Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Basic Res Cardiol ; 119(2): 215-242, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38472506

ABSTRACT

Atrial myopathy is a condition that consists of electrical, structural, contractile, and autonomic remodeling of the atria and is the substrate for development of atrial fibrillation, the most common arrhythmia. Pathophysiologic mechanisms driving atrial myopathy are inflammation, oxidative stress, atrial stretch, and neurohormonal signals, e.g., angiotensin-II and aldosterone. These mechanisms initiate the structural and functional remodeling of the atrial myocardium. Novel therapeutic strategies are being developed that target the pathophysiologic mechanisms of atrial myopathy. In this review, we will discuss the pathophysiology of atrial myopathy, as well as diagnostic and therapeutic strategies.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Muscular Diseases , Humans , Clinical Relevance , Heart Atria , Myocardium , Atrial Remodeling/physiology
2.
Am J Physiol Heart Circ Physiol ; 325(2): H398-H413, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37389953

ABSTRACT

Dysfunction or failure of the endothelial organ is a heterogenous and often ill-described feature of both cardiovascular and noncardiovascular disorders. Although seldom recognized as a separate clinical condition, endothelial cell dysfunction (ECD) is an established catalyst of disease. However, even in recent pathophysiological studies, ECD is frequently oversimplified as a binary state without gradation, based on the assessment of a single function (e.g., synthesis or activity of nitric oxide) and without considering spatiotemporal dimensions (local vs. generalized, acute vs. chronic). In this article, we suggest a simple scale to grade the severity of ECD and a definition of ECD in three dimensions: space, time, and severity. We also adopt a broader perspective on ECD by integrating and comparing gene expression data of endothelial cells from different organs and diseases and propose a concept that links common pathophysiological mechanisms. We hope that this will enhance the understanding of the pathophysiology of ECD and stimulate discussion in this field.


Subject(s)
Endothelial Cells , Vascular Diseases , Humans , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism
3.
J Thromb Haemost ; 21(5): 1266-1273, 2023 05.
Article in English | MEDLINE | ID: mdl-36740042

ABSTRACT

BACKGROUND: Statins efficiently lower cholesterol and also exert pleiotropic effects that extend beyond lipid lowering. In a recent pilot study, valuable information on the carboxypeptidase U (CPU) system in hyperlipidemia and the effect of statin therapy was collected. It was shown that proCPU levels are increased in hyperlipidemic patients. Statins significantly decreased proCPU levels and improved plasma fibrinolysis. Furthermore, it was suggested that patients with high baseline proCPU levels are most likely to benefit from statin therapy. OBJECTIVES: We aimed to further substantiate the effect of hyperlipidemia and statin therapy on CPU-related parameters in a larger cohort of hyperlipidemic and statin-treated individuals. METHODS: Blood was collected from 141 individuals treated with different dosages of atorvastatin (10-80 mg), 38 normolipidemic, and 37 hyperlipidemic controls. Lipid parameters and markers of fibrinolysis (proCPU and clot lysis time) were determined and compared between the groups. RESULTS: Pilot study results of high proCPU concentrations in hyperlipidemic patients and the proCPU-reducing effect of atorvastatin were confirmed. Accordingly, an improvement in plasma fibrinolytic potential was seen under the influence of atorvastatin. High interindividual variation in proCPU concentrations was observed in the hyperlipidemic cohort, with up to 80% higher proCPU levels compared with normolipidemic controls. Furthermore, proCPU concentration and the dosage of atorvastatin were inversely correlated. CONCLUSIONS: This study clearly shows that plasma proCPU concentrations and its expected effect on the fibrinolytic rate (as measured by clot lysis time) are increased in hyperlipidemic patients and that these effects can be normalized (and even further reduced compared with normolipidemic patients) by atorvastatin treatment.


