Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
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
2.
J Tradit Complement Med ; 12(4): 330-334, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35747353

ABSTRACT

Background and aim: Hydrocotyle bonariensis Comm ex Lamm (Araliaceae) is one of these plants sufficiently exploited in traditional African medicine for its hypotensive effect. However, the pharmacological effects of those plants on cardiac functions are not well known. The potassium currents IKs and IKr, responsible for the repolarization of cardiac cell action potential, strongly influence the human cardiac rhythm. Therefore, modulators of these currents have a beneficial or undesirable medical importance in relation to cardiac arrhythmias. In order to optimize the therapeutic use of this medicinal plant, we studied the effects of hydro-ethanolic leaf extract of Hydrocotyle bonariensis on both potassium currents. Experimental procedure: The patch clamp experiments for IK currents recording were performed on the HEK 293 (Human Embryonic Kidney 293) cell line, stably transfected with either KCNQ1 and KCNE1 genes encoding the channel responsible for the "IKs" current (HEK293 IKs), or with hERG (human ether-a-go-go related gene) gene encoding "IKr" current (HEK293 IKr). Results and conclusion: This study revealed that the hydro-ethanolic leaf extract of H. bonariensis significantly inhibits the slow potassium component (IKs) without altering the fast potassium component (IKr). The extract at 0.5 mg/ml decreases IKs conductance by 24 ± 4.1% (n = 6) without modifying its activation threshold suggesting a direct blockade of the slow potassium channel. This selective action of the extract on the IKs current reflects a class III anti-arrhythmic effect.

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.
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.
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.
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
7.
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
8.
Sci Rep ; 7(1): 14199, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079724

ABSTRACT

It is generally accepted that voltage-gated Ca2+ channels, CaV, regulate Ca2+ homeostasis in excitable cells following plasma membrane depolarization. Here, we show that the Ca2+ protein α1D of CaV1.3 channel is overexpressed in colorectal cancer biopsies compared to normal tissues. Gene silencing experiments targeting α1D reduced the migration and the basal cytosolic Ca2+ concentration of HCT116 colon cancer cell line and modified the cytosolic Ca2+ oscillations induced by the sodium/calcium exchanger NCX1/3 working in its reverse mode. Interestingly, NCX1/3 regulated membrane potential of HCT116 cells only when α1D was silenced, and blocking NCX1/3 increased cytosolic Ca2+ concentration and cell migration. However, membrane depolarization did not induce an increase in intracellular Ca2+. Patch-clamp experiments clearly showed that the inward Ca2+ current was absent. Finally, flow cytometry and immunofluorescence studies showed that α1D protein was localized at the plasma membrane, in cytosol and cell nuclei. Altogether, we uncover a novel signaling pathway showing that α1D is involved in the regulation of Ca2+ homeostasis and cell migration by a mechanism independent of its plasma membrane canonical function but that involved plasma membrane Na+/Ca2+ exchanger.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Cell Movement , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Intracellular Space/metabolism , Active Transport, Cell Nucleus , Cell Membrane/metabolism , Cell Nucleus/metabolism , Colonic Neoplasms/physiopathology , Cytosol/metabolism , Electrophysiological Phenomena , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Sodium-Calcium Exchanger/metabolism
9.
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
10.
Oncotarget ; 8(23): 37681-37693, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28445150

ABSTRACT

Inwardly rectifying potassium channels (Kir), and especially the barium-sensitive Kir4.1 encoded by KCNJ10, are key regulators of glial functions. A lower expression or mislocation of Kir4.1 is detected in human brain tumors. MicroRNAs participate in the regulation of ionic channels and associated neurologic disorders. Here, we analyze effects of miR-5096 on the Kir4.1 expression and function in two glioblastoma cell lines, U87 and U251. Using whole-cell patch-clamp and western-blot analysis, we show that cell loading with miR-5096 decreases the Kir4.1 protein level and associated K+ current. Cell treatment with barium, a Kir4.1 blocker, or cell loading of miR-5096 both increase the outgrowth of filopodia in glioma cells, as observed by time-lapse microscopy. Knocking-down Kir4.1 expression by siRNA transfection similarly increased both filopodia formation and invasiveness of glioma cells as observed in Boyden chamber assay. MiR-5096 also promotes the release of extracellular vesicles by which it increases its own transfer to surrounding cells, in a Kir4.1-dependent manner in U251 but not in U87. Altogether, our results validate Kir4.1 as a miR-5096 target to promote invasion of glioblastoma cells. Our data highlight the complexity of microRNA effects and the role of K+ channels in cancer.


