Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Biochem Biophys Res Commun ; 642: 83-89, 2023 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-36566566

RESUMEN

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.


Asunto(s)
Neuropatías Amiloides Familiares , Prealbúmina , Humanos , Interleucina-6 , Interleucina-8 , Neuropatías Amiloides Familiares/tratamiento farmacológico , Amiloide , Fibroblastos
2.
Pflugers Arch ; 472(12): 1719-1732, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047172

RESUMEN

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.


Asunto(s)
Fibrosis Endomiocárdica/metabolismo , Miofibroblastos/metabolismo , Canales Catiónicos TRPM/metabolismo , Potenciales de Acción , Anciano , Animales , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Miocardio/citología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/fisiología , Fenantrenos
3.
Cell Mol Biol Lett ; 25(1): 50, 2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33292162

RESUMEN

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.


Asunto(s)
Antígenos de Superficie/metabolismo , Calcio/metabolismo , Diferenciación Celular/fisiología , Canales Iónicos/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Anciano , Proliferación Celular/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Expresión Génica/fisiología , Humanos , Masculino , Células Madre Multipotentes/metabolismo , Miocitos del Músculo Liso/metabolismo
4.
Stem Cells ; 36(5): 761-774, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29359518

RESUMEN

The subventricular zone (SVZ) is the major stem cell niche in the brain of adult mammals. Within this region, neural stem cells (NSC) proliferate, self-renew and give birth to neurons and glial cells. Previous studies underlined enrichment in calcium signaling-related transcripts in adult NSC. Because of their ability to mobilize sustained calcium influxes in response to a wide range of extracellular factors, store-operated channels (SOC) appear to be, among calcium channels, relevant candidates to induce calcium signaling in NSC whose cellular activities are continuously adapted to physiological signals from the microenvironment. By Reverse Transcription Polymerase Chain Reaction (RT-PCR), Western blotting and immunocytochemistry experiments, we demonstrate that SVZ cells express molecular actors known to build up SOC, namely transient receptor potential canonical 1 (TRPC1) and Orai1, as well as their activator stromal interaction molecule 1 (STIM1). Calcium imaging reveals that SVZ cells display store-operated calcium entries. Pharmacological blockade of SOC with SKF-96365 or YM-58483 (also called BTP2) decreases proliferation, impairs self-renewal by shifting the type of SVZ stem cell division from symmetric proliferative to asymmetric, thereby reducing the stem cell population. Brain section immunostainings show that TRPC1, Orai1, and STIM1 are expressed in vivo, in SOX2-positive SVZ NSC. Injection of SKF-96365 in brain lateral ventricle diminishes SVZ cell proliferation and reduces the ability of SVZ cells to form neurospheres in vitro. The present study combining in vitro and in vivo approaches uncovers a major role for SOC in the control of SVZ NSC population and opens new fields of investigation for stem cell biology in health and disease. Stem Cells 2018;36:761-774.


Asunto(s)
Encéfalo/citología , Calcio/metabolismo , Autorrenovación de las Células/fisiología , Células-Madre Neurales/citología , Células Madre Adultas/metabolismo , Animales , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Proliferación Celular/fisiología , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Neuronas/metabolismo
5.
J Cell Physiol ; 232(4): 725-730, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27685887

RESUMEN

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.


Asunto(s)
Fibroblastos/metabolismo , Miocardio/citología , Transducción de Señal , Animales , Forma de la Célula , Fibroblastos/citología , Fibrosis , Humanos , Mecanotransducción Celular
6.
Biochim Biophys Acta ; 1850(6): 1215-23, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25721215

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Procesamiento Proteico-Postraduccional , Brefeldino A/farmacología , Glicosilación , Células HEK293 , Humanos , Potenciales de la Membrana , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Fenotipo , Transporte de Proteínas/efectos de los fármacos , Transfección
7.
Ann Noninvasive Electrocardiol ; 20(1): 28-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24943134

RESUMEN

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.


Asunto(s)
Ajmalina/administración & dosificación , Antiarrítmicos/administración & dosificación , Síndrome de Brugada/complicaciones , Síndrome de Brugada/diagnóstico , Electrocardiografía , Distrofia Miotónica/complicaciones , Adolescente , Adulto , Anciano , Síndrome de Brugada/fisiopatología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
8.
J Mol Cell Cardiol ; 68: 12-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24412532

RESUMEN

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.


