %, respectively). POS treatment also changed the expression of proteins involved in the regulation of extracellular matrix proteins (TGF-ß and TIMP-4, increased by 50 % and decreased by 58 %, respectively) in TCFCM. In conclusion, POS presented a potent trypanocidal effect both in 2D and in TCFCM, and the reduction of the parasite load was associated with a reduction of fibrosis in the absence of external immunological effectors.


Subject(s)
Chagas Cardiomyopathy/drug therapy , Endomyocardial Fibrosis/drug therapy , Myocytes, Cardiac/drug effects , Triazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Animals , Cell Culture Techniques , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/parasitology , Endomyocardial Fibrosis/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fetus , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Inhibitory Concentration 50 , Laminin/genetics , Laminin/metabolism , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , Parasite Load , Primary Cell Culture , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/pathogenicity , Tissue Inhibitor of Metalloproteinase-4
8.
Matrix Biol ; 91-92: 167-175, 2020 09.
Article in English | MEDLINE | ID: mdl-32438054

ABSTRACT

Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.


Subject(s)
Endomyocardial Fibrosis/immunology , Extracellular Matrix/immunology , Fibroblasts/immunology , Heart Failure/immunology , Myocardial Infarction/immunology , T-Lymphocytes/immunology , Age Factors , Antigens, CD/genetics , Antigens, CD/immunology , Cell Communication/genetics , Cell Communication/immunology , Cytokines/genetics , Cytokines/immunology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibroblasts/pathology , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Humans , Inflammation , Macrophages/immunology , Macrophages/pathology , Male , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Signal Transduction , T-Lymphocytes/classification , T-Lymphocytes/pathology
9.
Matrix Biol ; 91-92: 117-135, 2020 09.
Article in English | MEDLINE | ID: mdl-32416242

ABSTRACT

Fibroblasts are the primary regulator of cardiac extracellular matrix (ECM). In response to disease stimuli cardiac fibroblasts undergo cell state transitions to a myofibroblast phenotype, which underlies the fibrotic response in the heart and other organs. Identifying regulators of fibroblast state transitions would inform which pathways could be therapeutically modulated to tactically control maladaptive extracellular matrix remodeling. Indeed, a deeper understanding of fibroblast cell state and plasticity is necessary for controlling its fate for therapeutic benefit. p38 mitogen activated protein kinase (MAPK), which is part of the noncanonical transforming growth factor ß (TGFß) pathway, is a central regulator of fibroblast to myofibroblast cell state transitions that is activated by chemical and mechanical stress signals. Fibroblast intrinsic signaling, local and global cardiac mechanics, and multicellular interactions individually and synergistically impact these state transitions and hence the ECM, which will be reviewed here in the context of cardiac fibrosis.


Subject(s)
Endomyocardial Fibrosis/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Myocardial Infarction/genetics , Myofibroblasts/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Cell Differentiation , Cell Lineage/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/pathology , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Humans , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/pathology , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Matrix Biol ; 91-92: 75-91, 2020 09.
Article in English | MEDLINE | ID: mdl-32446910

ABSTRACT

The cardiac stroma plays essential roles in health and following cardiac damage. The major player of the stroma with respect to extracellular matrix deposition, maintenance and remodeling is the poorly defined fibroblast. It has long been recognized that there is considerable variability to the fibroblast phenotype. With the advent of new, high throughput analytical methods our understanding and appreciation of this heterogeneity has grown dramatically. This review aims to explore the diversity of cardiac fibroblasts and highlights new insights into the diverse nature of these cells and their progenitors as revealed by single cell sequencing and fate mapping studies. We propose that at least in part the observed heterogeneity is related to the existence of a differentiation cascade within stromal cells. Beyond in-organ heterogeneity, we also discuss how the stromal response to damage differs between non-regenerating organs such as the heart and regenerating organs such as skeletal muscle. In exploring possible causes for these differences, we outline that although fibrogenic cells from different organs overlap in many properties, they still possess organ-specific transcriptional signatures and differentiation biases that make them functionally distinct.