Subject(s)
Glioblastoma/metabolism , MicroRNAs/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Cell Movement , Cells, Cultured , Humans , Pentamidine , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/pharmacology , Transfection
11.
Biochim Biophys Acta ; 1850(6): 1215-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25721215

ABSTRACT

BACKGROUND: Like many voltage-gated sodium channels, the cardiac isoform Nav1.5 is well known as a glycoprotein which necessarily undergoes N-glycosylation processing during its transit to the plasma membrane. In some cardiac disorders, especially the Brugada syndrome (BrS), mutations in Nav1.5 encoding gene lead to intracellular retention and consequently trafficking defect of these proteins. We used two BrS mutants as tools to clarify both Nav1.5 glycosylation states and associated secretory behaviors. METHODS: Patch-clamp recordings and surface biotinylation assays of HEK293T cells expressing wild-type (WT) and/or mutant Nav1.5 proteins were performed to assess the impact of mutant co-expression on the membrane activity and localization of WT channels. Enzymatic deglycosylation assays and brefeldin A (BFA) treatments were also employed to further characterize recombinant and native Nav1.5 maturation. RESULTS: The present data demonstrate that Nav1.5 channels mainly exist as two differentially glycosylated forms. We reveal that dominant negative effects induced by BrS mutants upon WT channel current result from the abnormal surface expression of the fully-glycosylated forms exclusively. Furthermore, we show that core-glycosylated channels can be found at the surface membrane of BFA-treated or untreated cells, but obviously without generating any sodium current. CONCLUSIONS: Our findings provide evidence that native and recombinant Nav1.5 subunits are expressed as two distinct matured forms. Fully-glycosylated state of Nav1.5 seems to determine its functionality whereas core-glycosylated forms might be transported to the plasma membrane through an unconventional Golgi-independent secretory route. GENERAL SIGNIFICANCE: This work highlights that N-linked glycosylation processing would be critical for Nav1.5 membrane trafficking and function.


Subject(s)
Cell Membrane/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Protein Processing, Post-Translational , Brefeldin A/pharmacology , Glycosylation , HEK293 Cells , Humans , Membrane Potentials , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype , Protein Transport/drug effects , Transfection
12.
Ann Noninvasive Electrocardiol ; 20(1): 28-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24943134

ABSTRACT

BACKGROUND: Myotonic dystrophy type 1 (DM1) generates missplicing of the SCN5A gene, encoding the cardiac sodium channel (Nav 1.5). Brugada syndrome, which partly results from Nav 1.5 dysfunction and causes increased VF occurrence, can be unmasked by ajmaline. We aimed to investigate the response to ajmaline challenge in DM1 patients and its potential impact on their sudden cardiac death risk stratification. METHODS: Among 36 adult DM1 patients referred to our institution, electrophysiological study and ajmaline challenge were performed in 12 patients fulfilling the following criteria: (1) PR interval >200 ms or QRS duration >100 ms; (2) absence of complete left bundle branch block; (3) absence of permanent ventricular pacing; (4) absence of implantable cardioverter-defibrillator (ICD); (5) preserved left-ventricular ejection fraction >50%; and (6) absence of severe muscular impairment. Of note, DM1 patients with ajmaline-induced Brugada pattern (BrP) were screened for SCN5A. RESULTS: In all the 12 patients studied, the HV interval was <70 ms. A BrP was unmasked in three patients but none carried an SCN5A mutation. Ajmaline-induced sustained ventricular tachycardia occurred in one patient with BrP, who finally received an ICD. The other patients did not present any cardiac event during the entire follow-up (15 ± 4 months). CONCLUSION: Our study is the first to describe a high prevalence of ajmaline-induced BrP in DM1 patients. The indications, the safety, and the implications of ajmaline challenge in this particular setting need to be determined by larger prospective studies.


Subject(s)
Ajmaline/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Brugada Syndrome/complications , Brugada Syndrome/diagnosis , Electrocardiography , Myotonic Dystrophy/complications , Adolescent , Adult , Aged , Brugada Syndrome/physiopathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies
13.
Heart Rhythm ; 11(8): 1393-400, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24768612

ABSTRACT

BACKGROUND: Myotonic dystrophy type 1 (DM1), a muscular dystrophy due to CTG expansion in the DMPK gene, can cause cardiac conduction disorders and sudden death. These cardiac manifestations are similar to those observed in loss-of-function SCN5A mutations, which are also responsible for Brugada syndrome (BrS). OBJECTIVE: The purpose of this study was to investigate DM1 effects on clinical expression of a loss-of-function SCN5A mutation causing BrS. METHODS: We performed complete clinical evaluation, including ajmaline test, in 1 family combining DM1 and BrS. We screened the known BrS susceptibility genes. We characterized an SCN5A mutation using whole-cell patch-clamp experiments associated with cell surface biotinylation. RESULTS: The proband, a 15-year-old female, was a survivor of out-of-hospital cardiac arrest with ventricular fibrillation. She combined a DMPK CTG expansion from the father's side and an SCN5A mutation (S910L) from the mother's side. S910L is a trafficking defective mutant inducing a dominant negative effect when transfected with wild-type Nav1.5. This loss-of-function SCN5A mutation caused a Brugada phenotype during the mother's ajmaline test. Surprisingly, in the father, a DM1 patient without SCN5A mutation, ajmaline also unmasked a Brugada phenotype. Furthermore, association of both genetic abnormalities in the proband exacerbated the response to ajmaline with a massive conduction defect. CONCLUSION: Our study is the first to describe the deleterious effect of DM1 on clinical expression of a loss-of-function SCN5A mutation and to show a provoked BrS phenotype in a DM1 patient. The modification of the ECG pattern by ajmaline supports the hypothesis of a link between DM1 and Nav1.5 loss of -function.


Subject(s)
Brugada Syndrome/genetics , Mutation, Missense , Myotonic Dystrophy/diagnosis , NAV1.5 Voltage-Gated Sodium Channel/genetics , Adolescent , Brugada Syndrome/diagnosis , Brugada Syndrome/metabolism , Cells, Cultured , DNA Mutational Analysis , Electrocardiography , Female , Genetic Predisposition to Disease , Genotype , Humans , Myotonic Dystrophy/genetics , Myotonic Dystrophy/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Pedigree , Phenotype
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.
Leuk Res ; 37(8): 948-55, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23726264

ABSTRACT

Cytarabine combined with an anthracycline or an anthracenedione represents the usual intensive induction therapy for the treatment of AML. However, this protocol induces severe side effects and treatment-related mortality due to the lack of selectivity of these cytotoxic agents. In this paper, we present the study of the first galactosidase-responsive molecular "Trojan Horse" programmed for the delivery of doxorubicin exclusively inside AML blasts over-expressing the folate receptor (FR). This targeting system allows the selective killing of AML blasts without affecting normal endothelial, cardiac or hematologic cells from healthy donors suggesting that FDC could reduce adverse events usually recorded with anthracyclines.


Subject(s)
Cell Proliferation/drug effects , Doxorubicin/pharmacology , Neoplastic Stem Cells/drug effects , beta-Galactosidase/metabolism , Acute Disease , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Blast Crisis/drug therapy , Blast Crisis/metabolism , Blast Crisis/pathology , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Delivery Systems/methods , Female , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folate Receptor 2/genetics , Folate Receptor 2/metabolism , Folic Acid/chemistry , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Male , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Young Adult
16.
Am J Physiol Cell Physiol ; 304(9): C881-94, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23426965

ABSTRACT

Calcium homeostasis is critical for several vital functions in excitable and nonexcitable cells and has been shown to be impaired in many pathologies including Duchenne muscular dystrophy (DMD). Various studies using murine models showed the implication of calcium entry in the dystrophic phenotype. However, alteration of store-operated calcium entry (SOCE) and transient receptor potential vanilloid 2 (TRPV2)-dependant cation entry has not been investigated yet in human skeletal muscle cells. We pharmacologically characterized basal and store-operated cation entries in primary cultures of myotubes prepared from muscle of normal and DMD patients and found, for the first time, an increased SOCE in DMD myotubes. Moreover, this increase cannot be explained by an over expression of the well-known SOCE actors: TRPC1/4, Orai1, and stromal interaction molecule 1 (STIM1) mRNA and proteins. Thus we investigated the modes of regulation of this cation entry. We firstly demonstrated the important role of the scaffolding protein α1-syntrophin, which regulates SOCE in primary human myotubes through its PDZ domain. We also studied the implication of phospholipase C (PLC) and protein kinase C (PKC) in SOCE and showed that their inhibition restores normal levels of SOCE in DMD human myotubes. In addition, the involvement of TRPV2 in calcium deregulation in DMD human myotubes was explored. We showed an abnormal elevation of TRPV2-dependant cation entry in dystrophic primary human myotubes compared with normal ones. These findings show that calcium homeostasis mishandling in DMD myotubes depends on SOCE under the influence of Ca(2+)/PLC/PKC pathway and α1-syntrophin regulation as well as on TRPV2-dependant cation influx.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , TRPV Cation Channels/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Channels/metabolism , Cells, Cultured , Estrenes/pharmacology , Gadolinium/pharmacology , Gene Expression , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Maleimides/pharmacology , Membrane Proteins/genetics , Muscular Dystrophy, Duchenne/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nifedipine/pharmacology , ORAI1 Protein , Patch-Clamp Techniques , Primary Cell Culture , Protein Kinase C/metabolism , Pyrrolidinones/pharmacology , Sarcoplasmic Reticulum/metabolism , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Type C Phospholipases/metabolism
17.
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
18.
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
19.
PLoS One ; 7(5): e38331, 2012.
Article in English | MEDLINE | ID: mdl-22675453

ABSTRACT

Cardiac Na(+) channels encoded by the SCN5A gene are essential for initiating heart beats and maintaining a regular heart rhythm. Mutations in these channels have recently been associated with atrial fibrillation, ventricular arrhythmias, conduction disorders, and dilated cardiomyopathy (DCM).We investigated a young male patient with a mixed phenotype composed of documented conduction disorder, atrial flutter, and ventricular tachycardia associated with DCM. Further family screening revealed DCM in the patient's mother and sister and in three of the mother's sisters. Because of the complex clinical phenotypes, we screened SCN5A and identified a novel mutation, R219H, which is located on a highly conserved region on the fourth helix of the voltage sensor domain of Na(v)1.5. Three family members with DCM carried the R219H mutation.The wild-type (WT) and mutant Na(+) channels were expressed in a heterologous expression system, and intracellular pH (pHi) was measured using a pH-sensitive electrode. The biophysical characterization of the mutant channel revealed an unexpected selective proton leak with no effect on its biophysical properties. The H(+) leak through the mutated Na(v)1.5 channel was not related to the Na(+) permeation pathway but occurred through an alternative pore, most probably a proton wire on the voltage sensor domain.We propose that acidification of cardiac myocytes and/or downstream events may cause the DCM phenotype and other electrical problems in affected family members. The identification of this clinically significant H(+) leak may lead to the development of more targeted treatments.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Dilated/physiopathology , Protons , Sodium Channels/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/genetics , Base Sequence , Cardiomyopathy, Dilated/genetics , Cell Line , Humans , Hydrogen-Ion Concentration , Male , Molecular Sequence Data , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Oocytes/metabolism , Pedigree , Phenotype , Sodium Channels/genetics , Xenopus
20.
Pflugers Arch ; 460(1): 77-86, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20204400

ABSTRACT

Na(v)1.6 is the major voltage-gated sodium channel at nodes of Ranvier. This channel has been shown to produce a robust persistent inward current in whole-cell experiments. Na(v)1.6 plays an important role in axonal conduction and may significantly contribute to the pathophysiology of the injured nervous system through this persistent current. However, the underlying molecular mechanisms and regulation of the persistent current are not well understood. Using the whole-cell configuration of the patch-clamp technique, we investigated the Na(v)1.6 transient and persistent currents in HEK-293. Previous studies have shown that the persistent current depended on the content of the patch electrode. Therefore, we characterised the single-channel properties of the persistent current with an intact intracellular medium using the cell-attached configuration of the patch-clamp technique. In HEK-293 cells, the Na(v)1.6 persistent current recorded in the whole-cell configuration was 3-5% of the peak transient current. In single-channel recording, the ratio between peak and persistent open probability confirmed the magnitude of the persistent current observed in the whole-cell configuration. The cell-attached configuration revealed that the molecular mechanism of the whole-cell persistent current is a consequence of single Na(v)1.6 channels reopening.


Subject(s)
Ion Channel Gating , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Action Potentials , Cell Line , Humans , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Sodium Channels/genetics , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...