Asunto(s)
Angiotensina II/fisiología , Señalización del Calcio , Canales de Cloruro/fisiología , Fibroblastos/metabolismo , Proteínas de Neoplasias/fisiología , Anciano , Anoctamina-1 , Transporte Biológico , Membrana Celular/metabolismo , Células Cultivadas , Cloruros/metabolismo , Femenino , Atrios Cardíacos/citología , Humanos , Cinética , Masculino , Receptor de Angiotensina Tipo 1/metabolismo
9.
J Gen Physiol ; 156(5)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38557788

RESUMEN

DSC1, a Drosophila channel with sequence similarity to the voltage-gated sodium channel (NaV), was identified over 20 years ago. This channel was suspected to function as a non-specific cation channel with the ability to facilitate the permeation of calcium ions (Ca2+). A honeybee channel homologous to DSC1 was recently cloned and shown to exhibit strict selectivity for Ca2+, while excluding sodium ions (Na+), thus defining a new family of Ca2+ channels, known as CaV4. In this study, we characterize CaV4, showing that it exhibits an unprecedented type of inactivation, which depends on both an IFM motif and on the permeating divalent cation, like NaV and CaV1 channels, respectively. CaV4 displays a specific pharmacology with an unusual response to the alkaloid veratrine. It also possesses an inactivation mechanism that uses the same structural domains as NaV but permeates Ca2+ ions instead. This distinctive feature may provide valuable insights into how voltage- and calcium-dependent modulation of voltage-gated Ca2+ and Na+ channels occur under conditions involving local changes in intracellular calcium concentrations. Our study underscores the unique profile of CaV4 and defines this channel as a novel class of voltage-gated Ca2+ channels.


Asunto(s)
Calcio , Canales de Sodio Activados por Voltaje , Abejas , Animales , Canales de Sodio Activados por Voltaje/química , Iones
10.
J Physiol ; 590(17): 4307-19, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22802584

RESUMEN

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.


Asunto(s)
Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Miofibroblastos/citología , Miofibroblastos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Células Cultivadas , Fenómenos Electrofisiológicos , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/química , Canal de Sodio Activado por Voltaje NAV1.5/genética , Técnicas de Placa-Clamp , Subunidades de Proteína , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
J Tradit Complement Med ; 12(4): 330-334, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35747353

RESUMEN

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.

12.
Cell Calcium ; 103: 102546, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35144094

RESUMEN

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.


Asunto(s)
Calcio , Optogenética , Animales , Calcio/metabolismo , Diferenciación Celular/fisiología , Ratones , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo
13.
Biochem Biophys Res Commun ; 408(1): 6-11, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21420933

RESUMEN

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.


Asunto(s)
Fibroblastos/fisiología , Ventrículos Cardíacos/metabolismo , Lisofosfolípidos/fisiología , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Función Ventricular/fisiología , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Gliburida/farmacología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Interleucina-6/metabolismo , Canales KATP/metabolismo , Lisofosfolípidos/farmacología , Ratones , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/farmacología , Esfingosina/fisiología , Receptores de Esfingosina-1-Fosfato , Receptores de Sulfonilureas , Función Ventricular/efectos de los fármacos
14.
Cancers (Basel) ; 13(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298643

RESUMEN

Glioblastoma is the most frequent and deadly form of primary brain tumors. Despite multimodal treatment, more than 90% of patients experience tumor recurrence. Glioblastoma contains a small population of cells, called glioblastoma stem cells (GSC) that are highly resistant to treatment and endowed with the ability to regenerate the tumor, which accounts for tumor recurrence. Transcriptomic studies disclosed an enrichment of calcium (Ca2+) signaling transcripts in GSC. In non-excitable cells, store-operated channels (SOC) represent a major route of Ca2+ influx. As SOC regulate the self-renewal of adult neural stem cells that are possible cells of origin of GSC, we analyzed the roles of SOC in cultures of GSC previously derived from five different glioblastoma surgical specimens. Immunoblotting and immunocytochemistry experiments showed that GSC express Orai1 and TRPC1, two core SOC proteins, along with their activator STIM1. Ca2+ imaging demonstrated that SOC support Ca2+ entries in GSC. Pharmacological inhibition of SOC-dependent Ca2+ entries decreased proliferation, impaired self-renewal, and reduced expression of the stem cell marker SOX2 in GSC. Our data showing the ability of SOC inhibitors to impede GSC self-renewal paves the way for a strategy to target the cells considered responsible for conveying resistance to treatment and tumor relapse.

15.
Pflugers Arch ; 460(1): 77-86, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20204400

RESUMEN

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.


Asunto(s)
Activación del Canal Iónico , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Potenciales de Acción , Línea Celular , Humanos , Canal de Sodio Activado por Voltaje NAV1.6 , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Canales de Sodio/genética , Factores de Tiempo , Transfección
16.
Cells ; 9(7)2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32668787

RESUMEN

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.