Subject(s)
Carboxypeptidase B2 , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pilot Projects , Thrombolytic Therapy
4.
Am J Physiol Heart Circ Physiol ; 323(4): H763-H773, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36018757

ABSTRACT

Cardiac arrhythmias are associated with cardiovascular morbidity and mortality. Cardiac electrophysiology studies (EPS) use intracardiac catheter recording and stimulation for profound evaluation of the heart's electrical properties. The main clinical application is investigation and treatment of rhythm disorders. These techniques have been translated to the murine setting to open opportunities for detailed evaluation of the impact of different characteristics (including genetics) and interventions on cardiac electrophysiology and -pathology. Currently, a detailed description of the technique of murine transjugular EPS (which is the standard route of catheter introduction) is lacking. This article provides detailed information on EPS in mice via the transjugular route. This includes catheter placement, stimulation protocols, intracardiac tracing interpretation, artifact reduction, and surface ECG recording. In addition, reference values as obtained in C57BL/6N mice are presented for common electrophysiological parameters. This detailed methodological description aims to increase accessibility and standardization of EPS in mice. Ultimately, also human research and patient care may benefit from translation of the knowledge obtained in preclinical models using this technique.NEW & NOTEWORTHY Electrophysiology studies (EPS) allow in-depth evaluation of cardiac electrophysiology and -pathology. These techniques have been adapted to the murine setting for (translational) studies, mainly focusing on arrhythmogenesis. Despite the frequent application of EPS via the transjugular route, a thorough description of the technique is currently lacking. This article aims to function as a comprehensive guide, also elaborating (for the first time) on nonsurgical aspects such as catheter positioning, tracing artifacts, stimulation protocols, and reference values.


Subject(s)
Arrhythmias, Cardiac , Electrophysiologic Techniques, Cardiac , Animals , Electrocardiography , Electrophysiologic Techniques, Cardiac/methods , Heart , Humans , Mice , Mice, Inbred C57BL
5.
J Vis Exp ; (175)2021 09 24.
Article in English | MEDLINE | ID: mdl-34633365

ABSTRACT

Atrial fibrillation (AF) is the most common arrhythmia caused by structural remodeling of the atria, also called atrial myopathy. Current therapies only target the electrical abnormalities and not the underlying atrial myopathy. For the development of novel therapies, a reproducible large animal model of atrial myopathy is necessary. This paper presents a model of sterile pericarditis-induced atrial myopathy in Aachener minipigs. Sterile pericarditis was induced by spraying sterile talcum and leaving a layer of sterile gauze over the atrial epicardial surface. This led to inflammation and fibrosis, two crucial components of the pathophysiology of atrial myopathy, making the atria susceptible to the induction of AF. Two pacemaker electrodes were positioned epicardially on each atrium and connected to two pacemakers from different manufacturers. This strategy allowed for repeated non-invasive atrial programmed stimulation to determine the inducibility of AF at specified time points after surgery. Different protocols to test AF inducibility were used. The advantages of this model are its clinical relevance, with AF inducibility and the rapid induction of inflammation and fibrosis-both present in atrial myopathy-and its reproducibility. The model will be useful in the development of novel therapies targeting atrial myopathy and AF.


Subject(s)
Atrial Fibrillation , Muscular Diseases , Pericarditis , Animals , Atrial Fibrillation/etiology , Pericarditis/etiology , Reproducibility of Results , Swine , Swine, Miniature
6.
Circ Heart Fail ; 14(7): e006898, 2021 07.
Article in English | MEDLINE | ID: mdl-34247489

ABSTRACT

BACKGROUND: Peripartum cardiomyopathy (PPCM) is a life-threatening disease in women without previously known cardiovascular disease. It is characterized by a sudden onset of heart failure before or after delivery. Previous studies revealed that the generation of a 16-kDa PRL (prolactin) metabolite, the subsequent upregulation of miR-146a, and the downregulation of the target gene Erbb4 is a common driving factor of PPCM. METHODS: miRNA profiling was performed in plasma of PPCM patients (n=33) and postpartum-matched healthy CTRLs (controls; n=36). Elevated miRNAs in PPCM plasma, potentially targeting ERBB4 (erythroblastic leukemia viral oncogene homolog 4), were overexpressed in cardiomyocytes using lentiviral vectors. Next, cardiac function, cardiac morphology, and PPCM phenotype were investigated after recurrent pregnancies of HZ (heterozygous) cardiomyocyte-specific Erbb4 mice (Erbb4F/+ αMHC-Cre+, n=9) with their age-matched nonpregnant CTRLs (n=9-10). RESULTS: Here, we identify 9 additional highly conserved miRNAs (miR-199a-5p and miR-199a-3p, miR-145a-5p, miR-130a-3p, miR-135a-5p, miR-221-3p, miR-222-3p, miR-23a-3p, and miR19b-3p) that target tyrosine kinase receptor ERBB4 and are over 4-fold upregulated in plasma of PPCM patients at the time of diagnosis. We confirmed that miR-146a, miR-199a-5p, miR-221-3p, miR-222-3p, miR-23a-3p, miR-130a-5p, and miR-135-3p overexpression decreases ERBB4 expression in cardiomyocytes (-29% to -50%; P<0.05). In addition, we demonstrate that genetic cardiomyocyte-specific downregulation of Erbb4 during pregnancy suffices to induce a variant of PPCM in mice, characterized by left ventricular dilatation (postpartum second delivery: left ventricular internal diameter in diastole, +19±7% versus HZ-CTRL; P<0.05), increased atrial natriuretic peptide (ANP) levels (4-fold increase versus HZ-CTRL mice, P<0.001), decreased VEGF (vascular endothelial growth factor) and VE-cadherin levels (-33±17%, P=0.07; -27±20%, P<0.05 versus HZ-CTRL), and histologically enlarged cardiomyocytes (+20±21%, versus HZ-CTRL, P<0.05) but without signs of myocardial apoptosis and inflammation. CONCLUSIONS: ERBB4 is essential to protect the maternal heart from peripartum stress. Downregulation of ERBB4 in cardiomyocytes induced by multiple miRNAs in the peripartum period may be crucial in PPCM pathophysiology. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT00998556.


Subject(s)
Cardiomyopathies/physiopathology , Heart Failure/genetics , MicroRNAs/genetics , Receptor, ErbB-4/genetics , Animals , Cardiomyopathies/genetics , Cardiovascular Diseases/genetics , Female , Heart Failure/metabolism , Humans , Mice , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Peripartum Period/metabolism , Pregnancy , Receptor, ErbB-4/metabolism
7.
Am J Physiol Heart Circ Physiol ; 320(6): H2416-H2428, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33989083

ABSTRACT

Endothelial cells (ECs) secrete different paracrine signals that modulate the function of adjacent cells; two examples of these paracrine signals are nitric oxide (NO) and neuregulin-1 (NRG1), a cardioprotective growth factor. Currently, it is undetermined whether one paracrine factor can compensate for the loss of another. Herein, we hypothesized that NRG1 can compensate for endothelial NO synthase (eNOS) deficiency. We characterized eNOS null and wild-type (WT) mice by cardiac ultrasound and histology and we determined circulating NRG1 levels. In a separate experiment, eight groups of mice were divided into four groups of eNOS null mice and WT mice; half of the mice received angiotensin II (ANG II) to induce a more severe phenotype. Mice were randomized to daily injections with NRG1 or vehicle for 28 days. eNOS deficiency increased NRG1 plasma levels, indicating that ECs increase their NRG1 expression when NO production is deleted. eNOS deficiency also increased blood pressure, lowered heart rate, induced cardiac fibrosis, and affected diastolic function. In eNOS null mice, ANG II administration not only increased cardiac fibrosis but also induced cardiac hypertrophy and renal fibrosis. NRG1 administration prevented cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. Moreover, Nrg1 expression in the myocardium is shown to be regulated by miR-134. This study indicates that administration of endothelium-derived NRG1 can compensate for eNOS deficiency in the heart and kidneys.NEW & NOTEWORTHY ECs compensate for eNOS deficiency by increasing the secretion of NRG1. NRG1 administration prevents cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. NRG1 expression is regulated by miR-134.


