Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Appl Physiol (1985) ; 136(5): 1195-1208, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38572539

ABSTRACT

Hypertensive postmenopausal women are more likely to develop adverse cardiac remodeling and respond less effectively to drug treatment than men. High-intensity interval exercise (HIIE) is a nonpharmacological strategy for the treatment of hypertension; however, the effectiveness in women remains uncertain. This study was designed to evaluate 1) the effects of HIIE training upon morphological and functional markers of cardiovascular health in female SHR and 2) to determine whether the hormonal shift induced by ovariectomy could influence cardiovascular responses to HIIE. Thirty-six SHR were randomly assigned to four groups: ovariectomized sedentary, ovariectomized trained, sham-operated sedentary, and sham-operated trained. The trained rats performed HIIE 5 days/wk for 8 wk. Blood pressure and echocardiographic measurements were performed before and after training in animals. Cardiac response to ß-adrenergic stimulation and the expression of calcium regulatory proteins and estrogen receptors in heart samples were assessed. Endothelium-dependent vasorelaxation in response to acetylcholine was evaluated in aortic rings as well as the expression of nitric oxide synthase isoforms (eNOS and P-eNOS) by Western blotting. In both groups of trained SHR, HIIE induced eccentric cardiac remodeling with greater inotropic and chronotropic effects, as well as an increase in SERCA and ß1AR expression. However, although the trained rats showed improved endothelial function and expression of eNOS and P-eNOS in the aorta, there was no demonstrated effect on blood pressure. In addition, the responses to HIIE training were not affected by ovariectomy. This work highlights the importance of assessing the cardiovascular efficacy and safety of different exercise modalities in women.NEW & NOTEWORTHY This study reports the effects of high-intensity interval exercise (HIIE) training on cardiac and endothelial function in female hypertensive rats. Despite a lack of effect on blood pressure (BP), HIIE training induces eccentric cardiac remodeling with greater functionals effects. Furthermore, training has beneficial effects on endothelial function. However, ovarian hormones do not seem to modulate cardiac and aortic adaptations to this training modality. All this underlines the need to consider training modalities on the cardiovascular system in women.


Subject(s)
Blood Pressure , High-Intensity Interval Training , Hypertension , Ovariectomy , Physical Conditioning, Animal , Rats, Inbred SHR , Animals , Female , High-Intensity Interval Training/methods , Rats , Blood Pressure/physiology , Hypertension/physiopathology , Hypertension/metabolism , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/methods , Nitric Oxide Synthase Type III/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Ventricular Remodeling/physiology
2.
Biochem Biophys Res Commun ; 642: 83-89, 2023 01 29.
Article in English | MEDLINE | ID: mdl-36566566

ABSTRACT

Extracellular aggregates of wild-type human transthyretin are associated with heart diseases such as wild-type transthyretin (TTR)-derived amyloidosis (ATTR-wt). Due to their strategic location, cardiac fibroblasts act as sentinel cells that sense injury and activate the inflammasome. No studies of the effects of TTR amyloid aggregation on the secretion of inflammatory factors by primary human cardiac fibroblasts (hCFs) have been reported yet. The intracellular internalization of TTR aggregates, which correspond to the early stage of ATTR-wt, were determined using immunofluorescence and Western blotting of cell lysates. A further objective of this study was to analyze the secretion of inflammatory factors by hCFs after analysis of TTR amyloid aggregation using X-MAP® Luminex Assay techniques. We show that TTR aggregates are internalized in hCFs and induce the secretion of both Brain Natriuretic Peptide (BNP) and N-terminal pro B-type Natriuretic Peptide(NT-proBNP). Also, pro-inflammatory mediators such as interleukin-6 (IL-6) and IL-8 are secreted without significant changes in the levels of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs). In conclusion, these findings suggest that IL-6 and IL-8 play important roles in the development of ATTR-wt, and indicate that IL-6 in particular could be a potentially important therapeutic target in patients with ATTR-wt.