Subject(s)
Endomyocardial Fibrosis/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Myofibroblasts/metabolism , Stromal Cells/metabolism , Animals , Cell Differentiation , Cell Lineage/genetics , Cell Tracking/methods , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myocardium/cytology , Myocardium/metabolism , Myofibroblasts/cytology , Organ Specificity , Signal Transduction , Single-Cell Analysis/methods , Stromal Cells/cytology , Transcription, Genetic
11.
Bull Exp Biol Med ; 168(5): 658-661, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32246376

ABSTRACT

Reduced expression of the key regulator of cardiac metabolism, transcription factor PPARα, in surgical samples of the auricles from patients with coronary heart disease and heart failure was detected by real-time quantitative PCR. These changes indicate reduced activity of this factor and a shift of energy metabolism from oxidative phosphorylation to glycolysis typical of dedifferentiated cells. Electron microscopy revealed dedifferentiated cardiomyocytes with disassembled contractile apparatus and disorganized sarcomeres. In the examined specimens from patients with heart failure, severe myocardial fibrosis was revealed.


Subject(s)
Energy Metabolism/physiology , Heart/physiology , Myocytes, Cardiac/metabolism , PPAR alpha/physiology , Regeneration/physiology , Biopsy , Cell Dedifferentiation/genetics , Coronary Disease/genetics , Coronary Disease/metabolism , Coronary Disease/pathology , Coronary Disease/physiopathology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Energy Metabolism/genetics , Gene Expression Regulation , Glycolysis/genetics , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Oxidative Phosphorylation , PPAR alpha/genetics , PPAR alpha/metabolism
12.
Mol Biol Rep ; 47(4): 2605-2617, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32130618

ABSTRACT

Atrial fibrillation (AF) is a commonly encountered heart arrhythmia and a risk factor for cardiovascular system. The purpose of the present study was to explore the role of long non-coding RNA myocardial infarction-associated transcript (MIAT) in AF and AF-induced myocardial fibrosis and the possible mechanisms involved in this process. We successfully induced an AF rat model. Expression of MIAT presented a dramatic increase, while microRNA (miR)-133a-3p presented a dramatic decrease in atrium tissues of rats with AF induction. In addition, we also found that MIAT was highly expressed and miR-133a-3p was significantly reduced in peripheral blood leukocyte of AF patients. For biological function exploration of MIAT/miR-133a-3p axis, MIAT was knocked down using small hairpin RNA (shRNA) lentivirus injection and the rescue experiments were performed simultaneously by inhibiting miR-133a-3p using anti-miR-133a-3p lentivirus injection in rats with AF. MIAT downregulation significantly alleviated AF, increased atrial effective refractory period (AERP), and reduced the duration of AF as well as cardiomyocytes apoptosis. Whereas these effects of MIAT downregulation on AF were reversed by anti-miR-133a-3p administration. Luciferase reporter revealed that miR-133a-3p was directly regulated by MIAT. Moreover, MIAT knockdown effectively reduced AF-induced atrial fibrosis by detecting reduced collagen in the right atria and inhibited expression of fibrosis-related gene expression of collagen I, collagen III, connective tissue growth factor (CTGF) and transforming growth factor-ß1 (TGF-ß1) in rats with AF, these findings were in contrast with the findings for rats with inhibition of miR-133a-3p. In conclusion, our study demonstrated the role of MIAT downregulation in alleviating AF and AF-induced myocardial fibrosis, and the functional regulatory pathway of MIAT targeting miR-133a-3p.


Subject(s)
Atrial Fibrillation/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Apoptosis/genetics , Atrial Fibrillation/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , China , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Female , Fibrosis/metabolism , Humans , Male , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , RNA, Long Noncoding/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics
13.
Matrix Biol ; 91-92: 188-203, 2020 09.
Article in English | MEDLINE | ID: mdl-32205152

ABSTRACT

The involvement of fibrosis as an underlying pathology in heart diseases is becoming increasingly clear. In recent years, fibrosis has been granted a causative role in heart diseases and is now emerging as a major contributor to Atrial Fibrillation (AF) pathogenesis. AF is the most common arrhythmia encountered in the clinic, but the substrate for AF is still being debated. Consensus in the field is a combination of cardiac tissue remodeling, inflammation and genetic predisposition. The extracellular matrix (ECM) is subject of growing investigation, since measuring circulatory biomarkers of ECM formation and degradation provides both diagnostic and prognostic information. However, fibrosis is not just fibrosis. Each specific collagen biomarker holds information on regulatory mechanisms, as well as information about which section of the ECM is being remodeled, providing a detailed description of cardiac tissue homeostasis. This review entails an overview of the implication of fibrosis in AF, the different collagens and their significance, and the potential of using biomarkers of ECM remodeling as tools for understanding AF pathogenesis and identifying patients at risk for further disease progression.


Subject(s)
Atrial Fibrillation/blood , Endomyocardial Fibrosis/blood , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , Atrial Fibrillation/diagnosis , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Biomarkers/blood , Cytokines/blood , Cytokines/genetics , Endomyocardial Fibrosis/diagnosis , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/pathology , Extracellular Matrix Proteins/blood , Fibroblasts/pathology , Gene Expression Regulation , Heart Atria/metabolism , Heart Atria/pathology , Heart Conduction System/metabolism , Heart Conduction System/pathology , Homeostasis/genetics , Humans , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Prognosis , Proteolysis , Signal Transduction
14.
Aging Cell ; 18(3): e12926, 2019 06.
Article in English | MEDLINE | ID: mdl-30834643

ABSTRACT

Biological aging dynamically alters normal immune and cardiac function, favoring the production of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) and increased instances of cardiac distress. Cardiac failure is the primary reason for hospitalization of the elderly (65+ years). The elderly are also increasingly susceptible to developing chronic bacterial infections due to aging associated immune abnormalities. Since bacterial infections compound the rates of cardiac failure in the elderly, and this phenomenon is not entirely understood, the interplay between the immune system and cardiovascular function in the elderly is of great interest. Using Mycobacterium avium, an opportunistic pathogen, we investigated the effect of mycobacteria on cardiac function in aged mice. Young (2-3 months) and old (18-20 months) C57BL/6 mice were intranasally infected with M. avium strain 104, and we compared the bacterial burden, immune status, cardiac electrical activity, pathology, and function of infected mice against uninfected age-matched controls. Herein, we show that biological aging may predispose old mice infected with M. avium to mycobacterial dissemination into the heart tissue and this leads to cardiac dysfunction. M. avium infected old mice had significant dysrhythmia, cardiac hypertrophy, increased recruitment of CD45+ leukocytes, cardiac fibrosis, and increased expression of inflammatory genes in isolated heart tissue. This is the first study to report the effect of mycobacteria on cardiac function in an aged model. Our findings are critical to understanding how nontuberculous mycobacterium (NTM) and other mycobacterial infections contribute to cardiac dysfunction in the elderly population.


Subject(s)
Arrhythmias, Cardiac/microbiology , Cardiomegaly/microbiology , Endomyocardial Fibrosis/microbiology , Mycobacterium Infections, Nontuberculous/immunology , Nontuberculous Mycobacteria , Aging/immunology , Aging/pathology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Disease Susceptibility , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Inflammation/microbiology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Leukocyte Common Antigens/immunology , Mice , Mice, Inbred C57BL , Mycobacterium Infections, Nontuberculous/pathology , Mycobacterium avium , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
15.
DNA Cell Biol ; 37(10): 821-830, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30227089

ABSTRACT

Premenopausal women have a reduced risk for cardiovascular disease. Estrogen deficiency augments cardiac inflammation and oxidative stress and, thereby, aggravates myocardial fibrosis (MF) and diastolic dysfunction in hypertensive female rats. However, estrogen replacement therapy has no effect on myocardial infarction and cardiac fibrosis in postmenopausal women. Further clinical studies showed that high blood glucose levels in patients with diabetes is an important cause of MF, but the underlying mechanism is unclear. To experimentally address this issue, diabetes mellitus (DM) was induced by injecting streptozotocin and administering a high-fat diet in ovariectomized (OVX) rats. High degrees of fibrosis and apoptosis were detected in the cardiac tissue of these rats, together with increased expression of iNOS. Further treatment with the G protein-coupled estrogen receptor 30 (GPR30) agonist G1 decreased iNOS expression and the apoptosis rate in cardiac tissue significantly and inhibited cardiac fibroblast (CF) proliferation. Similar trends were observed in cultured CFs treated with high concentrations of fat and glucose. In addition, treatment with the iNOS-specific inhibitor W1400 attenuated iNOS and vimentin expression, which is associated with a marked reduction in MF. These results suggest that GPR30 activation inhibits MF in diabetic OVX female rats by suppressing cardiac iNOS activity and consequently NO levels. Thus, GPR30 activation may provide novel cardioprotection strategies for postmenopausal women, especially those with DM.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Endomyocardial Fibrosis/genetics , Fibroblasts/metabolism , Nitric Oxide Synthase Type II/genetics , Ovariectomy , Receptors, G-Protein-Coupled/genetics , Amidines/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/prevention & control , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation , Humans , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Streptozocin , Vimentin/genetics , Vimentin/metabolism
16.
Adv Exp Med Biol ; 1087: 259-273, 2018.
Article in English | MEDLINE | ID: mdl-30259373

ABSTRACT

Fibrosis refers to a process involving the accumulation of extracellular matrix components. It could happen in chronic organ injury or during the recovery of acute organ injury. The severity of fibrosis interferes with the function of the organ involved. Numerous studies have been carried out to explore the mechanism of fibrosis, including parenchyma injury, fibrillar ECM accumulation, fibroblast activation, microvasculature rarefaction, and a mononuclear infiltrate. Unfortunately, its underlying mechanism is at largely unknown. The studying of noncoding RNAs has provided novel insight for circRNA-miRNA-mRNA in learning disease progress. Emerging evidence has shown that circRNA is related to fibrosis activity and could potentially be a monitoring factor for fibrosis or, more excitingly, could be a target for treatment. In this chapter, we will first present the basic mechanism of organ fibrosis. Then we will focus on the recent studies about how circRNA dysregulation contributes to organ fibrosis. Finally, the advantages and potential challenges of circRNA-based therapeutics for the treatment of fibroproliferative diseases will be discussed.


Subject(s)
Fibrosis/genetics , RNA/genetics , Animals , Disease Models, Animal , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Fibroblasts/metabolism , Fibrosis/diagnosis , Fibrosis/physiopathology , Fibrosis/therapy , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Microcirculation , Organ Specificity , Prognosis , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , RNA/metabolism , RNA, Circular , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
17.
Toxicology ; 410: 182-192, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30114436

ABSTRACT

Modulation of epigenetic marks has promised efficacy for treating fibrosis. Cardiac fibroblast is the primary source of activated myofibroblasts that produce extracellular matrix (ECM) in cardiac fibrosis, but the mechanisms underlying this process are incompletely understood. Here we show that microRNA-369-5p (miR-369-5p) through DNMT3A hypermethylation and suppression of the Patched1 pathway leads to fibroblast proliferation in cardiac fibrosis. Forty adult male Sprague-Dawley (SD) rats were randomly divided into two groups (sham and AAC group), cardiac fibrosis was produced by abdominal aortic constriction, and the operation of abdominal aortic constriction was carried out according to the method described. Cardiac fibroblasts (CFs) were harvested from SD neonate rats and cultured. Importantly, miR-369-5p bind directly to DNMT3A with high affinity. MiR-369-5p leads to inhibition of DNMT3A enzyme activity. Exogenous miR-369-5p in cells induces aberrant DNA methylation of the Patched1, resulting in hypermethylation of low to moderately methylated regions. Moreover, Overexpression of miR-369-5p in cardiac fibroblast cells inhibits proliferation. We identify DNMT3A as miR-369-5p target genes and demonstrate that inhibition of miR-369-5p expression augments cell proliferation by activating DNMT3A and suppression of the Patched1 pathway. Together, our results highlight miR-369-5p mediated DNMT3A epigenetic silencing of Patched1 as a mechanism of fibroblast proliferation in cardiac fibrosis.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Endomyocardial Fibrosis/genetics , Epigenesis, Genetic/drug effects , MicroRNAs/genetics , Patched-1 Receptor/genetics , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Constriction, Pathologic , DNA Methylation/genetics , DNA Methyltransferase 3A , Echocardiography, Doppler , Endomyocardial Fibrosis/diagnostic imaging , Epigenesis, Genetic/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Fibroblasts/pathology , Male , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
18.
Med Sci Monit ; 24: 4121-4127, 2018 Jun 16.
Article in English | MEDLINE | ID: mdl-29908129

ABSTRACT

BACKGROUND The role of miR-181a in the development of cardiac disease and in particular, myocardial fibrosis following myocardial infarction (MI) remains unknown. The aim of this study was to explore the role of miR-181a in myocardial fibrosis in a rat model of MI and the expression of TGF-ß receptor III (TßRIII). MATERIAL AND METHODS Forty adult male Wistar rats were randomly divided into an MI model group (n=30) and a control group with (n=10). The rat MI model involved ligating the left anterior descending (LAD) coronary artery in the model group; the control group was treated with a sham operation. Cardiac function was assessed using cardiac ultrasound. Myocardial fibroblasts were extracted from the rat hearts and transfected with a miR-mimic or miR-inhibitor, and cell growth was measured using an MTT assay. The level of miR-181a expression was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blots. RESULTS miR-181a expression was significantly increased during the progression of MI (P<0.05). Over-expression of miR-181a was associated with increased deposition of extracellular matrix (ECM) components, collagen I and fibronectin. This effect was reversed with the use of a miR-181a inhibitor (P<0.05). Upregulation of miR-181a suppressed the expression of TGF-ß receptor III (TßRIII) by binding with 3'-UTR. CONCLUSIONS In this rat model of MI, the findings were that miR-181a had a role in the progression of myocardial fibrosis. The findings require further studies to determine whether miR-181a might provide a novel therapeutic target to limit myocardial fibrosis following MI.


