Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
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.
Physiol Rep ; 11(4): e15524, 2023 02.
Article in English | MEDLINE | ID: mdl-36807709

ABSTRACT

In spontaneously hypertensive rats, exercise can lead to a post-exercise decrease in blood pressure, named post-exercise hypotension (PEH). This can be following physical training but also after a single bout of mild to moderate exercise when measured with tail-cuff or externalized catheter methods. Our aim was to assess the PEH obtained with different calculation methods and to compare the magnitude of this effect induced by a moderate-intensity continuous exercise or a high-intensity intermittent exercise. Thirteen 16-week-old male spontaneously hypertensive rats performed two types of aerobic exercise (continuous or intermittent) on a treadmill. Arterial pressure was recorded by telemetry for 24 h which was started 3 h before physical exercise. Based on the literature, PEH was first evaluated with two different baseline values, and then with three different approaches. We observed that the identification of PEH depended on the method used to measure the rest value, and that its amplitude was also influenced by the calculation approach and the type of exercise performed. Hence, the calculation method and the amplitude of the detected PEH can significantly influence their physiological and pathophysiological inferences.


Subject(s)
Hypertension , Hypotension , Physical Conditioning, Animal , Post-Exercise Hypotension , Rats , Animals , Male , Rats, Inbred SHR , Blood Pressure/physiology
3.
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
4.
Cell Calcium ; 103: 102546, 2022 05.
Article in English | MEDLINE | ID: mdl-35144094

ABSTRACT

Calcium takes part in numerous cellular processes such as proliferation, migration, differentiation, or cell death and plays a particular role in myogenesis of skeletal muscle. Indeed, intracellular calcium signaling participates, in a non-negligeable manner, to the "on" signal of muscle differentiation from undifferentiated cells to differentiated myotubes. Therefore, this differentiation can be modulated by controlling calcium activity with electrical or optogenetic stimulation approaches. In this study, we used the optogenetic tool channelrhodopsin 2 (ChR2) to control calcium activity and to modulate skeletal muscle differentiation. Using primary cultures of mouse myotubes, we showed that ChR2 stimulation was well-adapted to control intracellular calcium activity at the single cell or whole culture scale. To modulate the calcium-dependent myotube differentiation, we used an optical stimulation protocol based on GCAMP6s-decoded spontaneous calcium activity patterns of differentiated myotubes. The optical training of myotubes increased the fusion index and their contractile ability. This study demonstrates that handling a mature calcium signature with such optogenetic tool improves the differentiation of primary murine myotubes.


Subject(s)
Calcium , Optogenetics , Animals , Calcium/metabolism , Cell Differentiation/physiology , Mice , Muscle Contraction , Muscle Fibers, Skeletal/metabolism
5.
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
6.
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
7.
Cells ; 9(7)2020 07 13.
Article in English | MEDLINE | ID: mdl-32668787

ABSTRACT

Anomalies in constitutive calcium entry (CCE) have been commonly attributed to cell dysfunction in pathological conditions such as cancer. Calcium influxes of this type rely on channels, such as transient receptor potential (TRP) channels, to be constitutively opened and strongly depend on membrane potential and a calcium driving force. We developed an optogenetic approach based on the expression of the halorhodopsin chloride pump to study CCE in non-excitable cells. Using C2C12 cells, we found that halorhodopsin can be used to achieve a finely tuned control of membrane polarization. Escalating the membrane polarization by incremental changes in light led to a concomitant increase in CCE through transient receptor potential vanilloid 2 (TRPV2) channels. Moreover, light-induced calcium entry through TRPV2 channels promoted cell migration. Our study shows for the first time that by modulating CCE and related physiological responses, such as cell motility, halorhodopsin serves as a potentially powerful tool that could open new avenues for the study of CCE and associated cellular behaviors.