Subject(s)
Amyloid Neuropathies, Familial , Prealbumin , Humans , Interleukin-6 , Interleukin-8 , Amyloid Neuropathies, Familial/drug therapy , Amyloid , Fibroblasts
3.
Heart Rhythm ; 19(8): 1352-1362, 2022 08.
Article in English | MEDLINE | ID: mdl-35447308

ABSTRACT

BACKGROUND: The intrinsic cardiac nervous system (ICNS) refers to clusters of neurons, located within the heart, that participate in the neuronal regulation of cardiac functions and that are involved in the initiation of cardiac arrhythmias. Therefore, deciphering its role in cardiac physiology and physiopathology is mandatory. OBJECTIVE: The aim of this study was to provide a phenotypic, electrophysiological, and pharmacological characterization of the mouse ICNS, which is still poorly characterized. METHODS: Global cardiac innervation and phenotypic diversity were investigated using immunohistochemistry on cleared murine hearts and on tissue sections. The patch clamp technique was used for the electrophysiological and pharmacological characterization of isolated mouse intracardiac neurons. RESULTS: We have identified the expression of 7 distinct neuronal markers within the mouse ICNS, thus proving the neurochemical diversity of this network. Of note, it was the first time that the existence of neurons expressing the calcium-binding protein calbindin, neuropeptide Y, and cocaine and amphetamine regulated transcript peptide was described in the mouse. Electrophysiology studies also revealed the existence of 4 different neuronal populations on the basis of their electrical behavior. Finally, we showed that these neurons can be modulated by several neuromodulators. CONCLUSION: This study showed that the mouse ICNS presents a molecular and functional complexity similar to other species and is therefore a suitable model to decipher the role of individual neuronal subtypes regarding the modulation of cardiac function and the initiation of cardiac arrhythmias.


Subject(s)
Arrhythmias, Cardiac , Heart , Animals , Heart/innervation , Mice , Nervous System , Neurons/metabolism , Patch-Clamp Techniques
4.
Cells ; 10(4)2021 04 17.
Article in English | MEDLINE | ID: mdl-33920685

ABSTRACT

If polyunsaturated fatty acids (PUFAs) are generally accepted to be good for health, the mechanisms of their bona fide benefits still remain elusive. Membrane phospholipids (PLs) of the cardiovascular system and skeletal muscles are particularly enriched in PUFAs. The fatty acid composition of PLs is known to regulate crucial membrane properties, including elasticity and plasticity. Since muscle cells undergo repeated cycles of elongation and relaxation, we postulated in the present study that PUFA-containing PLs could be central players for muscle cell adaptation to mechanical constraints. By a combination of in cellulo and in silico approaches, we show that PUFAs, and particularly the ω-3 docosahexaenoic acid (DHA), regulate important properties of the plasma membrane that improve muscle cell resilience to mechanical constraints. Thanks to their unique property to contortionate within the bilayer plane, they facilitate the formation of vacuole-like dilation (VLD), which, in turn, avoid cell breakage under mechanical constraints.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Phospholipids/pharmacology , Stress, Mechanical , Animals , Arachidonic Acid/analysis , Cell Line , Docosahexaenoic Acids/analysis , Male , Mice, Inbred C57BL , Molecular Dynamics Simulation , Organ Specificity/drug effects , Osmosis , Principal Component Analysis
5.
Cell Mol Biol Lett ; 25(1): 50, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33292162

ABSTRACT

BACKGROUND: Human cardiac stem cells expressing the W8B2 marker (W8B2+ CSCs) were recently identified and proposed as a new model of multipotent CSCs capable of differentiating into smooth muscle cells, endothelial cells and immature myocytes. Nevertheless, no characterization of ion channel or calcium activity during the differentiation of these stem cells has been reported. METHODS: The objectives of this study were thus to analyze (using the TaqMan Low-Density Array technique) the gene profile of W8B2+ CSCs pertaining to the regulation of ion channels, transporters and other players involved in the calcium homeostasis of these cells. We also analyzed spontaneous calcium activity (via the GCaMP calcium probe) during the in vitro differentiation of W8B2+ CSCs into cardiac myocytes. RESULTS: Our results show an entirely different electrophysiological genomic profile between W8B2+ CSCs before and after differentiation. Some specific nodal genes, such as Tbx3, HCN, ICaT, L, KV, and NCX, are overexpressed after this differentiation. In addition, we reveal spontaneous calcium activity or a calcium clock whose kinetics change during the differentiation process. A pharmacological study carried out on differentiated W8B2+ CSCs showed that the NCX exchanger and IP3 stores play a fundamental role in the generation of these calcium oscillations. CONCLUSIONS: Taken together, the present results provide important information on ion channel expression and intrinsic calcium dynamics during the differentiation process of stem cells expressing the W8B2 marker.