Subject(s)
Endothelial Cells/metabolism , Heart Rate/genetics , Heart/drug effects , MicroRNAs/metabolism , Myocardium/pathology , Neuregulin-1/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Diastole/drug effects , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation , Heart Rate/drug effects , Kidney/pathology , Mice , Mice, Knockout , Neuregulin-1/pharmacology , Nitric Oxide Synthase Type III/metabolism , Random Allocation , Vasoconstrictor Agents/pharmacology
8.
J Am Heart Assoc ; 10(3): e019169, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33470124

ABSTRACT

The myocardium consists of different cell types, of which endothelial cells, cardiomyocytes, and fibroblasts are the most abundant. Communication between these different cell types, also called paracrine signaling, is essential for normal cardiac function, but also important in cardiac remodeling and heart failure. Systematic studies on the expression of ligands and their corresponding receptors in different cell types showed that for 60% of the expressed ligands in a particular cell, the receptor is also expressed. The fact that many ligand-receptor pairs are present in most cells, including the major cell types in the heart, indicates that autocrine signaling is a widespread phenomenon. Autocrine signaling in cardiac remodeling and heart failure is involved in all pathophysiological mechanisms generally observed: hypertrophy, fibrosis, angiogenesis, cell survival, and inflammation. Herein, we review ligand-receptor pairs present in the major cardiac cell types based on RNA-sequencing expression databases, and we review current literature on extracellular signaling proteins with an autocrine function in the heart; these include C-type natriuretic peptide, fibroblast growth factors 2, F21, and 23, macrophage migration inhibitory factor, heparin binding-epidermal growth factor, angiopoietin-like protein 2, leptin, adiponectin, follistatin-like 1, apelin, neuregulin 1, vascular endothelial growth factor, transforming growth factor ß, wingless-type integration site family, member 1-induced secreted protein-1, interleukin 11, connective tissue growth factor/cellular communication network factor, and calcitonin gene‒related peptide. The large number of autocrine signaling factors that have been studied in the literature supports the concept that autocrine signaling is an essential part of myocardial biology and disease.


Subject(s)
Autocrine Communication/physiology , Heart Failure/therapy , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Animals , Cells, Cultured , Heart Failure/metabolism , Heart Failure/pathology , Humans , Myocytes, Cardiac/pathology , Signal Transduction
9.
Am J Physiol Heart Circ Physiol ; 319(2): H443-H455, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32618511

ABSTRACT

Neuregulin-1 (NRG1) is a paracrine growth factor, secreted by cardiac endothelial cells (ECs) in conditions of cardiac overload/injury. The current concept is that the cardiac effects of NRG1 are mediated by activation of erythroblastic leukemia viral oncogene homolog (ERBB)4/ERBB2 receptors on cardiomyocytes. However, recent studies have shown that paracrine effects of NRG1 on fibroblasts and macrophages are equally important. Here, we hypothesize that NRG1 autocrine signaling plays a role in cardiac remodeling. We generated EC-specific Erbb4 knockout mice to eliminate endothelial autocrine ERBB4 signaling without affecting paracrine NRG1/ERBB4 signaling in the heart. We first observed no basal cardiac phenotype in these mice up to 32 wk. We next studied these mice following transverse aortic constriction (TAC), exposure to angiotensin II (ANG II), or myocardial infarction in terms of cardiac performance, myocardial hypertrophy, myocardial fibrosis, and capillary density. In general, no major differences between EC-specific Erbb4 knockout mice and control littermates were observed. However, 8 wk following TAC both myocardial hypertrophy and fibrosis were attenuated by EC-specific Erbb4 deletion, albeit these responses were normalized after 20 wk. Similarly, 4 wk after ANG II treatment, myocardial fibrosis was less pronounced compared with control littermates. These observations were supported by RNA-sequencing experiments on cultured endothelial cells showing that NRG1 controls the expression of various hypertrophic and fibrotic pathways. Overall, this study shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling. This study contributes to understanding the spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury.NEW & NOTEWORTHY The role of NRG1/ERBB signaling in endothelial cells is not completely understood. Our study contributes to the understanding of spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury and shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling.


Subject(s)
Autocrine Communication , Cardiomyopathies/metabolism , Endothelial Cells/metabolism , Hypertrophy, Left Ventricular/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-4/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Paracrine Communication , Receptor, ErbB-4/deficiency , Receptor, ErbB-4/genetics , Signal Transduction
10.
Am J Physiol Heart Circ Physiol ; 318(5): H1233-H1244, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32275471

ABSTRACT

An important physiological role of the aorta is to convert the pulsatile blood flow that originates in the heart to a nearly continuous flow in the peripheral vessels. Previously, we demonstrated that basal, unstimulated nitric oxide (NO) production is more abundant in large as compared with muscular arteries and that it is an important regulator of arterial (aortic) stiffness. Hence, endothelial function and NO bioavailability are important determinants of aortic biomechanics, and mouse models with altered NO signaling might be of interest to investigate the (patho)physiological role of the NO signaling as a dynamic regulator of arterial stiffness. We aimed to characterize the ex vivo biomechanical properties of aortic segments from mice with no (eNOS-/-), normal [wild type (WT)], or high (eNOS-tg) endothelial NO synthase (eNOS) expression. Isobaric aortic diameter and compliance were lower in eNOS-/- mice and increased in eNOS-tg mice as compared with WT mice. Interestingly, these differences remained when NO levels were pharmacologically restored ex vivo, suggesting that they were not merely the result of a lack or excess of the vasodilator effects of NO. Analysis of basal vascular smooth muscle cell tone and the phasic as well as the tonic contraction in response to α1-adrenergic stimulation with phenylephrine revealed that the chronic lack of eNOS expression affected aortic reactivity similarly but with different magnitude as compared with acute eNOS blockade using Nω-nitro-l-arginine methyl ester in WT and eNOS-tg mice, suggesting that chronical distortion of NO signaling triggered several compensatory mechanisms that reflect the organism's attempt to restore the contractile imbalance and maintain optimal central hemodynamics.NEW & NOTEWORTHY Endothelial function and NO bioavailability are important determinants of aortic biomechanics and function. With a new technique we investigated the ex vivo aortic segment biomechanics of different mouse models with altered NO signaling. Our experiments clearly show that chronic distortion of NO signaling triggered several compensatory mechanisms that reflect the organism's attempt to maintain optimal central hemodynamics.


Subject(s)
Aorta/physiology , Nitric Oxide Synthase Type III/metabolism , Vascular Stiffness , Animals , Aorta/metabolism , Biomechanical Phenomena , Male , Mice , Mice, Inbred C57BL , Muscle Tonus , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Signal Transduction , Vasoconstriction
11.
Cell Oncol (Dordr) ; 43(3): 335-352, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32219702

ABSTRACT

BACKGROUND: The epidermal growth factor receptor family consists of four members, ErbB1 (epidermal growth factor receptor-1), ErbB2, ErbB3, and ErbB4, which all have been found to play important roles in tumor development. ErbB4 appears to be unique among these receptors, because it is the only member with growth inhibiting properties. ErbB4 plays well-defined roles in normal tissue development, in particular the heart, the nervous system, and the mammary gland system. In recent years, information on the role of ErbB4 in a number of tumors has emerged and its general direction points towards a tumor suppressor role for ErbB4. However, there are some controversies and conflicting data, warranting a review on this topic. CONCLUSIONS: Here, we discuss the role of ErbB4 in normal physiology and in breast, lung, colorectal, gastric, pancreatic, prostate, bladder, and brain cancers, as well as in hepatocellular carcinoma, cholangiocarcinoma, and melanoma. Understanding the role of ErbB4 in cancer is not only important for the treatment of tumors, but also for the treatment of other disorders in which ErbB4 plays a major role, e.g. cardiovascular disease.


Subject(s)
Neoplasms/metabolism , Receptor, ErbB-4/metabolism , Embryonic Development , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/pathology , Receptor, ErbB-4/genetics , Signal Transduction
12.
J Mol Cell Cardiol ; 138: 75-87, 2020 01.
Article in English | MEDLINE | ID: mdl-31756323

ABSTRACT

The myocardium is a highly structured pluricellular tissue which is governed by an intricate network of intercellular communication. Endothelial cells are the most abundant cell type in the myocardium and exert crucial roles in both healthy myocardium and during myocardial disease. In the last decade, microRNAs have emerged as new actors in the regulation of cellular function in almost every cell type. Here, we review recent evidence on the regulatory function of different microRNAs expressed in endothelial cells, also called endothelial microRNAs, in healthy and diseased myocardium. Endothelial microRNA emerged as modulators of angiogenesis in the myocardium, they are implicated in the paracrine role of endothelial cells in regulating cardiac contractility and homeostasis, and interfere in the crosstalk between endothelial cells and cardiomyocytes.


Subject(s)
Cardiovascular Diseases/genetics , Endothelial Cells/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Animals , Humans , MicroRNAs/genetics , Myocardium/pathology , Neovascularization, Physiologic/genetics , Paracrine Communication/genetics
13.
Circ Heart Fail ; 12(10): e006288, 2019 10.
Article in English | MEDLINE | ID: mdl-31607147

ABSTRACT

Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1-a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.


Subject(s)
Endothelial Cells/metabolism , Heart Failure/metabolism , Neuregulin-1/metabolism , Animals , Cardiovascular Agents/therapeutic use , Chronic Disease , Endothelial Cells/drug effects , ErbB Receptors/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Ligands , Molecular Targeted Therapy , Neuregulin-1/therapeutic use , Signal Transduction
14.
Physiol Genomics ; 51(6): 186-196, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30978160

ABSTRACT

Cardiac microvascular endothelial cells (CMVECs) are the most numerous cells in the myocardium and orchestrate cardiogenesis during development, regulate adult cardiac function, and modulate pathophysiology of heart failure. It has been shown that the transcriptome of CMVECs differs from other endothelial cell types, but transcriptomic changes in cardiac endothelial cells during cardiac maturation and cardiac remodeling have not been studied. CMVECs were isolated from rat hearts based on CD31 expression and were immediately processed for RNA sequencing. We compared gene expression levels from primary CMVECs of neonatal hearts, normal adult hearts, and infarcted hearts. Between neonatal and adult CMVECs, 6,838 genes were differentially expressed, indicating that CMVECs undergo a substantial transformation during postnatal cardiac growth. A large fraction of genes upregulated in neonatal CMVECs are part of mitosis pathways, whereas a large fraction of genes upregulated in adult CMVECs are part of cellular response, secretory, signaling, and cell adhesion pathways. Between CMVECs of normal adult hearts and infarcted hearts, 159 genes were differentially expressed. We found a limited degree of overlap (55 genes) between the differentially expressed genes in neonatal and infarcted-hearts. Of 46 significantly upregulated genes in the infarcted heart, 46% were also upregulated in neonatal hearts relative to sham. Of 113 significantly downregulated genes in the infarcted-hearts, 30% were also downregulated in neonatal hearts relative to sham. These data demonstrate that CMVECs undergo dramatic changes from neonatal to adult and more subtle changes between normal state and cardiac remodeling.


Subject(s)
Endothelial Cells/metabolism , Heart/physiology , Transcriptome/genetics , Ventricular Remodeling/genetics , Animals , Cell Adhesion/genetics , Cells, Cultured , Down-Regulation/genetics , Female , Gene Expression Profiling/methods , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA/methods , Signal Transduction/genetics , Up-Regulation/genetics
15.
Eur Heart J ; 40(26): 2155-2163, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30957868

ABSTRACT

Randomized clinical trials initially used heart failure (HF) patients with low left ventricular ejection fraction (LVEF) to select study populations with high risk to enhance statistical power. However, this use of LVEF in clinical trials has led to oversimplification of the scientific view of a complex syndrome. Descriptive terms such as 'HFrEF' (HF with reduced LVEF), 'HFpEF' (HF with preserved LVEF), and more recently 'HFmrEF' (HF with mid-range LVEF), assigned on arbitrary LVEF cut-off points, have gradually arisen as separate diseases, implying distinct pathophysiologies. In this article, based on pathophysiological reasoning, we challenge the paradigm of classifying HF according to LVEF. Instead, we propose that HF is a heterogeneous syndrome in which disease progression is associated with a dynamic evolution of functional and structural changes leading to unique disease trajectories creating a spectrum of phenotypes with overlapping and distinct characteristics. Moreover, we argue that by recognizing the spectral nature of the disease a novel stratification will arise from new technologies and scientific insights that will shape the design of future trials based on deeper understanding beyond the LVEF construct alone.