Subject(s)
Calcium/metabolism , Cell Movement , Membrane Potentials , Optogenetics , Animals , Calcium Channels/metabolism , Cell Line , Cell Movement/radiation effects , Halorhodopsins/metabolism , Humans , Light , Membrane Potentials/radiation effects , Mice , Myoblasts/metabolism , Myoblasts/radiation effects , TRPV Cation Channels/metabolism
8.
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
9.
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
10.
Sci Rep ; 7(1): 11108, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894267

ABSTRACT

Excitation-contraction coupling in muscle cells is initiated by a restricted membrane depolarization delimited within the neuromuscular junction. This targeted depolarization triggers an action potential that propagates and induces a global cellular calcium response and a consequent contraction. To date, numerous studies have investigated this excitation-calcium response coupling by using different techniques to depolarize muscle cells. However, none of these techniques mimic the temporal and spatial resolution of membrane depolarization observed in the neuromuscular junction. By using optogenetics in C2C12 muscle cells, we developed a technique to study the calcium response following membrane depolarization induced by photostimulations of membrane surface similar or narrower than the neuromuscular junction area. These stimulations coupled to confocal calcium imaging generate a global cellular calcium response that is the consequence of a membrane depolarization propagation. In this context, this technique provides an interesting, contactless and relatively easy way of investigation of calcium increase/release as well as calcium decrease/re-uptake triggered by a propagated membrane depolarization.


Subject(s)
Calcium Signaling , Calcium/metabolism , Muscle Fibers, Skeletal/metabolism , Optogenetics , Animals , Biomarkers , Calcium Signaling/radiation effects , Cell Line , Gene Expression , Genes, Reporter , Light , Mice , Microscopy, Confocal , Muscle Fibers, Skeletal/radiation effects , Myoblasts/metabolism , Optogenetics/methods , Recombinant Fusion Proteins
11.
Prog Biophys Mol Biol ; 130(Pt B): 273-280, 2017 11.
Article in English | MEDLINE | ID: mdl-28546113

ABSTRACT

Transient receptor potential vanilloid type 2, TRPV2, is a calcium-permeable cation channel belonging to the TRPV channel family. Although this channel has been first characterized as a noxious heat sensor, its mechanosensor property recently gained importance in various physiological functions. TRPV2 has been described as a stretch-mediated channel and a regulator of calcium homeostasis in several cell types and has been shown to be involved in the stretch-dependent responses in cardiomyocytes. Hence, several studies in the last years support the idea that TRPV2 play a key role in the function and structure of the heart, being involved in the cardiac compensatory mechanisms in response to pathologic or exercise-induced stress. We present here an overview of the current literature and concepts of TRPV2 channels involvement (i) in the mechanical coupling mechanisms in heart and (ii) in the mechanisms that lead to cardiomyopathies. All these studies lead us to think that TRPV2 may also be an important cardiac drug target based on its major physiological roles in heart.


Subject(s)
Heart Diseases/metabolism , Mechanical Phenomena , Myocytes, Cardiac/metabolism , TRPV Cation Channels/metabolism , Animals , Cardiomyopathy, Dilated/metabolism , Heart/physiology , Heart/physiopathology , Heart Diseases/physiopathology , Humans
12.
EMBO J ; 35(4): 414-28, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26772186

ABSTRACT

Extracellular pH variations are seen as the principal endogenous signal that triggers activation of Acid-Sensing Ion Channels (ASICs), which are basically considered as proton sensors, and are involved in various processes associated with tissue acidification. Here, we show that human painful inflammatory exudates, displaying non-acidic pH, induce a slow constitutive activation of human ASIC3 channels. This effect is largely driven by lipids, and we identify lysophosphatidylcholine (LPC) and arachidonic acid (AA) as endogenous activators of ASIC3 in the absence of any extracellular acidification. The combination of LPC and AA evokes robust depolarizing current in DRG neurons at physiological pH 7.4, increases nociceptive C-fiber firing, and induces pain behavior in rats, effects that are all prevented by ASIC3 blockers. Lipid-induced pain is also significantly reduced in ASIC3 knockout mice. These findings open new perspectives on the roles of ASIC3 in the absence of tissue pH variation, as well as on the contribution of those channels to lipid-mediated signaling.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Arachidonic Acid/metabolism , Lysophosphatidylcholines/metabolism , Nociceptors/physiology , Animals , Cell Line , Ganglia, Spinal/cytology , Humans , Mice, Knockout , Pain , Rats
13.
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
14.
Am J Physiol Cell Physiol ; 297(5): C1133-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19692655