Subject(s)
Antigens, Surface/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Ion Channels/metabolism , Myocytes, Cardiac/metabolism , Stem Cells/metabolism , Aged , Cell Proliferation/physiology , Cells, Cultured , Endothelial Cells/metabolism , Female , Gene Expression/physiology , Humans , Male , Multipotent Stem Cells/metabolism , Myocytes, Smooth Muscle/metabolism
6.
Pflugers Arch ; 472(12): 1719-1732, 2020 12.
Article in English | MEDLINE | ID: mdl-33047172

ABSTRACT

Cardiac fibroblasts play an important role in cardiac matrix turnover and are involved in cardiac fibrosis development. Ca2+ is a driving belt in this phenomenon. This study evaluates the functional expression and contribution of the Ca2+-activated channel TRPM4 in atrial fibroblast phenotype. Molecular and electrophysiological investigations were conducted in human atrial fibroblasts in primary culture and in atrial fibroblasts obtained from wild-type and transgenic mice with disrupted Trpm4 gene (Trpm4-/-). A typical TRPM4 current was recorded on human cells (equal selectivity for Na+ and K+, activation by internal Ca2+, voltage sensitivity, conductance of 23.2 pS, inhibition by 9-phenanthrol (IC50 = 6.1 × 10-6 mol L-1)). Its detection rate was 13% on patches at days 2-4 in culture but raised to 100% on patches at day 28. By the same time, a cell growth was observed. This growth was smaller when cells were maintained in the presence of 9-phenanthrol. Similar cell growth was measured on wild-type mice atrial fibroblasts during culture. However, this growth was minimized on Trpm4-/- mice fibroblasts compared to control animals. In addition, the expression of alpha smooth muscle actin increased during culture of atrial fibroblasts from wild-type mice. This was not observed in Trpm4-/- mice fibroblasts. It is concluded that TRPM4 participates in fibroblast growth and could thus be involved in cardiac fibrosis.


Subject(s)
Endomyocardial Fibrosis/metabolism , Myofibroblasts/metabolism , TRPM Cation Channels/metabolism , Action Potentials , Aged , Animals , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Mice , Myocardium/cytology , Myofibroblasts/drug effects , Myofibroblasts/physiology , Phenanthrenes
7.
Dis Model Mech ; 13(6)2020 06 15.
Article in English | MEDLINE | ID: mdl-32303571

ABSTRACT

The balance within phospholipids (PLs) between saturated fatty acids and monounsaturated or polyunsaturated fatty acids is known to regulate the biophysical properties of cellular membranes. As a consequence, in many cell types, perturbing this balance alters crucial cellular processes, such as vesicular budding and the trafficking/function of membrane-anchored proteins. The worldwide spread of the Western diet, which is highly enriched in saturated fats, has been clearly correlated with the emergence of a complex syndrome known as metabolic syndrome (MetS). MetS is defined as a cluster of risk factors for cardiovascular diseases, type 2 diabetes and hepatic steatosis; however, no clear correlations have been established between diet-induced fatty acid redistribution within cellular PLs and the severity/chronology of the symptoms associated with MetS or the function of the targeted organs. To address this issue, in this study we analyzed PL remodeling in rats exposed to a high-fat/high-fructose diet (HFHF) over a 15-week period. PL remodeling was analyzed in several organs, including known MetS targets. We show that fatty acids from the diet can redistribute within PLs in a very selective manner, with phosphatidylcholine being the preferred sink for this redistribution. Moreover, in the HFHF rat model, most organs are protected from this redistribution, at least during the early onset of MetS, at the expense of the liver and skeletal muscles. Interestingly, such a redistribution correlates with clear-cut alterations in the function of these organs.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Fatty Acids/metabolism , Metabolic Syndrome/metabolism , Phospholipids/metabolism , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Dietary Sugars , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Fructose , Lipidomics , Liver/metabolism , Liver/pathology , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Rats, Wistar , Time Factors
8.
Aging Cell ; 18(2): e12894, 2019 04.
Article in English | MEDLINE | ID: mdl-30680911

ABSTRACT

With the onset of advanced age, cardiac-associated pathologies have increased in prevalence. The hallmarks of cardiac aging include cardiomyocyte senescence, fibroblast proliferation, inflammation, and hypertrophy. The imbalance between levels of reactive oxygen species (ROS) and antioxidant enzymes is greatly enhanced in aging cells, promoting cardiac remodeling. In this work, we studied the long-term impact of phenolic compounds (PC) on age-associated cardiac remodeling. Three-month-old Wistar rats were treated for 14 months till middle-age with either 2.5, 5, 10, or 20 mg kg-1  day-1 of PC. PC treatment showed a dose-dependent preservation of cardiac ejection fraction and fractional shortening as well as decreased hypertrophy reflected by left ventricular chamber diameter and posterior wall thickness as compared to untreated middle-aged control animals. Analyses of proteins from cardiac tissue showed that PC attenuated several hypertrophic pathways including calcineurin/nuclear factor of activated T cells (NFATc3), calcium/calmodulin-dependent kinase II (CAMKII), extracellular regulated kinase 1/2 (ERK1/2), and glycogen synthase kinase 3ß (GSK 3ß). PC-treated groups exhibited reduced plasma inflammatory and fibrotic markers and revealed as well ameliorated extracellular matrix remodeling and interstitial inflammation by a downregulated p38 pathway. Myocardia from PC-treated middle-aged rats presented less fibrosis with suppression of profibrotic transforming growth factor-ß1 (TGF-ß1) Smad pathway. Additionally, reduction of apoptosis and oxidative damage in the PC-treated groups was reflected by elevated antioxidant enzymes and reduced RNA/DNA damage markers. Our findings pinpoint that a daily consumption of phenolic compounds could preserve the heart from the detrimental effects of aging storm.


Subject(s)
Aging , Models, Biological , Phenols/pharmacology , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects , Administration, Oral , Animals , Apoptosis/drug effects , Diet , Dose-Response Relationship, Drug , Echocardiography , Male , Oxidative Stress/drug effects , Phenols/administration & dosage , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ventricular Dysfunction, Left/metabolism
9.
Antioxidants (Basel) ; 7(6)2018 Jun 04.
Article in English | MEDLINE | ID: mdl-29866989

ABSTRACT

The high diversity of phenolic compounds (PC) found in food matrices makes it challenging to analyze their bioavailability and their impact on health and functional metabolism. It is well recognized that PC do modulate the composition of the gut microbiota (GM), however, the literature still lacks significant data concerning the link between the metabolic fate of the ingested compounds and their bioactivity, mainly when considering the secondary metabolites produced. In this study, we assessed the metabolic fate of PC for a period covering 14 months of daily intake to identify the metabolites that could be responsible for the effects of PC on the GM observed in our previous work. Urinary analysis of polyphenol metabolites was performed using a high resolution mass spectrometry LC-QTOF-MS method. Among the sixteen metabolites identified, 3-hydroxyphenylacetic acid and 2-(4-hydroxyphenyl) propionic acid were detected simultaneously and, therefore, correlated with the growth of Bifidobacterium in the rat GM. In addition, Daidzedin, detected only at 14 months post-treatment, mostly interfered with the growth inhibition of Clostridium (Cluster I). In conclusion, the impact of the long-term intake of PC on rat GM seems to be related to specific metabolites produced after ingestion of the parental compounds and this may also be due to their additional synergistic effects.

10.
FEBS J ; 285(3): 518-530, 2018 02.
Article in English | MEDLINE | ID: mdl-29211342

ABSTRACT

Recently, a new population of resident cardiac stem cells (CSCs) positive for the W8B2 marker has been identified. These CSCs are considered to be an ideal cellular source to repair myocardial damage after infarction. However, the electrophysiological profile of these cells has not been characterized yet. We first establish the conditions of isolation and expansion of W8B2+ CSCs from human heart biopsies using a magnetic sorting system followed by flow cytometry cell sorting. These cells display a spindle-shaped morphology, are highly proliferative, and possess self-renewal capacity demonstrated by their ability to form colonies. Besides, W8B2+ CSCs are positive for mesenchymal markers but negative for hematopoietic and endothelial ones. RT-qPCR and immunostaining experiments show that W8B2+ CSCs express some early cardiac-specific transcription factors but lack the expression of cardiac-specific structural genes. Using patch clamp in the whole-cell configuration, we show for the first time the electrophysiological signature of BKCa current in these cells. Accordingly, RT-PCR and western blotting analysis confirmed the presence of BKCa at both mRNA and protein levels in W8B2+ CSCs. Interestingly, BKCa channel inhibition by paxilline decreased cell proliferation in a concentration-dependent manner and halted cell cycle progression at the G0/G1 phase. The inhibition of BKCa also decreased the self-renewal capacity but did not affect migration of W8B2+ CSCs. Taken together, our results are consistent with an important role of BKCa channels in cell cycle progression and self-renewal in human cardiac stem cells.


Subject(s)
Antigens, Surface/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myocytes, Cardiac/metabolism , Stem Cells/metabolism , Biomarkers/metabolism , Calcium Channel Blockers/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Separation , Cell Shape/drug effects , Cells, Cultured , Flow Cytometry , Humans , Immunomagnetic Separation , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Microspheres , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , RNA, Messenger/metabolism , Resting Phase, Cell Cycle/drug effects , Stem Cells/cytology , Stem Cells/drug effects
11.
J Food Sci ; 83(1): 246-251, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29227528

ABSTRACT

The aim of this work is to evaluate the impact on the rat microbiota of long-term feeding with phenolic compounds (PC) rich grape pomace extracts. Thirty, 2-mo-old rats, were divided into 5 groups. Four groups were treated with different concentrations of PC (2.5, 5, 10, and 20 mg/kg/d diluted in 0.1% DMSO), and 1 group received 0.1% Dimethyl Sulfoxide (DMSO) alone (control group). The daily treatment lasted 14 mo. Major phenolic compounds constituents were characterized by the high-performance liquid chromatography and free radical scavenging capacity was measured by means of the DPPH assay. Fecal samples from young rats (2-mo old), and rats daily fed with PC or DMSO were collected at 6 and 14 mo posttreatment. The gut microbiota composition was analyzed by quantitative polymerase chain reaction. Bifidobacterium was significantly higher in the groups PC 2.5 and PC 5 than in control and young rats. Lactobacillus decreased with time in all treated and untreated groups. Bacteroides, Clostridium leptum subgroup (Clostridium cluster IV), and Enterococcus were not significantly changed by PC at any concentration when compared to control; nevertheless, after 14 mo of treatment all concentrations of PC abolished the increase of Clostridium sensu stricto (cluster I) (Clostridium Cluster I) observed in the control group when compared to young rats. PC do modulate selectively rat gut microbiome to a healthier phenotype in long-term feeding rats, and could counteract the adverse outcomes of aging on gut bacterial population. PRACTICAL APPLICATION: This research shows that phenolic-rich grape pomace extracts exhibiting a high antioxidant activity, selectively modulate rat gut microbiota to a healthier phenotype within age in a long-term feeding rats.


Subject(s)
Gastrointestinal Microbiome/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Vitis/chemistry , Animals , Bifidobacterium/isolation & purification , Clostridium/isolation & purification , Feces/microbiology , Gastrointestinal Tract/microbiology , Lactobacillus/isolation & purification , Male , Rats , Rats, Wistar
12.
Br J Pharmacol ; 174(23): 4449-4463, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28941245

ABSTRACT

BACKGROUND AND PURPOSE: We evaluated the concordance of results from two sets of nonclinical cardiovascular safety studies on pitolisant. EXPERIMENTAL APPROACH: Nonclinical studies envisaged both in the International Conference on Harmonization (ICH) S7B guideline and Comprehensive in vitro Pro-arrhythmia Assay (CiPA) initiative were undertaken. The CiPA initiative included in vitro ion channels, stem cell-derived human ventricular myocytes, and in silico modelling to simulate human ventricular electrophysiology. ICH S7B-recommended assays included in vitro hERG (KV 11.1) channels, in vivo dog studies with follow-up investigations in rabbit Purkinje fibres and the in vivo Carlsson rabbit pro-arrhythmia model. KEY RESULTS: Both sets of nonclinical data consistently excluded pitolisant from having clinically relevant QT-liability or pro-arrhythmic potential. CiPA studies revealed pitolisant to have modest calcium channel blocking and late INa reducing activities at high concentrations, which resulted in pitolisant reducing dofetilide-induced early after-depolarizations (EADs) in the ICH S7B studies. Studies in stem cell-derived human cardiomyocytes with dofetilide or E-4031 given alone and in combination with pitolisant confirmed these properties. In silico modelling confirmed that the ion channel effects measured are consistent with results from both the stem cell-derived cardiomyocytes and rabbit Purkinje fibres and categorized pitolisant as a drug with low torsadogenic potential. Results from the two sets of nonclinical studies correlated well with those from two clinical QT studies. CONCLUSIONS AND IMPLICATIONS: Our findings support the CiPA initiative but suggest that sponsors should consider investigating drug effects on EADs and the use of pro-arrhythmia models when the results from CiPA studies are ambiguous.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Myocytes, Cardiac/drug effects , Piperidines/adverse effects , Animals , Arrhythmias, Cardiac/physiopathology , Computer Simulation , Disease Models, Animal , Dogs , Electrocardiography , Female , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Male , Myocytes, Cardiac/metabolism , Purkinje Fibers/drug effects , Purkinje Fibers/metabolism , Rabbits , Research Design
13.
J Cell Physiol ; 232(4): 725-730, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27685887

ABSTRACT

Cardiac fibroblasts are commonly known as supporting cells of the cardiac network and exert many essential functions that are fundamental for normal cardiac growth as well as for cardiac remodeling process during pathological conditions. This review focuses on the roles of cardiac fibroblasts in the formation and regulation of the extracellular matrix components, and in maintaining structural, biochemical and mechanical properties of the heart. Additionally, though considered as non-excitable cells, we review the functional expression in cardiac fibroblasts of a wide variety of transmembrane ion channels which activity may contribute to key regulation of cardiac physiological processes. All together, cardiac fibroblasts which actively participate to fundamental regulation of cardiac physiology and physiopathology processes may represent pertinent targets for pharmacological approaches of cardiac diseases and lead to new tracks of therapeutic strategies. J. Cell. Physiol. 232: 725-730, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Fibroblasts/metabolism , Myocardium/cytology , Signal Transduction , Animals , Cell Shape , Fibroblasts/cytology , Fibrosis , Humans , Mechanotransduction, Cellular
14.
J Mol Cell Cardiol ; 68: 12-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412532

ABSTRACT

Cardiac fibroblasts are an integral part of the myocardial tissue and contribute to its remodelling. This study characterises for the first time the calcium-dependent chloride channels (CaCC) in the plasma membrane of primary human atrial cardiac fibroblasts by means of the iodide efflux and the patch clamp methods. The calcium ionophore A23187 and Angiotensin II (Ang II) activate a chloride conductance in cardiac fibroblasts that shares pharmacological similarities with calcium-dependent chloride channels. This chloride conductance is depressed by RNAi-mediated selective Anoctamine 1 (ANO1) but not by Anoctamine 2 (ANO2) which has been revealed as CaCC and is inhibited by the selective ANO1 inhibitor, T16inh-A01. The effect of Ang II on anion efflux is mediated through AT1 receptors (with an EC50 = 13.8 ± 1.3 nM). The decrease of anion efflux by calphostin C and bisindolylmaleimide I (BIM I) suggests that chloride conductance activation is dependent on PKC. We conclude that ANO1 contributes to CaCC current in human cardiac fibroblasts and that this is regulated by Ang II acting via the AT1 receptor pathway.


Subject(s)
Angiotensin II/physiology , Calcium Signaling , Chloride Channels/physiology , Fibroblasts/metabolism , Neoplasm Proteins/physiology , Aged , Anoctamin-1 , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Chlorides/metabolism , Female , Heart Atria/cytology , Humans , Kinetics , Male , Receptor, Angiotensin, Type 1/metabolism
15.
Cell Biochem Biophys ; 66(3): 723-36, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23400933

ABSTRACT

A dilated cardiomyopathy (DCM) is associated with Duchenne muscular dystrophy (DMD). The loss of dystrophin leads to membrane instability and calcium dysregulation in skeletal muscle but effects of such a loss are not elucidated at cardiomyocytes level. We sought to examine whether membrane and transverse tubules damages occur in ventricular myocytes from mdx mouse model of DMD and how they impact the function of single excitation-contraction coupling elements. Scanning ion conductance microscopy (SICM) was used to characterize the integrity loss of living mdx cardiomyocytes surface. 2D Fourier transform analysis of labeled internal networks (transverse tubules, alpha-actinin, dihydropyridine receptors, ryanodine receptors) was performed to evaluate internal alterations. During calcium measurements, "smart microperfusions" of depolarizing solutions were applied through SICM nanopipette, stimulating single tubules elements. These approaches revealed structural membrane surface (39% decrease for Z-groove ratio) and transverse tubules disorganization (21% transverse tubules ratio decrease) in mdx as compared to control. These disruptions were associated with functional alterations (sixfold increase of calcium signal duration and twofold increase of sparks frequency). In DCM associated with DMD, myocytes display evident membrane alterations at the surface level but also in the cell depth with a disruption of transverse tubules network as observed in other cases of heart failure. These ultrastructural changes are associated with changes in the function of some coupling elements. Thus, these profound disruptions may play a role in calcium dysregulation through excitation-contraction coupling elements perturbation and suggest a transverse tubules stabilizing role for dystrophin.


Subject(s)
Cell Membrane/ultrastructure , Excitation Contraction Coupling , Molecular Imaging , Myocytes, Cardiac/cytology , Animals , Calcium/metabolism , Cardiomyopathy, Dilated/pathology , Cell Membrane/metabolism , Intracellular Space/metabolism , Male , Mice , Mice, Inbred mdx , Myocytes, Cardiac/ultrastructure , Sarcolemma/metabolism , Sarcolemma/ultrastructure
16.
PLoS One ; 7(11): e48690, 2012.
Article in English | MEDLINE | ID: mdl-23133651