Subject(s)
Heart Failure/classification , Stroke Volume , Comorbidity , Disease Progression , Endothelium, Vascular/physiopathology , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocytes, Cardiac/physiology , Reference Values , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling
16.
Am J Physiol Heart Circ Physiol ; 316(6): H1417-H1425, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30951364

ABSTRACT

The myocardium is a highly structured tissue consisting of different cell types including cardiomyocytes, endothelial cells, fibroblasts, smooth muscle cells, inflammatory cells, and stem cells. Microvascular endothelial cells are the most abundant cell type in the myocardium and play crucial roles during cardiac development, in normal adult myocardium, and during myocardial diseases such as heart failure. In the last decade, epigenetic changes have been described regulating cellular function in almost every cell type in the organism. Here, we review recent evidence on different epigenetic changes that regulate intercellular communication in normal myocardium and during myocardial diseases, including cardiac remodeling. Epigenetic changes influence many intercellular communication signaling systems, including the nitric oxide, angiotensin, and endothelin signaling systems. In this review, we go beyond discussing classic endothelial function (for instance nitric oxide secretion) and will discuss epigenetic regulation of intercellular communication.


Subject(s)
Cell Communication/genetics , DNA Methylation , Epigenesis, Genetic , Heart Diseases/genetics , Myocardium/metabolism , Ventricular Remodeling/genetics , Acetylation , Angiotensins/genetics , Angiotensins/metabolism , Animals , Chromatin Assembly and Disassembly , Endothelins/genetics , Endothelins/metabolism , Fibrosis , Gene Expression Regulation , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Histones/genetics , Histones/metabolism , Humans , Myocardium/pathology , Nitric Oxide/metabolism , Signal Transduction
17.
Physiol Rep ; 7(4): e13934, 2019 02.
Article in English | MEDLINE | ID: mdl-30810292

ABSTRACT

Over the past few decades, isometric contraction studies of isolated thoracic aorta segments have significantly contributed to our overall understanding of the active, contractile properties of aortic vascular smooth muscle cells (VSMCs) and their cross-talk with endothelial cells. However, the physiological role of VSMC contraction or relaxation in the healthy aorta and its contribution to the pulse-smoothening capacity of the aorta is currently unclear. Therefore, we investigated the acute effects of VSMC contraction and relaxation on the isobaric biomechanical properties of healthy mouse aorta. An in-house developed set-up was used to measure isobaric stiffness parameters of periodically stretched (10 Hz) aortic segments at an extended pressure range, while pharmacologically modulating VSMC tone and endothelial cell function. We found that the effects of α1-adrenergic stimulation with phenylephrine on the pressure-stiffness relationship varied in sensitivity, magnitude and direction, with the basal, unstimulated NO production by the endothelium playing a pivotal role. We also investigated how arterial disease affected this system by using the angiotensin-II-treated mouse. Our results show that isobaric stiffness was increased and that the aortic segments demonstrated a reduced capacity for modulating the pressure-stiffness relationship. This suggests that not only increased isobaric stiffness at normal pressure, but also a reduced capacity of the VSMCs to limit the pressure-associated increase in aortic stiffness, may contribute to the pathogenesis of this mouse model. Overall, this study provides more insight in how aortic VSMC tone affects the pressure-dependency of aortic biomechanics at different physiological and pathological conditions.


Subject(s)
Aorta/physiology , Muscle Relaxation , Muscle, Smooth, Vascular/physiology , Vascular Stiffness , Vasoconstriction , Angiotensin II/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle Tonus , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/physiology
18.
Clin Exp Pharmacol Physiol ; 46(3): 204-215, 2019 03.
Article in English | MEDLINE | ID: mdl-30244497

ABSTRACT

Anthracycline chemotherapy has a prominent role in treating many forms of cancer. Unfortunately, cardiotoxic side effects represent a serious limitation to their use, with doxorubicin being the leading drug of the group. Indeed, anthracycline-induced cardiomyopathy is an important public health concern because it may not be detected for many years and remains a lifelong threat. Even after decades of investigation, neither the exact mode of action of anthracyclines nor the pathways leading to their side effect are fully understood. It is increasingly important to establish collaboration between oncologists and cardiologists to improve the management of cancer patient receiving anthracyclines. This article reviews the clinical course, pathogenesis, cardiac monitoring and new concepts in diagnosing and preventing anthracycline-induced cardiotoxicity.


Subject(s)
Anthracyclines/adverse effects , Cardiotoxicity/prevention & control , Cardiotoxicity/physiopathology , Animals , Antineoplastic Agents/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Humans
19.
Clin Exp Pharmacol Physiol ; 46(3): 255-265, 2019 03.
Article in English | MEDLINE | ID: mdl-30339273

ABSTRACT

We have previously shown that treatment with recombinant human neuregulin-1 (rhNRG-1) improves pulmonary arterial hypertension (PAH) in a monocrotaline (MCT)-induced animal model, by decreasing pulmonary arterial remodelling and endothelial dysfunction, as well as by restoring right ventricular (RV) function. Additionally, rhNRG-1 treatment showed direct myocardial anti-remodelling effects in a model of pressure loading of the RV without PAH. This work aimed to study the intrinsic cardiac effects of rhNRG-1 on experimental PAH and RV pressure overload, and more specifically on diastolic stiffness, at both the ventricular and cardiomyocyte level. We studied the effects of chronic rhNRG-1 treatment on ventricular passive stiffness in RV and LV samples from MCT-induced PAH animals and in the RV from animals with compensated and decompensated RV hypertrophy, through a mild and severe pulmonary artery banding (PAB). We also measured passive tension in isolated cardiomyocytes and quantified the expression of myocardial remodelling-associated genes and calcium handling proteins. Chronic rhNRG-1 treatment decreased passive tension development in RV and LV isolated from animals with MCT-induced PAH. This decrease was associated with increased phospholamban phosphorylation, and with attenuation of the expression of cardiac maladaptive remodelling markers. Finally, we showed that rhNRG-1 therapy decreased RV remodelling and cardiomyocyte passive tension development in PAB-induced RV hypertrophy animals, without compromising cardiac function, pointing to cardiac-specific effects in both hypertrophy stages. In conclusion, we demonstrated that rhNRG-1 treatment decreased RV intrinsic diastolic stiffness, through the improvement of calcium handling and cardiac remodelling signalling.


Subject(s)
Diastole/physiology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Neuregulin-1/pharmacology , Vascular Stiffness/drug effects , Ventricular Dysfunction, Right/drug therapy , Animals , Calcium Signaling/drug effects , Gene Expression Regulation/drug effects , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuregulin-1/therapeutic use , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Ventricular Remodeling/drug effects
20.
Front Physiol ; 9: 582, 2018.
Article in English | MEDLINE | ID: mdl-29867592

ABSTRACT

Induction of hypertension by angiotensin II (AngII) is a widely used experimental stimulus to study vascular aging in mice. It is associated with large artery stiffness, a hallmark of arterial aging and a root cause of increased cardiovascular risk. We reported earlier that long term (4 week) AngII treatment in mice altered the active, contractile properties of the arteries in a vascular bed-specific manner and that, in healthy mice aorta, active contractile properties of the aortic wall determine isobaric aortic stiffness. Given the huge physiological relevance of large artery stiffening, we aimed to characterize the early (1 week) changes in the active properties of the aorta of AngII-treated mice. We were not able to detect a significant effect of AngII treatment on anesthetized blood pressure or abdominal aorta pulse wave velocity. Ex vivo biomechanical and functional studies of the aorta revealed increased arterial stiffness and altered vascular smooth muscle cell (VSMC) and endothelial cell reactivity. Interestingly, the AngII-associated changes in the aorta could be largely attributed to alterations in basal VSMC tone and basal nitric oxide efficacy, indicating that, besides structural remodeling of the arterial wall, dysfunctional active components of the aorta play a crucial role in the pathophysiological mechanisms by which AngII treatment induces arterial stiffness.

SELECTION OF CITATIONS
SEARCH DETAIL
...