ABSTRACT

Evidence for a modulatory effect of cyclosporin A (CsA) on calcium signaling and cell survival in dystrophin-deficient cells is presented. Our previous works strongly supported the hypothesis of an overactivation of Ca(2+) release via inositol 1,4,5-trisphosphate (IP3) receptors (IP3R) in dystrophin-deficient cells, both during membrane depolarization and at rest, through spontaneous Ca(2+) release events. Forced expression of mini-dystrophin in these cells contributed, during stimulation and in resting condition, to the recovery of a controlled calcium homeostasis. In the present work, we demonstrate that CsA exposure displayed a dual-modulator effect on calcium signaling in dystrophin-deficient cells. Short-time incubation induced a decrease of IP3-dependent calcium release, leading to patterns of release similar to those observed in myotubes expressing mini-dystrophin, whereas long-time incubation reduced the expression of the type I of IP3 receptors (IP3R-1) RNA levels. Moreover, both IP3R-1 knockdown and blockade through 2-aminoethoxydiphenyle borate or CsA induced improved survival of dystrophin-deficient myotubes, demonstrating the cell death dependence on the IP3-dependent calcium signaling as well as the protective effect of CsA. Inhibition of the IP3 pathway could be a very interesting approach for reducing the natural cell death of dystrophin-deficient cells in development.


Subject(s)
Calcium Signaling/physiology , Dystrophin/deficiency , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Muscle Fibers, Skeletal/metabolism , Animals , Blotting, Western , Calcium Signaling/drug effects , Cell Death/drug effects , Cell Death/physiology , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression , Gene Expression Regulation/drug effects , Image Processing, Computer-Assisted , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Microscopy, Confocal , Muscle Fibers, Skeletal/drug effects , RNA, Messenger/analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
15.
FASEB J ; 21(2): 608-17, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17202249

ABSTRACT

Calcium mishandling in Duchenne dystrophic muscle suggested that dystrophin, a membrane-associated cytoskeleton protein, might regulate calcium signaling cascade such as calcium influx pathway. It was previously shown that abnormal calcium entries involve uncontrolled stretch-activated currents and store-operated Ca2+ currents supported by TRPC1 channels. Moreover, our recent work demonstrated that reintroduction of minidystrophin in dystrophic myotubes restores normal capacitative calcium entries (CCEs). However, until now, no molecular link between the dystrophin complex and calcium entry channels has been described. This study is the first to show by coimmunoprecipitation assays the molecular association of TRPC1 with dystrophin and alpha1-syntrophin in muscle cells. TRPC1 was also associated with alpha1-syntrophin in dystrophic muscle cells independently of dystrophin. Furthermore, glutathione S-transferase (GST) pull-down assays showed that TRPC1 binds to the alpha1-syntrophin PDZ domain. Transfected recombinant alpha1-syntrophin formed a complex with TRPC1 channels and restored normal CCEs in dystrophic muscle cells. We suggest that normal regulation of CCEs in skeletal muscle depends on the association between TRPC1 channels and alpha1-syntrophin that may anchor the store-operated channels to the dystrophin-associated protein complex (DAPC). The loss of this molecular association could participate in the calcium alterations observed in dystrophic muscle cells. This study provides a new model for the regulation of calcium influx by interaction with the scaffold of the DAPC in muscle cells.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Dystrophin/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , TRPC Cation Channels/metabolism , Animals , Binding Sites , Calcium-Binding Proteins/genetics , Cell Line , Cytoskeleton/metabolism , Dystrophin-Associated Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcolemma/metabolism , TRPC Cation Channels/genetics
16.
J Muscle Res Cell Motil ; 27(5-7): 375-86, 2006.
Article in English | MEDLINE | ID: mdl-16897576

ABSTRACT

Calcium mishandling in Duchenne muscular dystrophy (DMD) suggested that dystrophin, a membrane-associated cytoskeleton protein, may regulate calcium-signalling cascades such as calcium entries. Calcium overload in human DMD myotubes is dependent on their contractile activity suggesting the involvement of channels being activated during contraction and/or calcium release. Forced expression of mini-dystrophin in dystrophin-deficient myotubes, reactivates appropriate sarcolemmal expression of dystrophin-associated proteins and restores normal calcium handling in the cytosol. Furthermore, the recombinant mini-dystrophin reduced the store-operated calcium influx across the sarcolemma, and the mitochondrial calcium uptake during this influx. A slow component of calcium release dependent on IP3R, as well as the production of IP3, were also reduced to normal levels by expression of mini-dystrophin. Our studies provide a new model for the convergent regulation of transmembrane calcium influx and IP3-dependent calcium release by the dystrophin-based cytoskeleton (DBC). We also suggest molecular association of such channels with DBC which may provide the scaffold for assembling a multiprotein-signalling complex that modulates the channel activity. This suggests that the loss of this molecular association could participate in the alteration of calcium homeostasis observed in DMD muscle cells.


Subject(s)
Cytoskeleton/metabolism , Dystrophin/physiology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Humans , Inositol 1,4,5-Trisphosphate Receptors/physiology , Mitochondria/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/physiopathology
17.
J Gen Physiol ; 128(2): 219-30, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847098

ABSTRACT

We present here evidence for the enhancement, at rest, of an inositol 1,4,5-trisphosphate (IP3)-mediated calcium signaling pathway in myotubes from dystrophin-deficient cell lines (SolC1(-)) as compared to a cell line from the same origin but transfected with mini-dystrophin (SolD(+)). With confocal microscopy, the number of sites discharging calcium (release site density [RSD]) was quantified and found more elevated in SolC1(-) than in SolD(+) myotubes. Variations of membrane potential had no significant effect on this difference, and higher resting [Ca2+]i in SolC1(-) (Marchand, E., B. Constantin, H. Balghi, M.C. Claudepierre, A. Cantereau, C. Magaud, A. Mouzou, G. Raymond, S. Braun, and C. Cognard. 2004. Exp. Cell Res. 297:363-379) cannot explain alone higher RSD. The exposure with SR Ca(2+) channel inhibitors (ryanodine and 2-APB) and phospholipase C inhibitor (U73122) significantly reduced RSD in both cell types but with a stronger effect in dystrophin-deficient SolC1(-) myotubes. Immunocytochemistry allowed us to localize ryanodine receptors (RyRs) as well as IP3 receptors (IP3Rs), IP3R-1 and IP3R-2 isoforms, indicating the presence of both RyRs-dependent and IP3-dependent release systems in both cells. We previously reported evidence for the enhancement, through a Gi protein, of the IP3-mediated calcium signaling pathway in SolC1(-) as compared to SolD(+) myotubes during a high K(+) stimulation (Balghi, H., S. Sebille, B. Constantin, S. Patri, V. Thoreau, L. Mondin, E. Mok, A. Kitzis, G. Raymond, and C. Cognard. 2006. J. Gen. Physiol. 127:171-182). Here we show that, at rest, these regulation mechanisms are also involved in the modulation of calcium release activities. The enhancement of resting release activity may participate in the calcium overload observed in dystrophin-deficient myotubes, and our findings support the hypothesis of the regulatory role of mini-dystrophin on intracellular signaling.


Subject(s)
Calcium Channels/physiology , Calcium Signaling/physiology , Dystrophin/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/analysis , Cell Line , Chelating Agents/pharmacology , Cytoplasm/metabolism , Down-Regulation , Dystrophin/deficiency , Dystrophin/genetics , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Estrenes/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C3H , Mice, Knockout , Microscopy, Confocal , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nuclear Envelope/metabolism , Phosphodiesterase Inhibitors/pharmacology , Potassium/pharmacology , Pyrrolidinones/pharmacology , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism
18.
J Gen Physiol ; 127(2): 171-82, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16446505

ABSTRACT

We present here evidence for the enhancement of an inositol 1,4,5-trisphosphate (IP3) mediated calcium signaling pathway in myotubes from dystrophin-deficient cell lines (SolC1(-)) as compared to a cell line from the same origin but transfected with mini-dystrophin (SolD(+)). With confocal microscopy, we demonstrated that calcium rise, induced by the perifusion of a solution containing a high potassium concentration, was higher in SolC1(-) than in SolD(+) myotubes. The analysis of amplitude and kinetics of the calcium increase in SolC1(-) and in SolD(+) myotubes during the exposure with SR Ca2+ channel inhibitors (ryanodine and 2-APB) suggested the presence of two mechanisms of SR calcium release: (1) a fast SR calcium release that depended on ryanodine receptors and (2) a slow SR calcium release mediated by IP3 receptors. Detection analyses of mRNAs (reverse transcriptase [RT]-PCR) and proteins (Western blot and immunolocalization) demonstrated the presence of the three known isoforms of IP3 receptors in both SolC1(-) and SolD(+) myotubes. Furthermore, analysis of the kinetics of the rise in calcium revealed that the slow IP3-dependent release may be increased in the SolC1(-) as compared to the SolD(+), suggesting an inhibitory effect of mini-dystrophin in this signaling pathway. Upon incubation with pertussis toxin (PTX), an inhibitory effect similar to that of the IP3R inhibitor (2-APB) was observed on K+-evoked calcium release. This result suggests the involvement of a Gi protein upstream of the IP3 pathway in these stimulation conditions. A hypothetical model is depicted in which both Gi protein and IP3 production could be involved in K+-evoked calcium release as well as a possible interaction with mini-dystrophin. Our findings demonstrate the existence of a potential relationship between mini-dystrophin and SR calcium release as well as a regulatory role of mini-dystrophin on intracellular signaling.


Subject(s)
Calcium Signaling , Calcium/metabolism , Dystrophin/physiology , GTP-Binding Proteins/physiology , Inositol 1,4,5-Trisphosphate/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Blotting, Western , Calcium Channels/analysis , Calcium Channels/chemistry , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels/physiology , Cell Line , Down-Regulation , Dystrophin/analysis , Dystrophin/deficiency , Dystrophin/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Gene Expression , Inositol 1,4,5-Trisphosphate Receptors , Mice , Mice, Inbred C3H , Microscopy, Confocal , Pertussis Toxin/pharmacology , Potassium/pharmacology , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/physiology
19.
Mol Cell Biochem ; 229(1-2): 163-71, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11936842

ABSTRACT

The effects of extracellular Mg2+ on both dynamic changes of [Ca2+]i and apoptosis rate were analysed. The consequences of spatial and temporal dynamic changes of intracellular Ca2+ on apoptosis, in thapsigargin- and the calcium-ionophore 4BrA23187-treated MCF7 cells were first determined. Both 4BrA23187 and thapsigargin induced an instant increase of intracellular Ca2+ concentrations ([Ca2+]i) which remained quite elevated (> 150 nM) and lasted for several hours. [Ca2+]i increases were equivalent in the cytosol and the nucleus. The treatments that induced apoptosis in MCF7 cells were systematically associated with high and sustained [Ca2+]i (150 nM) for several hours. The initial [Ca2+]i increase was not determinant in the events triggering apoptosis. Thapsigargin-mediated apoptosis and [Ca2+]i rise were abrogated when cells were pretreated with the calcium chelator BAPTA. The role of the extracellular Mg2+ concentration has been studied in thapsigargin treated cells. High (10 mM) extracellular Mg2+, caused an increase in basal [Mg2+]i from 0.8+/-0.3 to 1.6+/-0.5 mM. As compared to 1.4 mM extracellular Mg2+, 1 microM thapsigargin induces, in 10 mM Mg2+, a reduced percentage from 22 to 11% of fragmented nuclei, a lower sustained [Ca2+]i and a lower Ca2+ influx through the plasma membrane. In conclusion, the cell death induced by thapsigargin was dependent on high and sustained [Ca2+]i which was inhibited by high extracellular and intracellular Mg2+.


Subject(s)
Apoptosis , Calcium/metabolism , Magnesium/pharmacology , Thapsigargin/pharmacology , Female , Humans , Microscopy, Video , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...