ABSTRACT

Brugada syndrome (BrS) is an inherited autosomal dominant cardiac channelopathy. Several mutations on the cardiac sodium channel Na(v)1.5 which are responsible for BrS lead to misfolded proteins that do not traffic properly to the plasma membrane. In order to mimic patient heterozygosity, a trafficking defective mutant, R1432G was co-expressed with Wild Type (WT) Na(v)1.5 channels in HEK293T cells. This mutant significantly decreased the membrane Na current density when it was co-transfected with the WT channel. This dominant negative effect did not result in altered biophysical properties of Na(v)1.5 channels. Luminometric experiments revealed that the expression of mutant proteins induced a significant reduction in membrane expression of WT channels. Interestingly, we have found that the auxiliary Na channel ß(1)-subunit was essential for this dominant negative effect. Indeed, the absence of the ß(1)-subunit prevented the decrease in WT sodium current density and surface proteins associated with the dominant negative effect. Co-immunoprecipitation experiments demonstrated a physical interaction between Na channel α-subunits. This interaction occurred only when the ß(1)-subunit was present. Our findings reveal a new role for ß(1)-subunits in cardiac voltage-gated sodium channels by promoting α-α subunit interaction which can lead to a dominant negative effect when one of the α-subunits shows a trafficking defective mutation.


Subject(s)
Brugada Syndrome/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/physiology , DNA, Complementary/metabolism , Electrophysiology/methods , Genes, Dominant , Green Fluorescent Proteins/metabolism , HEK293 Cells , Heterozygote , Humans , Immunoblotting/methods , Immunoprecipitation , Microscopy, Fluorescence/methods , Mutation , Patch-Clamp Techniques , Protein Binding , Sodium/chemistry
17.
J Physiol ; 590(17): 4307-19, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22802584

ABSTRACT

Fibroblasts play a major role in heart physiology. They are at the origin of the extracellular matrix renewal and production of various paracrine and autocrine factors. In pathological conditions, fibroblasts proliferate, migrate and differentiate into myofibroblasts leading to cardiac fibrosis. This differentiated status is associated with changes in expression profile leading to neo-expression of proteins such as ionic channels. The present study investigates further electrophysiological changes associated with fibroblast differentiation focusing on the activity of voltage-gated sodium channels in human atrial fibroblasts and myofibroblasts. Using the patch clamp technique we show that human atrial myofibroblasts display a fast inward voltage gated sodium current with a density of 13.28 ± 2.88 pA pF(-1) whereas no current was detectable in non-differentiated fibroblasts. Quantitative RT-PCR reveals a large amount of transcripts encoding the Na(v)1.5 α-subunit with a fourfold increased expression level in myofibroblasts when compared to fibroblasts. Accordingly, half of the current was blocked by 1 µm of tetrodotoxin and immunocytochemistry experiments reveal the presence of Na(v)1.5 proteins. Overall, this current exhibits similar biophysical characteristics to sodium currents found in cardiac myocytes except for the window current that is enlarged for potentials between -100 and -20 mV. Since fibrosis is one of the fundamental mechanisms implicated in atrial fibrillation, it is of great interest to investigate how this current could influence myofibroblast properties. Moreover, since several Na(v)1.5 mutations are related to cardiac pathologies, this study offers a new avenue on the fibroblasts involvement of these mutations.


Subject(s)
Heart Atria/cytology , Heart Atria/metabolism , Myofibroblasts/cytology , Myofibroblasts/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Aged , Aged, 80 and over , Cell Differentiation , Cells, Cultured , Electrophysiological Phenomena , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , HEK293 Cells , Humans , Male , Middle Aged , Mutation , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Protein Subunits , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 408(1): 6-11, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21420933

ABSTRACT

The aim of this study was to characterize the effects of sphingosine-1-phosphate (S1P) on cardiac ventricular fibroblasts. Impacts of S1P on fibroblast excitability, cell migration, proliferation and secretion were characterized. The patch-clamp technique in the whole-cell configuration was used to study the S1P-induced current from mouse ventricular fibroblasts. The expression level of the S1P receptor during cell culture duration was evaluated by western-blot. Fibroblast proliferation and migration were quantified using the methylene blue assay and the Boyden chamber technique, respectively. Finally, fibroblast secretion properties were estimated by quantification of the IL-6 and collagen levels using ELISA and SIRCOL collagen assays, respectively. We found that S1P activated SUR2/Kir6.1 channel and that this effect was sensitive to specific inhibition of the S1P receptor of type 3 (S1P3R). In contrast, S1P1R receptor inhibition had no effect. Moreover, the S1P-induced current increased with cell culture duration whereas S1P3R expression level remained constant. The activation of SUR2/Kir6.1 channel by S1P via S1P3R stimulated cell proliferation and decreased IL-6 and collagen secretions. S1P also stimulated fibroblast migration via S1P3R but independently from SUR2/Kir6.1 channel activation. This study demonstrates that S1P, via S1P3R, affects cardiac ventricular fibroblasts function independently or through activation of SUR2/Kir6.1 channel. The latter effect occurs after fibroblasts differentiate into myofibroblasts, opening a new potential therapeutic strategy to modulate fibrosis after cardiac physiopathological injury.


Subject(s)
Fibroblasts/physiology , Heart Ventricles/metabolism , Lysophospholipids/physiology , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Ventricular Function/physiology , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Glyburide/pharmacology , Heart Ventricles/cytology , Heart Ventricles/drug effects , Interleukin-6/metabolism , KATP Channels/metabolism , Lysophospholipids/pharmacology , Mice , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , Receptors, Lysosphingolipid/agonists , Sphingosine/pharmacology , Sphingosine/physiology , Sphingosine-1-Phosphate Receptors , Sulfonylurea Receptors , Ventricular Function/drug effects
19.
J Mol Cell Cardiol ; 46(4): 508-17, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19166858

ABSTRACT

The present work is aimed at identifying and characterizing, at a molecular and functional level, new ionic conductances potentially involved in the excitation-secretion coupling and proliferation of cardiac ventricular fibroblasts. Among potassium channel transcripts which were screened by high-throughput real-time PCR, SUR2 and Kir6.1 mRNAs were found to be the most abundant in ventricular fibroblasts. The corresponding proteins were not detected by western blot following 5 days of cell culture, but had appeared at 7 days, increasing with extended cell culture duration as the fibroblasts differentiated into myofibroblasts. Using the inside-out configuration of the patch-clamp technique, single potassium channels could be recorded. These had properties similar to those reported for SUR2/Kir6.1 channels, i.e. activation by pinacidil, inhibition by glibenclamide and activation by intracellular UDP. As already reported for this molecular signature, they were insensitive to intracellular ATP. In the whole-cell configuration, these channels have been shown to be responsible for a glibenclamide-sensitive macroscopic potassium current which can be activated not only by pinacidil, but also by nanomolar concentrations of the sphingolipid sphingosine-1-phosphate (S1P). The activation of this current resulted in an increase in cell proliferation and a decrease in IL-6 secretion, suggesting it has a functional role in situations where S1P increases. Overall, this work demonstrates for the first time that SUR2/Kir6.1 channels represent a significant potassium conductance in ventricular fibroblasts which may be activated in physio-pathological conditions and which may impact on fibroblast proliferation and function.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Fibroblasts/metabolism , Heart Ventricles/cytology , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/genetics , Actins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Glyburide/pharmacology , Heart Ventricles/metabolism , Interleukin-6/metabolism , Ion Channel Gating/drug effects , KATP Channels , Lysophospholipids/pharmacology , Mice , Pinacidil/pharmacology , Potassium Channels, Inwardly Rectifying/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Drug/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sulfonylurea Receptors
20.
Biochim Biophys Acta ; 1778(10): 2097-104, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18482576

ABSTRACT

Cardiac fibroblasts contribute to the structure and function of the myocardium. However their involvement in electrophysiological processes remains unclear; particularly in pathological situations when they proliferate and develop fibrosis. We have identified the connexins involved in gap junction channels between fibroblasts from adult mouse heart and characterized their functional coupling. RT-PCR and Western blotting results show that mRNA and proteins of connexin40 and connexin43 are expressed in cultured cardiac fibroblasts, while Cx45 is not detected. Analysis of gap junctional communications established by these connexins with the gap-FRAP technique demonstrates that fibroblasts are functionally coupled. The time constant of permeability, k, calculated from the fluorescence recovery curves between cell pairs is 0.066+/-0.005 min(-1) (n = 65). Diffusion analysis of Lucifer Yellow through gap junction channels with the scrape-loading method demonstrates that when they are completely confluent, a majority of fibroblasts are coupled forming an interconnecting network over a distance of several hundred micrometers. These data show that cardiac fibroblasts express connexin40 and connexin43 which are able to establish functional communications through homo and/or heterotypic junctions to form an extensive coupled cell network. It should then be interesting to study the conditions to improve efficiency of this coupling in pathological conditions.


Subject(s)
Connexin 43/metabolism , Connexins/metabolism , Fibroblasts/metabolism , Gap Junctions/metabolism , Heart Conduction System/metabolism , Myocardium , Animals , Cells, Cultured , Connexin 43/genetics , Connexins/genetics , Fibroblasts/cytology , Gap Junctions/chemistry , Mice , Myocardium/cytology , Myocardium/metabolism , Gap Junction alpha-5 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...