Asunto(s)
Calcio/metabolismo , Movimiento Celular , Potenciales de la Membrana , Optogenética , Animales , Canales de Calcio/metabolismo , Línea Celular , Movimiento Celular/efectos de la radiación , Halorrodopsinas/metabolismo , Humanos , Luz , Potenciales de la Membrana/efectos de la radiación , Ratones , Mioblastos/metabolismo , Mioblastos/efectos de la radiación , Canales Catiónicos TRPV/metabolismo
17.
J Mol Cell Cardiol ; 46(4): 508-17, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19166858

RESUMEN

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.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Fibroblastos/metabolismo , Ventrículos Cardíacos/citología , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Actinas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Gliburida/farmacología , Ventrículos Cardíacos/metabolismo , Interleucina-6/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Canales KATP , Lisofosfolípidos/farmacología , Ratones , Pinacidilo/farmacología , Canales de Potasio de Rectificación Interna/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Droga/genética , Esfingosina/análogos & derivados , Esfingosina/farmacología , Receptores de Sulfonilureas
18.
Aging Cell ; 18(2): e12894, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30680911

RESUMEN

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.


Asunto(s)
Envejecimiento , Modelos Biológicos , Fenoles/farmacología , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular/efectos de los fármacos , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Dieta , Relación Dosis-Respuesta a Droga , Ecocardiografía , Masculino , Estrés Oxidativo/efectos de los fármacos , Fenoles/administración & dosificación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Disfunción Ventricular Izquierda/metabolismo
19.
Biochim Biophys Acta ; 1772(8): 885-94, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17382525

RESUMEN

Cardiac hypertrophy is an adaptive process that occurs in response to increased physical stress on the heart. Hypertrophy, which may be induced by hypertension among other factors, is characterized by an increase in left ventricular mass and an associated increase in force production capacity. However, as sustained cardiac hypertrophy may lead to heart failure and sudden death, an understanding of the molecular processes involved in both the onset and consequences of hypertrophy is of significant importance. Calcium is a key player in the process underlying the development of cardiac hypertrophy. Recently, several Transient Receptor Potential proteins (TRPs), including calcium-permeable and calcium-regulated ion channels, have been shown to be related to various aspects of cardiac hypertrophy. TRPs are implicated in the development of cardiac hypertrophy (TRPC1, TRPC3, TRPC6), the electrophysiological perturbations associated with hypertrophy (TRPM4) and the progression to heart failure (TRPC7). This review describes the major characteristics of cardiac hypertrophy and focuses on the roles of TRPs in the physiological processes underlying hypertrophy.


Asunto(s)
Cardiomegalia/etiología , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Señalización del Calcio/fisiología , Cardiomegalia/genética , Cardiomegalia/metabolismo , Humanos , Potenciales de la Membrana/fisiología , Modelos Biológicos , Familia de Multigenes/fisiología , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/fisiología , Canales Catiónicos TRPM/fisiología , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
20.
J Med Liban ; 56(3): 168-73, 2008.
Artículo en Francés | MEDLINE | ID: mdl-18792554

RESUMEN

In the heart, two types of calcium currents were described, the L- and T-type. In addition to these two types, a dihydropyridine-resistant Ca2+ component has been described to be up-regulated in rat ventricular cardiomyocytes during their differentiation- dedifferentiation process. The aim of our study is to examine if such calcium current component is present in human cardiomyocytes. The patch clamp technique was used to record Ca2+ current in atrial cells. In the presence of 2 microM nifedipine, residual current was activated (-2.7 +/- 0.7 pA/pF, n = 6) in the same voltage range as the L-type, nifedipine-sensitive Ca2+ current (-2.1 +/- 0.4 pA/pF, n = 6), but its steady-state inactivation was negatively shifted by 10 mV. This nifedipine-resistant Ca2+ current was completely blocked by 500 microM cadmium chloride and significantly enhanced by 1 microM isoproterenol (-7.5 +/- 0.5 pA/pF, n = 6; p <0.01). These results give evidence that a nifedipine-resistant Ca2+ current, similar to the one which has been shown to be developmentally expressed in rat ventricular cardiomyocytes, is observed in human atrial cells. Its molecular identity, its expression level as well as its role in pathophysiologic conditions remain to be studied.


Asunto(s)
Canales de Calcio/efectos de los fármacos , Calcio/fisiología , Atrios Cardíacos/inervación , Miocitos Cardíacos/efectos de los fármacos , Cloruro de Cadmio , Dihidropiridinas/uso terapéutico , Humanos , Técnicas In Vitro , Nifedipino/uso terapéutico , Técnicas de Placa-Clamp
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda