Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 14(2)2021 02 22.
Article in English | MEDLINE | ID: mdl-33619211

ABSTRACT

Besides skeletal muscle abnormalities, Duchenne muscular dystrophy (DMD) patients present with dilated cardiomyopathy development, which considerably contributes to morbidity and mortality. Because the mechanisms responsible for the cardiac complications in the context of DMD are largely unknown, evidence-based therapy approaches are still lacking. This has increased the need for basic research efforts into animal models for DMD. Here, we characterized in detail the cardiovascular abnormalities of Dmdmdx rats, with the aim of determining the suitability of this recently established dystrophin-deficient small animal as a model for DMD.Various methods were applied to compare cardiovascular properties between wild-type and Dmdmdx rats, and to characterize the Dmdmdx cardiomyopathy. These methods comprised echocardiography, invasive assessment of left ventricular hemodynamics, examination of adverse remodeling and endothelial cell inflammation, and evaluation of vascular function, employing wire myography. Finally, intracellular Ca2+ transient measurements, and recordings of currents through L-type Ca2+ channels were performed in isolated single ventricular cardiomyocytes. We found that, similar to respective observations in DMD patients, the hearts of Dmdmdx rats show significantly impaired cardiac function, fibrosis and inflammation, consistent with the development of a dilated cardiomyopathy. Moreover, in Dmdmdx rats, vascular endothelial function is impaired, which may relate to inflammation and oxidative stress, and Ca2+ handling in Dmdmdx cardiomyocytes is abnormal.These findings indicate that Dmdmdx rats represent a promising small-animal model to elucidate mechanisms of cardiomyopathy development in the dystrophic heart, and to test mechanism-based therapies aiming to combat cardiovascular complications in DMD.


Subject(s)
Cardiomyopathies/pathology , Cardiovascular System , Disease Models, Animal , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Myocardium/pathology , Myocytes, Cardiac/pathology , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Cardiomyopathy, Dilated/complications , Dystrophin/metabolism , Endothelium, Vascular/pathology , Fibrosis/pathology , Heart Ventricles/physiopathology , Hemodynamics , Homeostasis , Humans , Inflammation , Kidney/metabolism , Lung/metabolism , Muscle, Skeletal/pathology , Myocytes, Cardiac/metabolism , Oxidative Stress , Peptidyl-Dipeptidase A , Phenotype , Rats , Stress, Mechanical
2.
Basic Res Cardiol ; 115(6): 76, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33258993

ABSTRACT

Ischemic mitral regurgitation (MR) is a frequent complication of myocardial infarction (MI) characterized by adverse remodeling both at the myocardial and valvular levels. Persistent activation of valvular endothelial cells leads to leaflet fibrosis through endothelial-to-mesenchymal transition (EMT). Tenascin C (TNC), an extracellular matrix glycoprotein involved in cardiovascular remodeling and fibrosis, was also identified in inducing epithelial-to-mesenchymal transition. In this study, we hypothesized that TNC also plays a role in the valvular remodeling observed in ischemic MR by contributing to valvular excess EMT. Moderate ischemic MR was induced by creating a posterior papillary muscle infarct (7 pigs and 7 sheep). Additional animals (7 pigs and 4 sheep) served as controls. Pigs and sheep were sacrificed after 6 weeks and 6 months, respectively. TNC expression was upregulated in the pig and sheep experiments at 6 weeks and 6 months, respectively, and correlated well with leaflet thickness (R = 0.68; p < 0.001 at 6 weeks, R = 0.84; p < 0.001 at 6 months). To confirm the translational potential of our findings, we obtained mitral valves from patients with ischemic cardiomyopathy presenting MR (n = 5). Indeed, TNC was also expressed in the mitral leaflets of these. Furthermore, TNC induced EMT in isolated porcine mitral valve endothelial cells (MVEC). Interestingly, Toll-like receptor 4 (TLR4) inhibition prevented TNC-mediated EMT in MVEC. We identified here for the first time a new contributor to valvular remodeling in ischemic MR, namely TNC, which induced EMT through TLR4. Our findings might set the path for novel therapeutic targets for preventing or limiting ischemic MR.


Subject(s)
Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mitral Valve Insufficiency/metabolism , Mitral Valve/metabolism , Myocardial Infarction/complications , Tenascin/metabolism , Aged , Aged, 80 and over , Animals , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Female , Humans , Male , Middle Aged , Mitral Valve/pathology , Mitral Valve/physiopathology , Mitral Valve Insufficiency/etiology , Mitral Valve Insufficiency/pathology , Mitral Valve Insufficiency/physiopathology , Sheep, Domestic , Signal Transduction , Sus scrofa , Toll-Like Receptor 4/metabolism , Up-Regulation
3.
J Hypertens ; 37(9): 1861-1870, 2019 09.
Article in English | MEDLINE | ID: mdl-30950975

ABSTRACT

BACKGROUND: Tenascin C (TN-C) is considered to play a pathophysiological role in maladaptive left ventricular remodeling. Yet, the mechanism underlying TN-C-dependent cardiac dysfunction remains elusive. METHOD: The present study was designed to investigate the effect of hypoxia and hypertrophic stimuli on TN-C expression in H9c2 cells and its putative regulation by epigenetic mechanisms, namely DNA promoter methylation and microRNAs. In addition, rats subjected to myocardial infarction (MI) were investigated. H9c2 cells were subjected to oxygen and glucose deprivation; incubated with angiotensin II (Ang II); or human TN-C (hTN-C) purified protein. Hypertrophic and fibrotic markers, TN-C promoter methylation as well as mir-335 expression were assessed by reverse transcription and quantitative polymerase chain reaction while TN-C protein levels were assessed by ELISA. RESULTS: Tn-C mRNA expression was markedly increased by both oxygen and glucose deprivation and Ang II (P < 0.01, respectively). In addition, Ang-II-dependent TN-C upregulation was explained by reduced promoter methylation (P < 0.05). Cells treated with hTN-C displayed upregulation of Bnp, Mmp2, ß-Mhc, integrin α6 and integrin ß1. Furthermore, hTN-C treated cells showed a significant reduction in adenosine monophosphate and adenosine triphosphate levels. In vivo, plasma and myocardial TN-C levels were increased 7 days post MI (P < 0.05, respectively). This increment in TN-C was accompanied by upregulation of mir-335 (P < 0.01). In conclusion, both hypoxic and hypertrophic stimuli lead to epigenetically driven TN-C upregulation and subsequent impairment of cellular energy metabolism in cardiomyoblasts. CONCLUSION: These findings might enlighten our understanding on maladaptive left ventricular remodeling and direct towards a strong involvement of TN-C.


Subject(s)
Cardiomegaly/metabolism , DNA Methylation , Hypoxia/metabolism , Myocardial Infarction/metabolism , Tenascin/metabolism , Angiotensin II , Animals , Coronary Artery Disease , Energy Metabolism , Epigenesis, Genetic , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Fibrosis , Heart Diseases/metabolism , Humans , Hypertrophy , Male , Matrix Metalloproteinase 2/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Nerve Tissue Proteins , Rats , Tenascin/genetics , Ventricular Remodeling
4.
Int J Cardiol ; 285: 72-79, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30904281

ABSTRACT

AIMS: Remote ischemic conditioning (RIC) is considered a potential clinical approach to reduce myocardial infarct size and ameliorate adverse post-infarct left ventricular (LV) remodeling, however the mechanisms are unknown. The aim was to clarify the impact of RIC on Neuregulin-1 (NRG-1)/ErbBs expression, inflammation and LV hemodynamic function. METHODS AND RESULTS: Male Sprague-Dawley rats were subjected to 30 min occlusion of the left coronary artery (LCA) followed by 2 weeks of reperfusion and separated into three groups: (1) sham operated (without LCA occlusion); (2) Myocardial ischemia/reperfusion (MIR) and (3) remote ischemic perconditioning group (MIR + RIPerc). Cardiac structural and functional changes were evaluated by echocardiography and on the isolated working heart system. The level of H3K4me3 at the NRG-1 promoter, and both plasma and LV tissue levels of NRG-1 were assessed. The expression of pro-inflammatory cytokines, ECM components and ErbB receptors were assessed by RT-qPCR. MIR resulted in a significant decrease in LV function and enlargement of LV chamber. This was accompanied with a decrease in the level of H3K4me3 at the NRG-1 promoter. Consequently NRG-1 protein levels were reduced in the infarcted myocardium. Subsequently, an upregulated influx of CD68+ macrophages, high expression of MMP-2 and -9 as well as an increase of IL-1ß, TLR-4, TNF-α, TNC expression were observed. In contrast, RIPerc significantly decreased inflammation and improved LV function in association with the enhancement of NRG-1 levels and ErbB3 expression. CONCLUSIONS: These findings may reveal a novel anti-remodeling and anti-inflammatory effect of RIPerc, involving activation of NRG-1/ErbB3 signaling.


Subject(s)
Heart Ventricles/physiopathology , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/complications , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology , Animals , Disease Models, Animal , Echocardiography , Heart Ventricles/diagnostic imaging , Male , Myocardial Infarction/etiology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Sprague-Dawley
5.
J Hypertens ; 36(4): 847-856, 2018 04.
Article in English | MEDLINE | ID: mdl-29283973

ABSTRACT

AIMS: Left ventricular (LV) hypertrophy is characterized by cardiomyocyte hypertrophy and interstitial fibrosis ultimately leading to increased myocardial stiffness and reduced contractility. There is substantial evidence that the altered expression of matrix metalloproteinases (MMP) and Tenascin-C (TN-C) are associated with the progression of adverse LV remodeling. However, the role of TN-C in the development of LV hypertrophy because of chronic pressure overload as well as the regulatory role of TN-C on MMPs remains unknown. METHODS AND RESULTS: In a knockout mouse model of TN-C, we investigated the effect of 10 weeks of pressure overload using transverse aortic constriction (TAC). Cardiac function was determined by magnetic resonance imaging. The expression of MMP-2 and MMP-9, CD147 as well as myocardial fibrosis were assessed by immunohistochemistry. The expression of TN-C was assessed by RT-qPCR and ELISA. TN-C knockout mice showed marked reduction in fibrosis (P < 0.001) and individual cardiomyocytes size (P < 0.01), in expression of MMP-2 (P < 0.05) and MMP-9 (P < 0.001) as well as preserved cardiac function (P < 0.01) in comparison with wild-type mice after 10 weeks of TAC. In addition, CD147 expression was markedly increased under pressure overload (P < 0.01), irrespectively of genotype. TN-C significantly increased the expression of the markers of hypertrophy such as ANP and BNP as well as MMP-2 in H9c2 cells (P < 0.05, respectively). CONCLUSION: Our results are pointed toward a novel signaling mechanism that contributes to LV remodeling via MMPs upregulation, cardiomyocyte hypertrophy as well as myocardial fibrosis by TN-C under chronic pressure overload.


Subject(s)
Hypertension/complications , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Myocardium/pathology , Tenascin/genetics , Tenascin/metabolism , Ventricular Remodeling/genetics , Animals , Basigin/genetics , Basigin/metabolism , Cardiac Output , Cell Line , Fibrosis , Genotype , Hypertrophy, Left Ventricular/physiopathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Signal Transduction , Ventricular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...