Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Circ Res ; 134(12): 1752-1766, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843295

ABSTRACT

Heart failure (HF) is characterized by a progressive decline in cardiac function and represents one of the largest health burdens worldwide. Clinically, 2 major types of HF are distinguished based on the left ventricular ejection fraction (EF): HF with reduced EF and HF with preserved EF. While both types share several risk factors and features of adverse cardiac remodeling, unique hallmarks beyond ejection fraction that distinguish these etiologies also exist. These differences may explain the fact that approved therapies for HF with reduced EF are largely ineffective in patients suffering from HF with preserved EF. Improving our understanding of the distinct cellular and molecular mechanisms is crucial for the development of better treatment strategies. This article reviews the knowledge of the immunologic mechanisms underlying HF with reduced and preserved EF and discusses how the different immune profiles elicited may identify attractive therapeutic targets for these conditions. We review the literature on the reported mechanisms of adverse cardiac remodeling in HF with reduced and preserved EF, as well as the immune mechanisms involved. We discuss how the knowledge gained from preclinical models of the complex syndrome of HF as well as from clinical data obtained from patients may translate to a better understanding of HF and result in specific treatments for these conditions in humans.


Subject(s)
Heart Failure , Stroke Volume , Ventricular Remodeling , Humans , Heart Failure/physiopathology , Heart Failure/immunology , Animals , Myocarditis/physiopathology , Myocarditis/immunology , Ventricular Function, Left , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology
2.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727271

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.


Subject(s)
Aortic Aneurysm, Thoracic , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Male , Mice , Angiotensin II/pharmacology , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Apoptosis/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction
3.
J Physiol ; 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38642051

ABSTRACT

Macrophages (MΦ) play pivotal roles in tissue homeostasis and repair. Their mechanical environment has been identified as a key modulator of various cell functions, and MΦ mechanosensitivity is likely to be critical - in particular in a rhythmically contracting organ such as the heart. Cultured MΦ, differentiated in vitro from bone marrow (MΦBM), form a popular research model. This study explores the activity of mechanosensitive ion channels (MSC) in murine MΦBM and compares it to MSC activity in MΦ enzymatically isolated from cardiac tissue (tissue-resident MΦ; MΦTR). We show that MΦBM and MΦTR have stretch-induced currents, indicating the presence of functional MSC in their plasma membrane. The current profiles in MΦBM and in MΦTR show characteristics of cation non-selective MSC such as Piezo1 or transient receptor potential channels. While Piezo1 ion channel activity is detectable in the plasma membrane of MΦBM using the patch-clamp technique, or by measuring cytosolic calcium concentration upon perfusion with the Piezo1 channel agonist Yoda1, no Piezo1 channel activity was observed in MΦTR. The selective transient receptor potential vanilloid 4 (TRPV4) channel agonist GSK1016790A induces calcium entry in MΦTR and in MΦBM. In MΦ isolated from left-ventricular scar tissue 28 days after cryoablation, stretch-induced current characteristics are not significantly different compared to non-injured control tissue, even though scarred ventricular tissue is expected to be mechanically remodelled and to contain an altered composition of pre-existing cardiac and circulation-recruited MΦ. Our data suggest that the in vitro differentiation protocols used to obtain MΦBM generate cells that differ from MΦ recruited from the circulation during tissue repair in vivo. Further investigations are needed to explore MSC identity in lineage-traced MΦ in scar tissue, and to compare mechanosensitivity of circulating monocytes with that of MΦBM. KEY POINTS: Bone marrow-derived (MΦBM) and tissue resident (MΦTR) macrophages have stretch-induced currents, indicating expression of functional mechanosensitive channels (MSC) in their plasma membrane. Stretch-activated current profiles show characteristics of cation non-selective MSC; and mRNA coding for MSC, including Piezo1 and TRPV4, is expressed in murine MΦBM and in MΦTR. Calcium entry upon pharmacological activation of TRPV4 confirms functionality of the channel in MΦTR and in MΦBM. Piezo1 ion channel activity is detected in the plasma membrane of MΦBM but not in MΦTR, suggesting that MΦBM may not be a good model to study the mechanotransduction of MΦTR. Stretch-induced currents, Piezo1 mRNA expression and response to pharmacological activation are not significantly changed in cardiac MΦ 28 days after cryoinjury compared to sham operated mice.