Subject(s)
Endomyocardial Fibrosis/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Animals , Cell Proliferation/physiology , Collagen Type I/metabolism , Disease Models, Animal , Disease Progression , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Myocardial Infarction/metabolism , Proteoglycans/biosynthesis , Proteoglycans/genetics , Rats , Rats, Wistar , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics
19.
Cardiol J ; 25(6): 732-741, 2018.
Article in English | MEDLINE | ID: mdl-29240957

ABSTRACT

Non-coding RNAs (ncRNAs) are a class of RNA molecules that do not encode proteins. ncRNAs are involved in cell proliferation, apoptosis, differentiation, metabolism, and other physiological processes as well as the pathogenesis of diseases. Cardiac fibrosis is increasingly recognized as a common final pathway in advanced heart diseases. Many studies have shown that the occurrence and development of cardiac fibrosis is closely related to the regulation of ncRNAs. This review will highlight recent updates regarding the involvement of ncRNAs in cardiac fibrosis, and their potential as emerging biomarkers and therapeutic targets.


Subject(s)
Endomyocardial Fibrosis , Genetic Therapy/methods , Myocardium/metabolism , RNA, Untranslated/genetics , Apoptosis/genetics , Biomarkers/metabolism , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/therapy , Humans , Myocardium/pathology , RNA, Untranslated/metabolism
20.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 11-23, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28987763

ABSTRACT

Cardiac dysfunction with progressive inflammation and fibrosis is a hallmark of Chagas disease caused by persistent Trypanosoma cruzi infection. Osteopontin (OPN) is a pro-inflammatory cytokine that orchestrates mechanisms controlling cell recruitment and cardiac architecture. Our main goal was to study the role of endogenous OPN as a modulator of myocardial CCL5 chemokine and MMP-2 metalloproteinase, and its pathological impact in a murine model of Chagas heart disease. Wild-type (WT) and OPN-deficient (spp1 -/-) mice were parasite-infected (Brazil strain) for 100days. Both groups developed chronic myocarditis with similar parasite burden and survival rates. However, spp1 -/- infection showed lower heart-to-body ratio (P<0.01) as well as reduced inflammatory pathology (P<0.05), CCL5 expression (P<0.05), myocyte size (P<0.05) and fibrosis (P<0.01) in cardiac tissues. Intense OPN labeling was observed in inflammatory cells recruited to infected heart (P<0.05). Plasma concentration of MMP-2 was higher (P<0.05) in infected WT than in spp1 -/- mice. Coincidently, specific immunostaining revealed increased gelatinase expression (P<0.01) and activity (P<0.05) in the inflamed hearts from T. cruzi WT mice, but not in their spp1 -/- littermates. CCL5 and MMP-2 induction occurred preferentially (P<0.01) in WT heart-invading CD8+ T cells and was mediated via phospho-JNK MAPK signaling. Heart levels of OPN, CCL5 and MMP-2 correlated (P<0.01) with collagen accumulation in the infected WT group only. Endogenous OPN emerges as a key player in the pathogenesis of chronic Chagas heart disease, through the upregulation of myocardial CCL5/MMP-2 expression and activities resulting in pro-inflammatory and pro-hypertrophic events, cardiac remodeling and interstitial fibrosis.


Subject(s)
Atrial Remodeling , Chagas Cardiomyopathy , Chemokine CCL5/metabolism , Matrix Metalloproteinase 2/metabolism , Myocarditis , Osteopontin/physiology , Ventricular Remodeling , Animals , Atrial Remodeling/genetics , Atrial Remodeling/immunology , Cells, Cultured , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/immunology , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Disease Models, Animal , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Osteopontin/genetics , Ventricular Remodeling/genetics , Ventricular Remodeling/immunology
SELECTION OF CITATIONS
SEARCH DETAIL