4.
Philos Trans R Soc Lond B Biol Sci ; 377(1864): 20210326, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36189808

ABSTRACT

Cardiomyocytes sense and shape their mechanical environment, contributing to its dynamics by their passive and active mechanical properties. While axial forces generated by contracting cardiomyocytes have been amply investigated, the corresponding radial mechanics remain poorly characterized. Our aim is to simultaneously monitor passive and active forces, both axially and radially, in cardiomyocytes freshly isolated from adult mouse ventricles. To do so, we combine a carbon fibre (CF) set-up with a custom-made atomic force microscope (AFM). CF allows us to apply stretch and to record passive and active forces in the axial direction. The AFM, modified for frontal access to fit in CF, is used to characterize radial cell mechanics. We show that stretch increases the radial elastic modulus of cardiomyocytes. We further find that during contraction, cardiomyocytes generate radial forces that are reduced, but not abolished, when cells are forced to contract near isometrically. Radial forces may contribute to ventricular wall thickening during contraction, together with the dynamic re-orientation of cells and sheetlets in the myocardium. This new approach for characterizing cell mechanics allows one to obtain a more detailed picture of the balance of axial and radial mechanics in cardiomyocytes at rest, during stretch, and during contraction. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.


Subject(s)
Myocytes, Cardiac , Animals , Carbon Fiber , Mice , Microscopy, Atomic Force/methods
5.
Adv Biol (Weinh) ; 6(7): e2000337, 2022 07.
Article in English | MEDLINE | ID: mdl-35481696

ABSTRACT

In the rapidly expanding field of molecular optogenetics, the performance of the engineered systems relies on the switching properties of the underlying genetically encoded photoreceptors. In this study, the bacterial phytochromes Cph1 and DrBphP are engineered, recombinantly produced in Escherichia coli, and characterized regarding their switching properties in order to synthesize biohybrid hydrogels with increased light-responsive stiffness modulations. The R472A mutant of the cyanobacterial phytochrome 1 (Cph1) is identified to confer the phytochrome-based hydrogels with an increased dynamic range for the storage modulus but a different light-response for the loss modulus compared to the original Cph1-based hydrogel. Stiffness measurements of human atrial fibroblasts grown on these hydrogels suggest that differences in the loss modulus at comparable changes in the storage modulus affect cell stiffness and thus underline the importance of matrix viscoelasticity on cellular mechanotransduction. The hydrogels presented here are of interest for analyzing how mammalian cells respond to dynamic viscoelastic cues. Moreover, the Cph1-R472A mutant, as well as the benchmarking of the other phytochrome variants, are expected to foster the development and performance of future optogenetic systems.


Subject(s)
Bacterial Proteins , Hydrogels , Mechanotransduction, Cellular , Optogenetics , Photoreceptors, Microbial , Phytochrome , Protein Kinases , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/radiation effects , Benchmarking , Cyanobacteria/genetics , Escherichia coli/metabolism , Fibroblasts , Genetic Engineering , Humans , Hydrogels/chemistry , Mechanotransduction, Cellular/radiation effects , Photoreceptors, Microbial/chemistry , Photoreceptors, Microbial/genetics , Photoreceptors, Microbial/radiation effects , Phytochrome/chemistry , Phytochrome/genetics , Phytochrome/radiation effects , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/radiation effects , Viscosity
6.
Biophys Rev ; 13(5): 587-610, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34765043

ABSTRACT

Passive mechanical tissue properties are major determinants of myocardial contraction and relaxation and, thus, shape cardiac function. Tightly regulated, dynamically adapting throughout life, and affecting a host of cellular functions, passive tissue mechanics also contribute to cardiac dysfunction. Development of treatments and early identification of diseases requires better spatio-temporal characterisation of tissue mechanical properties and their underlying mechanisms. With this understanding, key regulators may be identified, providing pathways with potential to control and limit pathological development. Methodologies and models used to assess and mimic tissue mechanical properties are diverse, and available data are in part mutually contradictory. In this review, we define important concepts useful for characterising passive mechanical tissue properties, and compare a variety of in vitro and in vivo techniques that allow one to assess tissue mechanics. We give definitions of key terms, and summarise insight into determinants of myocardial stiffness in situ. We then provide an overview of common experimental models utilised to assess the role of environmental stiffness and composition, and its effects on cardiac cell and tissue function. Finally, promising future directions are outlined.

7.
J Mol Cell Cardiol ; 158: 49-62, 2021 09.
Article in English | MEDLINE | ID: mdl-33974928

ABSTRACT

AIMS: Atrial Fibrillation (AF) is an arrhythmia of increasing prevalence in the aging populations of developed countries. One of the important indicators of AF is sustained atrial dilatation, highlighting the importance of mechanical overload in the pathophysiology of AF. The mechanisms by which atrial cells, including fibroblasts, sense and react to changing mechanical forces, are not fully elucidated. Here, we characterise stretch-activated ion channels (SAC) in human atrial fibroblasts and changes in SAC- presence and activity associated with AF. METHODS AND RESULTS: Using primary cultures of human atrial fibroblasts, isolated from patients in sinus rhythm or sustained AF, we combine electrophysiological, molecular and pharmacological tools to identify SAC. Two electrophysiological SAC- signatures were detected, indicative of cation-nonselective and potassium-selective channels. Using siRNA-mediated knockdown, we identified the cation-nonselective SAC as Piezo1. Biophysical properties of the potassium-selective channel, its sensitivity to calcium, paxilline or iberiotoxin (blockers), and NS11021 (activator), indicated presence of calcium-dependent 'big potassium channels' (BKCa). In cells from AF patients, Piezo1 activity and mRNA expression levels were higher than in cells from sinus rhythm patients, while BKCa activity (but not expression) was downregulated. Both Piezo1-knockdown and removal of extracellular calcium from the patch pipette resulted in a significant reduction of BKCa current during stretch. No co-immunoprecipitation of Piezo1 and BKCa was detected. CONCLUSIONS: Human atrial fibroblasts contain at least two types of ion channels that are activated during stretch: Piezo1 and BKCa. While Piezo1 is directly stretch-activated, the increase in BKCa activity during mechanical stimulation appears to be mainly secondary to calcium influx via SAC such as Piezo1. During sustained AF, Piezo1 is increased, while BKCa activity is reduced, highlighting differential regulation of both channels. Our data support the presence and interplay of Piezo1 and BKCa in human atrial fibroblasts in the absence of physical links between the two channel proteins.


Subject(s)
Arrhythmia, Sinus/metabolism , Atrial Fibrillation/metabolism , Atrial Remodeling/genetics , Heart Atria/metabolism , Ion Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myofibroblasts/metabolism , Signal Transduction/genetics , Adult , Aged , Aged, 80 and over , Arrhythmia, Sinus/pathology , Arrhythmia, Sinus/surgery , Atrial Fibrillation/pathology , Atrial Fibrillation/surgery , Atrial Remodeling/drug effects , Calcium/metabolism , Cells, Cultured , Female , Gene Knockdown Techniques , Heart Atria/pathology , Humans , Indoles/pharmacology , Ion Channels/genetics , Ion Transport/drug effects , Ion Transport/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Male , Middle Aged , Peptides/pharmacology , Signal Transduction/drug effects , Tetrazoles/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transfection
8.
Cells ; 10(3)2021 03 16.
Article in English | MEDLINE | ID: mdl-33809739

ABSTRACT

The mechanical environment of cardiac cells changes continuously and undergoes major alterations during diseases. Most cardiac diseases, including atrial fibrillation, are accompanied by fibrosis which can impair both electrical and mechanical function of the heart. A key characteristic of fibrotic tissue is excessive accumulation of extracellular matrix, leading to increased tissue stiffness. Cells are known to respond to changes in their mechanical environment, but the molecular mechanisms underlying this ability are incompletely understood. We used cell culture systems and hydrogels with tunable stiffness, combined with advanced biophysical and imaging techniques, to elucidate the roles of the stretch-activated channel Piezo1 in human atrial fibroblast mechano-sensing. Changing the expression level of Piezo1 revealed that this mechano-sensor contributes to the organization of the cytoskeleton, affecting mechanical properties of human embryonic kidney cells and human atrial fibroblasts. Our results suggest that this response is independent of Piezo1-mediated ion conduction at the plasma membrane, and mediated in part by components of the integrin pathway. Further, we show that Piezo1 is instrumental for fibroblast adaptation to changes in matrix stiffness, and that Piezo1-induced cell stiffening is transmitted in a paracrine manner to other cells by a signaling mechanism requiring interleukin-6. Piezo1 may be a new candidate for targeted interference with cardiac fibroblast function.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Heart Atria/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular , Cytoskeleton/metabolism , Cytoskeleton/pathology , Elastic Modulus , Extracellular Matrix/pathology , Fibroblasts/pathology , Fibrosis , HEK293 Cells , Heart Atria/pathology , Humans , Integrin beta1/metabolism , Interleukin-6/metabolism , Ion Channels/genetics , Paracrine Communication
9.
Naunyn Schmiedebergs Arch Pharmacol ; 394(3): 533-543, 2021 03.
Article in English | MEDLINE | ID: mdl-33064167

ABSTRACT

Cardiovascular diseases are exacerbated and driven by cardiac fibrosis. TGFß induces fibroblast activation and differentiation into myofibroblasts that secrete excessive extracellular matrix proteins leading to stiffening of the heart, concomitant cardiac dysfunction, and arrhythmias. However, effective pharmacotherapy for preventing or reversing cardiac fibrosis is presently unavailable. Therefore, drug repurposing could be a cost- and time-saving approach to discover antifibrotic interventions. The aim of this study was to investigate the antifibrotic potential of mesalazine in a cardiac fibroblast stress model. TGFß was used to induce a profibrotic phenotype in a human cardiac fibroblast cell line. After induction, cells were treated with mesalazine or solvent control. Fibroblast proliferation, key fibrosis protein expression, extracellular collagen deposition, and mechanical properties were subsequently determined. In response to TGFß treatment, fibroblasts underwent a profound phenoconversion towards myofibroblasts, determined by the expression of fibrillary αSMA. Mesalazine reduced differentiation nearly by half and diminished fibroblast proliferation by a third. Additionally, TGFß led to increased cell stiffness and adhesion, which were reversed by mesalazine treatment. Collagen 1 expression and deposition-key drivers of fibrosis-were significantly increased upon TGFß stimulation and reduced to control levels by mesalazine. SMAD2/3 and ERK1/2 phosphorylation, along with reduced nuclear NFκB translocation, were identified as potential modes of action. The current study provides experimental pre-clinical evidence for antifibrotic effects of mesalazine in an in vitro model of cardiac fibrosis. Furthermore, it sheds light on possible mechanisms of action and suggests further investigation in experimental and clinical settings.


Subject(s)
Cardiotonic Agents/therapeutic use , Mesalamine/therapeutic use , Myocardium/pathology , Actins/metabolism , Cardiotonic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line , Collagen Type I/metabolism , Drug Repositioning , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Humans , Mesalamine/pharmacology , Myocardium/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , NF-kappa B/metabolism , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism , Transforming Growth Factor beta
10.
Methods Mol Biol ; 2173: 217-231, 2020.
Article in English | MEDLINE | ID: mdl-32651921

ABSTRACT

Synthetic extracellular matrices with reversibly adjustable mechanical properties are essential for the investigation of how cells respond to dynamic mechanical cues as occurring in living organisms. One interesting approach to engineer dynamic biomaterials is the incorporation of photoreceptors from cyanobacteria or plants into polymer materials. Here, we give an overview of existing photoreceptor-based biomaterials and describe a detailed protocol for the synthesis of a phytochrome-based extracellular matrix (CyPhyGel). Using cell-compatible light in the red and far-red spectrum, the mechanical properties of this matrix can be adjusted in a fully reversible, wavelength-specific, and dose-dependent manner with high spatiotemporal control.


Subject(s)
Light , Phytochrome/metabolism , Extracellular Matrix/metabolism , Hydrogels/chemistry , Optogenetics/methods , Spatio-Temporal Analysis
11.
FEBS Open Bio ; 10(7): 1210-1218, 2020 07.
Article in English | MEDLINE | ID: mdl-32421922

ABSTRACT

Atrial fibrillation (AF) is regularly accompanied by cardiac fibrosis and concomitant heart failure. Due to the heterogeneous nature and complexity of fibrosis, the knowledge about the underlying mechanisms is limited, which prevents effective pharmacotherapy. A deeper understanding of cardiac fibroblasts is essential to meet this need. We previously described phenotypic and functional differences between atrial fibroblasts from patients in sinus rhythm and with AF. Herein, we established and characterized a novel human atrial fibroblast line, which displays typical fibroblast morphology and function comparable to primary cells but with improved proliferation capacity and low spontaneous myofibroblast differentiation. These traits make our model suitable for the study of fibrosis mechanisms and for drug screening aimed at developing effective antifibrotic pharmacotherapy.


Subject(s)
Fibroblasts/metabolism , Fibrosis/metabolism , Heart Atria/metabolism , Models, Biological , Cell Differentiation , Cell Proliferation , Cells, Cultured , Fibroblasts/pathology , Fibrosis/pathology , Heart Atria/pathology , Humans
12.
Front Physiol ; 10: 1526, 2019.
Article in English | MEDLINE | ID: mdl-31998137

ABSTRACT

Fibrosis is associated with aging and many cardiac pathologies. It is characterized both by myofibroblast differentiation and by excessive accumulation of extracellular matrix proteins. Fibrosis-related tissue remodeling results in significant changes in tissue structure and function, including passive mechanical properties. This research area has gained significant momentum with the recent development of new tools and approaches to better characterize and understand the ability of cells to sense and respond to their biophysical environment. We use a novel hydrogel, termed CyPhyGel, to provide an advanced in vitro model of remodeling-related changes in tissue stiffness. Based on light-controlled dimerization of a Cyanobacterial Phytochrome, it enables contactless and reversible tuning of hydrogel mechanical properties with high spatial and temporal resolution. Human primary atrial fibroblasts were cultured on CyPhyGels. After 4 days of culturing on stiff (~4.6 kPa) or soft (~2.7 kPa) CyPhyGels, we analyzed fibroblast cell area and stiffness. Cells grown on the softer substrate were smaller and softer, compared to cells grown on the stiffer substrate. This difference was absent when both soft and stiff growth substrates were combined in a single CyPhyGel, with the resulting cell areas being similar to those on homogeneously stiff gels and cell stiffnesses being similar to those on homogeneously soft substrates. Using CyPhyGels to mimic tissue stiffness heterogeneities in vitro, our results confirm the ability of cardiac fibroblasts to adapt to their mechanical environment, and suggest the presence of a paracrine mechanism that tunes fibroblast structural and functional properties associated with mechanically induced phenotype conversion toward myofibroblasts. In the context of regionally increased tissue stiffness, such as upon scarring or in diffuse fibrosis, such a mechanism could help to prevent abrupt changes in cell properties at the border zone between normal and diseased tissue. The light-tunable mechanical properties of CyPhyGels and their suitability for studying human primary cardiac cells make them an attractive model system for cardiac mechanobiology research. Further investigations will explore the interactions between biophysical and soluble factors in the response of cardiac fibroblasts to spatially and temporally heterogeneous mechanical cues.

13.
Biosens Bioelectron ; 115: 97-103, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29803867

ABSTRACT

The detection of antibodies from blood sera is crucial for diagnostic purposes. Miniaturized protein assays in combination with microfluidic setups hold great potential by enabling automated handling and multiplexed analyses. Yet, the separate expression, purification, and storage of many individual proteins are time consuming and limit applicability. In vitro cell-free expression has been proposed as an alternative procedure for the generation of protein assays. We report the successful in vitro expression of different model proteins from DNA templates with an optimized expression mix. His10-tagged proteins were specifically captured and immobilized on a Ni-NTA coated sensor surface directly from the in vitro expression mix. Finally, the specific binding of antibodies from rabbit-derived blood sera to the immobilized proteins was monitored by imaging reflectometric interferometry (iRIf). Antibodies in the blood sera could be identified by binding to the respective epitopes with minimal cross reactivity. The results show the potential of in vitro expression and label-free detection for binding assays in general and diagnostic purposes in specific.


Subject(s)
Antibodies/blood , Antigens/blood , Biosensing Techniques , Immobilized Proteins/chemistry , Antibodies/chemistry , Interferometry/methods
14.
Herzschrittmacherther Elektrophysiol ; 29(1): 62-69, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29392412

ABSTRACT

Fibrotic areas in cardiac muscle-be it in ventricular or atrial tissue-are considered as obstacles for conduction of the excitatory wave and can therefore facilitate re-entry, which may contribute to the sustenance of cardiac arrhythmias. Persistence of one of the most frequent arrhythmias, atrial fibrillation (AF), is accompanied by enhanced atrial fibrosis. Any kind of myocardial perturbation, whether via mechanical stress or ischemic damage, inflammation, or irregular and high-frequency electrical activity, activates fibroblasts. This leads to the secretion of paracrine factors and extracellular matrix proteins, especially collagen, and to the differentiation of fibroblasts into myofibroblasts. Excessive collagen production is the hallmark of fibrosis and impairs regular impulse propagation. In addition, direct electrical coupling between cardiomyocytes and nonmyocytes, such as fibroblasts and macrophages, via gap junctions affects conduction. Although fibroblasts are not electrically excitable, they express functional ion channels, in particular K+ channels and mechanosensitive channels, some of which could be involved in tissue remodeling. Here, we briefly review these aspects with special reference to AF.


Subject(s)
Atrial Fibrillation/physiopathology , Electrocardiography , Electrophysiological Phenomena/physiology , Fibroblasts/physiology , Heart Atria/physiopathology , Cell Differentiation/physiology , Collagen/metabolism , Fibrosis/physiopathology , Humans , Ion Channels/physiology , Myocytes, Cardiac/physiology , Myofibroblasts/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...