Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cell Calcium ; 123: 102925, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38908063

RESUMEN

AIMS: Previous studies have identified RyR2 W4645R mutation, located in the caffeine-binding site, to associate with CPVT1 pathology. Caffeine binding to its site is thought to displace the carboxyl-terminal domain to Ca2+-binding, allowing the tryptophan residue (W4645) to regulate Ca2+ sensitivity of RyR2. To gain insights into regulation of RyR2 Ca2+-binding and its interaction with caffeine-binding site, we introduced W4645R-RyR2 point mutation via CRISPR/Cas9 gene-editing in human induced pluripotent stem cell-derived cardiomyocytes (hiPSCCMs) and characterized their Ca2+-signaling phenotype compared to WT hiPSCCMs. METHODS AND RESULTS: W4645R-RyR2 cardiomyocytes had: (1) no significant change in ICa magnitude or voltage-dependence; (2) slightly reduced CICR; (3) altered relaxation kinetics of Ca2+-transients with no change in isoproterenol sensitivity; (4) complete loss of caffeine-triggered Ca2+ release; (5) larger SR Ca2+ leak resulting in 40 % lower SR Ca2+ content, as determined by myocytes' response to 4-CmC; (6) lower incidence of calcium sparks and asynchronous spontaneous SR Ca2+ releases. CONCLUSIONS: W4645R-RyR2 mutation induces loss of caffeine-triggered SR Ca2+ release and enhances SR Ca2+ leak that underlie asynchronous spontaneous Ca2+ releases, triggering arrhythmia and impairing cardiac function.

2.
Cardiovasc Res ; 120(1): 44-55, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-37890099

RESUMEN

AIMS: CRISPR/Cas9 gene edits of cardiac ryanodine receptor (RyR2) in human-induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) provide a novel platform for introducing mutations in RyR2 Ca2+-binding residues and examining the resulting excitation contraction (EC)-coupling remodelling consequences. METHODS AND RESULTS: Ca2+-signalling phenotypes of mutations in RyR2 Ca2+-binding site residues associated with cardiac arrhythmia (RyR2-Q3925E) or not proven to cause cardiac pathology (RyR2-E3848A) were determined using ICa- and caffeine-triggered Ca2+ releases in voltage-clamped and total internal reflection fluorescence-imaged wild type and mutant cardiomyocytes infected with sarcoplasmic reticulum (SR)-targeted ER-GCaMP6 probe. (i) ICa- and caffeine-triggered Fura-2 or ER-GCaMP6 signals were suppressed, even when ICa was significantly enhanced in Q3925E and E3848A mutant cardiomyocytes; (ii) spontaneous beating (Fura-2 Ca2+ transients) persisted in mutant cells without the SR-release signals; (iii) while 5-20 mM caffeine failed to trigger Ca2+-release in voltage-clamped mutant cells, only ∼20% to ∼70% of intact myocytes responded respectively to caffeine; (iv) and 20 mM caffeine transients, however, activated slowly, were delayed, and variably suppressed by 2-APB, FCCP, or ruthenium red. CONCLUSION: Mutating RyR2 Ca2+-binding residues, irrespective of their reported pathogenesis, suppressed both ICa- and caffeine-triggered Ca2+ releases, suggesting interaction between Ca2+- and caffeine-binding sites. Enhanced transmembrane calcium influx and remodelling of EC-coupling pathways may underlie the persistence of spontaneous beating in Ca2+-induced Ca2+ release-suppressed mutant myocytes.


Asunto(s)
Miocitos Cardíacos , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Cafeína/farmacología , Cafeína/metabolismo , Calcio/metabolismo , Fura-2/metabolismo , Miocitos Cardíacos/metabolismo , Mutación Puntual , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
3.
Int J Mol Sci ; 24(20)2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37894987

RESUMEN

Type-2 ryanodine receptor (RyR2) is the major Ca2+ release channel of the cardiac sarcoplasmic reticulum (SR) that controls the rhythm and strength of the heartbeat, but its malfunction may generate severe arrhythmia leading to sudden cardiac death or heart failure. S4938F-RyR2 mutation in the carboxyl-terminal was expressed in human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) using CRISPR/Cas9 gene-editing technique. Ca2+ signaling and electrophysiological properties of beating cardiomyocytes carrying the mutation were studied using total internal reflection fluorescence microscopy (TIRF) and patch clamp technique. In mutant cells, L-type Ca2+ currents (ICa), measured either by depolarizations to zero mV or repolarizations from +100 mV to -50 mV, and their activated Ca2+ transients were significantly smaller, despite their larger caffeine-triggered Ca2+ release signals compared to wild type (WT) cells, suggesting ICa-induced Ca2+ release (CICR) was compromised. The larger SR Ca2+ content of S4938F-RyR2 cells may underlie the higher frequency of spontaneously occurring Ca2+ sparks and Ca2+ transients and their arrhythmogenic phenotype.


Asunto(s)
Señalización del Calcio , Células Madre Pluripotentes Inducidas , Humanos , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
4.
Methods Mol Biol ; 2573: 41-52, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36040585

RESUMEN

Human-induced pluripotent stem cells (hiPSCs) provide a powerful platform to study biophysical and molecular mechanisms underlying the pathophysiology of genetic mutations associated with cardiac arrhythmia. Human iPSCs can be generated by reprograming of dermal fibroblasts of normal or diseased individuals and be differentiated into cardiac myocytes. Obtaining biopsies from patients afflicted with point mutations causing arrhythmia is often a cumbersome process even when patients are available. Recent development of CRISPR/Cas9 gene editing system makes it, however, possible to introduce arrhythmia-associated point mutations at the desired loci of the wild-type hiPSCs in relatively short times. This platform was used by us to compare the Ca2+ signaling phenotypes of cardiomyocytes harboring point mutations in cardiac Ca2+ release channel, type-2 ryanodine receptor (RyR2), since over 200 missense mutations in RYR2 gene appear to be associated with catecholaminergic polymorphic ventricular tachycardia (CPVT1). We have created cardiac myocytes harboring mutations in different domains of RyR2, to study not only their Ca2+ signaling consequences but also their drug and domain specificity as related to CPVT1 pathology. In this chapter, we describe our procedures to establish CRISPR/Cas9 gene-edited hiPSC-derived cardiomyocytes.


Asunto(s)
Células Madre Pluripotentes Inducidas , Canal Liberador de Calcio Receptor de Rianodina , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Sistemas CRISPR-Cas , Calcio/metabolismo , Edición Génica/métodos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Taquicardia Ventricular
5.
Can J Physiol Pharmacol ; 100(9): 848-857, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35679617

RESUMEN

How oxygen is sensed by the heart and what mechanisms mediate its sensing remain poorly understood. As recent reports show that low PO2 levels are detected by the cardiomyocytes in a few seconds, the rapid and short applications of low levels of oxygen (acute hypoxia), which avoid multiple effects of chronic hypoxia, may be used to probe the oxygen-sensing pathway of the heart. Here, we explored the oxygen-sensing pathway, focusing primarily on cellular surface membrane proteins that were first exposed to low PO2. Such studies suggest that acute hypoxia primarily targets the cardiac calcium channels, where either the channel itself or moieties closely associated with it, for instance heme-oxygenase-2 (HO-2) interacting through kinase phosphorylation, signal the α-subunit of the channel to the altered levels of PO2. Amino acids 1572-1651, the CaMKII phosphorylation sites (S1487 and S1545), CaM-binding sites (I1624 and Q1625), and Ser1928 of the carboxyl tail of the α-subunit appear to be critical residues that sense oxygen. Future studies on HO-2 knockout mice or CRISPR/Cas9 gene-edited human-induced pluripotent stem-cell-derived cardiomyocytes that reduce CaM-binding affinity are likely to provide deeper insights into the O2-sensing mechanisms.


Asunto(s)
Hipoxia , Oxígeno , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Hipoxia/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Fosforilación
6.
Cell Calcium ; 101: 102500, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34813985

RESUMEN

AIMS: To gain insights into FKBP regulation of cardiac ryanodine receptor (RyR2) and Ca2+ signaling, we introduced the point mutation (N771D-RyR2) corresponding to skeletal muscle mutation (N760D-RyR1) associated with central core disease (CCD) via CRISPR/Cas9 gene-editing in the RyR2 FKBP binding site expressed in human induced pluripotent stem cell-derived cardiomyocytes (hiPSCCMs). Patients inflicted with CCD and other hereditary skeletal muscle diseases often show higher incidence of atrial or ventricular arrhythmias. METHODS AND RESULTS: Ca2+ imaging of voltage-clamped N771D-RyR2 mutant compared to WT hiPSCCMs showed: (1) ∼30% suppressed ICa with no significant changes in the gating kinetics of ICa; (2) 29% lower SR Ca2+ content and 33% lower RyR2 Ca2+ leak; (3) higher CICR gain and 30-35% increased efficiency of ICa-triggered Ca2±release; (4) higher incidence of aberrant SR Ca2+ releases, DADs, and Ca2+ sparks; (5) no change in fractional Ca2+-release, action potential morphology, sensitivity to isoproterenol, and sarcomeric FKBP-binding pattern. CONCLUSIONS: The more frequent spontaneous Ca2+ releases and longer Ca2+ sparks underlie the increased incidence of DADs and cellular arrhythmogenesis of N771D-RyR2 mutant. The smaller RyR2 Ca2±leak and SR content result from suppressed ICathat is compensated by higher CICR gain.


Asunto(s)
Señalización del Calcio , Células Madre Pluripotentes Inducidas , Canal Liberador de Calcio Receptor de Rianodina , Sitios de Unión , Calcio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
7.
Heart Rhythm ; 18(2): 250-260, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32931925

RESUMEN

BACKGROUND: Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) created from patients with catecholaminergic polymorphic ventricular tachycardia 1 (CPVT1) have been used to study CPVT1 arrhythmia. OBJECTIVE: The purpose of this study was to evaluate the Ca2+ signaling aberrancies and pharmacological sensitivities of 3 CRISPR/Cas9-introduced CPVT1 mutations located in different molecular domains of ryanodine receptor 2 (RyR2). METHODS: CRISPR/Cas9-engineered hiPSC-CMs carrying RyR2 mutations-R420Q, Q4201R, and F2483I-were voltage clamped, and their electrophysiology, pharmacology, and Ca2+ signaling phenotypes measured using total internal reflection fluorescence microscopy. RESULTS: R420Q and Q4201R mutant hiPSC-CMs exhibit irregular, long-lasting, spatially wandering Ca2+ sparks and aberrant Ca2+ releases similar to F2483I unlike the wild-type myocytes. Large sarcoplasmic reticulum (SR) Ca2+ leaks and smaller SR Ca2+ contents were detected in cells expressing Q4201R and F2483I, but not R420Q. Fractional Ca2+ release and calcium-induced calcium release gain were higher in Q4201R than in R420Q and F2483I hiPSC-CMs. JTV519 was equally effective in suppressing Ca2+ sparks, waves, and SR Ca2+ leaks in hiPSC-CMs derived from all 3 mutant lines. Flecainide and dantrolene similarly suppressed SR Ca2+ leaks, but were less effective in decreasing spark frequency and durations. CONCLUSION: CRISPR/Cas9 gene editing of hiPSCs provides a novel approach in studying CPVT1-associated RyR2 mutations and suggests that Ca2+-signaling aberrancies and drug sensitivities may vary depending on the mutation site.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/genética , Proteína 9 Asociada a CRISPR/metabolismo , Calcio/metabolismo , Señalización del Calcio , Diferenciación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , ADN/genética , Análisis Mutacional de ADN , Humanos , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/patología , Fenotipo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología
8.
Biol Cell ; 113(3): 133-145, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33275284

RESUMEN

BACKGROUND INFORMATION: Wnt/ß-catenin signalling, in the microenvironment of pluripotent stem cells (PSCs), plays a critical role in their differentiation and proliferation. Contradictory reports on the role of Wnt/ß-catenin signalling in PSCs self-renewal and differentiation, however, render these mechanisms largely unclear. RESULTS: Wnt/ß-catenin signalling pathway in human-induced pluripotent stem cells (hiPSCs) was activated by inhibiting glycogen synthase kinase 3 (GSK3), driving the cells into a mesodermal/mesenchymal state, exhibiting proliferative, invasive and anchorage-independent growth properties, where over 70% of cell population became CD 44 (+)/CD133 (+). Wnt/ß-catenin signalling activation also altered the metabolic state of hiPSCs from aerobic glycolysis to oxidative metabolism and changed their drug and oxidative stress sensitivities. These effects of GSK3 inhibition were suppressed in HIF1α-stabilised cells. CONCLUSIONS: Persistent activation of Wnt/ß-catenin signalling endows hiPSCs with proliferative/invasive 'teratoma-like' states, shifting their metabolic dependence and allowing HIF1α-stabilisation to inhibit their proliferative/invasive properties. SIGNIFICANCE: The hiPSC potential to differentiate into 'teratoma-like' cells suggest that stem cells may exist in two states with differential metabolic and drug dependency.


Asunto(s)
Células Madre Pluripotentes Inducidas , Vía de Señalización Wnt/fisiología , Diferenciación Celular , Línea Celular , Proliferación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo
9.
Cell Calcium ; 90: 102244, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32585508

RESUMEN

Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) have been extensively used for in vitro modeling of human cardiovascular disease, drug screening and pharmacotherapy, but little rigorous studies have been reported on their biophysical or Ca2+ signaling properties. There is also considerable concern as to the level of their maturity and whether they can serve as reliable models for adult human cardiac myocytes. Ultrastructural difference such as lack of t-tubular network, their polygonal shapes, disorganized sarcomeric myofilament, and their rhythmic automaticity, among others, have been cited as evidence for immaturity of hiPSC-CMs. In this review, we will deal with Ca2+ signaling, its regulation, and its stage of maturity as compared to the mammalian adult cardiomyocytes. We shall summarize the data on functional aspects of Ca2+signaling and its parameters that include: L-type calcium channel (Cav1.2), ICa-induced Ca2+release, CICR, and its parameters, cardiac Na/Ca exchanger (NCX1), the ryanodine receptors (RyR2), sarco-reticular Ca2+pump, SERCA2a/PLB, and the contribution of mitochondrial Ca2+ to hiPSC-CMs excitation-contraction (EC)-coupling as compared with adult mammalian cardiomyocytes. The comparative studies suggest that qualitatively hiPSC-CMs have similar Ca2+signaling properties as those of adult cardiomyocytes, but quantitative differences do exist. This review, we hope, will allow the readers to judge for themselves to what extent Ca2+signaling of hiPSC-CMs represents the adult form of this signaling pathway, and whether these cells can be used as good models of human cardiomyocytes.


Asunto(s)
Señalización del Calcio , Células Madre Pluripotentes Inducidas/citología , Mamíferos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Adulto , Animales , Diferenciación Celular , Humanos , Modelos Biológicos
10.
Stem Cells Dev ; 28(14): 920-932, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31119982

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are potential sources for cardiac regeneration and drug development. hiPSC-CMs express all the cardiac ion channels and the unique cardiac Ca2+-signaling phenotype. In this study, we tested for expression of acid sensing ion channels (ASICs) in spontaneously beating cardiomyocytes derived from three different hiPSC lines (IMR-90, iPSC-K3, and Ukki011-A). Rapid application of solutions buffered at pH 6.7, 6.0, or 5.0 triggered rapidly activating and slowly inactivating voltage-independent inward current that reversed at voltages positive to ENa, was suppressed by 5 µM amiloride and withdrawal of [Na+]o, like neuronal ASIC currents. ASIC currents were expressed at much lower percentages and densities in undifferentiated hiPSC and in dermal fibroblasts. ASIC1 mRNA and protein were measured in first 60 days but decreased in 100 days postdifferentiation hiPSC cultures. Hyperacidification (pH 5 and 6) also triggered large Ca2+ transients in intact hiPSC-CMs that were neither ruthenium red nor amiloride-sensitive, but were absent in whole cell-clamped hiPSC-CMs. Neither ASIC1 current nor its protein was detected in rat adult cardiomyocytes, but hyperacidification did activate smaller and slowly activating currents with drug sensitivity similar to TRPV channels. Considering ASIC expression in developing but not adult myocardium, a role in heart development is likely.


Asunto(s)
Canales Iónicos Sensibles al Ácido/biosíntesis , Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Línea Celular Tumoral , Dermis/citología , Dermis/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología
11.
Cell Calcium ; 78: 1-14, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30579812

RESUMEN

AIMS: The effects of acute (100 s) hypoxia and/or acidosis on Ca2+ signaling parameters of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are explored here for the first time. METHODS AND RESULTS: 1) hiPSC-CMs express two cell populations: rapidly-inactivating ICa myocytes (τi<40 ms, in 4-5 day cultures) and slowly-inactivating ICa (τi ≥ 40 ms, in 6-8 day cultures). 2) Hypoxia suppressed ICa by 10-20% in rapidly- and 40-55% in slowly-inactivating ICa cells. 3) Isoproterenol enhanced ICa in hiPSC-CMs, but either enhanced or did not alter the hypoxic suppression. 4) Hypoxia had no differential suppressive effects in the two cell-types when Ba2+ was the charge carrier through the calcium channels, implicating Ca2+-dependent inactivation in O2 sensing. 5) Acidosis suppressed ICa by ∼35% and ∼25% in rapidly and slowly inactivating ICa cells, respectively. 6) Hypoxia and acidosis suppressive effects on Ca-transients depended on whether global or RyR2-microdomain were measured: with acidosis suppression was ∼25% in global and ∼37% in RyR2 Ca2+-microdomains in either cell type, whereas with hypoxia suppression was ∼20% and ∼25% respectively in global and RyR2-microdomaine in rapidly and ∼35% and ∼45% respectively in global and RyR2-microdomaine in slowly-inactivating cells. CONCLUSIONS: Variability in ICa inactivation kinetics rather than cellular ancestry seems to underlie the action potential morphology differences generally attributed to mixed atrial and ventricular cell populations in hiPSC-CMs cultures. The differential hypoxic regulation of Ca2+-signaling in the two-cell types arises from differential Ca2+-dependent inactivation of the Ca2+-channel caused by proximity of Ca2+-release stores to the Ca2+ channels.


Asunto(s)
Acidosis/metabolismo , Calcio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Señalización del Calcio , Hipoxia de la Célula , Células Cultivadas , Humanos
12.
Cell Calcium ; 73: 104-111, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29730419

RESUMEN

Type-2 ryanodine receptors (RyR2s) play a pivotal role in cardiac excitation-contraction coupling by releasing Ca2+ from sarcoplasmic reticulum (SR) via a Ca2+ -induced Ca2+ release (CICR) mechanism. Two strategies have been used to study the structure-function characteristics of RyR2 and its disease associated mutations: (1) heterologous cell expression of the recombinant mutant RyR2s, and (2) knock-in mouse models harboring RyR2 point mutations. Here, we establish an alternative approach where Ca2+ signaling aberrancy caused by the RyR2 mutation is studied in human cardiomyocytes with robust CICR mechanism. Specifically, we introduce point mutations in wild-type RYR2 of human induced pluripotent stem cells (hiPSCs) by CRISPR/Cas9 gene editing, and then differentiate them into cardiomyocytes. To verify the reliability of this approach, we introduced the same disease-associated RyR2 mutation, F2483I, which was studied by us in hiPSC-derived cardiomyocytes (hiPSC-CMs) from a patient biopsy. The gene-edited F2483I hiPSC-CMs exhibited longer and wandering Ca2+ sparks, elevated diastolic Ca2+ leaks, and smaller SR Ca2+ stores, like those of patient-derived cells. Our CRISPR/Cas9 gene editing approach validated the feasibility of creating myocytes expressing the various RyR2 mutants, making comparative mechanistic analysis and pharmacotherapeutic approaches for RyR2 pathologies possible.


Asunto(s)
Sistemas CRISPR-Cas/fisiología , Señalización del Calcio/fisiología , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/fisiología , Canal Liberador de Calcio Receptor de Rianodina/genética , Secuencia de Bases , Diferenciación Celular/fisiología , Humanos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
13.
J Mol Cell Cardiol ; 114: 58-71, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29032102

RESUMEN

Ischemic heart disease is an arrhythmogenic condition, accompanied by hypoxia, acidosis, and impaired Ca2+ signaling. Here we report on effects of acute hypoxia and acidification in rat neonatal cardiomyocytes cultures. RESULTS: Two populations of neonatal cardiomyocyte were identified based on inactivation kinetics of L-type ICa: rapidly-inactivating ICa (τ~20ms) myocytes (prevalent in 3-4-day cultures), and slow-inactivating ICa (τ≥40ms) myocytes (dominant in 7-day cultures). Acute hypoxia (pO2<5mmHg for 50-100s) suppressed ICa reversibly in both cell-types to different extent and with different kinetics. This disparity disappeared when Ba2+ was the channel charge carrier, or when the intracellular Ca2+ buffering capacity was increased by dialysis of high concentrations of EGTA and BAPTA, suggesting critical role for calcium-dependent inactivation. Suppressive effect of acute acidosis on ICa (~40%, pH6.7), on the other hand, was not cell-type dependent. Isoproterenol enhanced ICa in both cell-types, but protected only against suppressive effects of acidosis and not hypoxia. Hypoxia and acidosis suppressed global Ca2+ transients by ~20%, but suppression was larger, ~35%, at the RyR2 microdomains, using GCaMP6-FKBP targeted probe. Hypoxia and acidosis also suppressed mitochondrial Ca2+ uptake by 40% and 10%, respectively, using mitochondrial targeted Ca2+ biosensor (mito-GCaMP6). CONCLUSION: Our studies suggest that acute hypoxia suppresses ICa in rapidly inactivating cell population by a mechanism involving Ca2+-dependent inactivation, while compromised mitochondrial Ca2+ uptake seems also to contribute to ICa suppression in slowly inactivating cell population. Proximity of cellular Ca2+ pools to sarcolemmal Ca2+ channels may contribute to the variability of inactivation kinetics of ICa in the two cell populations, while acidosis suppression of ICa appears mediated by proton-induced block of the calcium channel.


Asunto(s)
Acidosis/patología , Señalización del Calcio , Hipoxia/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Acidosis/metabolismo , Enfermedad Aguda , Animales , Animales Recién Nacidos , Bario/metabolismo , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Concentración de Iones de Hidrógeno , Hipoxia/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Isoproterenol/farmacología , Cinética , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factores de Tiempo
14.
Can J Physiol Pharmacol ; 95(10): 1100-1107, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28350969

RESUMEN

The sinoatrial (SA) node is the primary site from which the mammalian heart is paced, but the mechanisms underlying the pacemaking still remain clouded. It is generally believed that the hyperpolarization-activated current If, encoded by hyperpolarization-activated cyclic nucleotide-gated (HCN) genes, contributes significantly to pacing, which in tandem with inward current generated by efflux of Ca2+ via the Na+-Ca2+ exchanger (NCX), resulting from the released Ca2+, mediates the diastolic depolarization. Here, we review the data that implicate If as the "pacemaker current" and conclude that there is not only a significant discrepancy between the range of diastolic depolarization potential (-60 to -40 mV) and the activation potential of If (negative to -70 mV), but that also the kinetics of If and its pharmacology are incompatible with the frequency of a heartbeat in rodents and humans. We propose that If serves as a functional insulator, which protects the SA-nodal cells against the large negative electrical sink of atrial tissue connected to it with connexins. We also evaluate the role of If and calcium signaling in mediating the diastolic depolarization in rat neonatal cardiomyocytes (rN-CM), and human induced pluripotent stem-cell derived cardiomyocytes (hiPSC-CM), and provide evidence for a possible involvement of mitochondrial Ca2+ in initiating the oscillatory events required for the spontaneous pacing.


Asunto(s)
Relojes Biológicos , Frecuencia Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Nodo Sinoatrial/metabolismo , Potenciales de Acción , Animales , Señalización del Calcio , Diferenciación Celular , Conexinas/metabolismo , Humanos , Mitocondrias Cardíacas/metabolismo , Modelos Cardiovasculares , Ratas , Nodo Sinoatrial/citología , Factores de Tiempo
15.
J Pharmacol Exp Ther ; 360(1): 239-248, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27815365

RESUMEN

Cardiac arrhythmia is a major cause of mortality in cardiovascular pathologies. A host of drugs targeted to sarcolemmal Na+, Ca2+, and K+ channels has had limited success clinically. Recently, Ca2+ signaling has been target of pharmacotherapy based on finding that leaky ryanodine receptors elevate local Ca2+ concentrations causing membrane depolarizations that trigger arrhythmias. In this study, we report that xanthohumol, an antioxidant extracted from hops showing therapeutic effects in other pathologies, suppresses aberrant ryanodine receptor Ca2+ release. The effects of xanthohumol (5-1000 nM) on Ca2+ signaling pathways were probed in isolated rat ventricular myocytes incubated with Fluo-4 AM using the perforated patch-clamp technique. We found that 5-50 nM xanthohumol reduced the frequency of spontaneously occurring Ca2+ sparks (>threefold) and Ca2+ waves in control myocytes and in cells subjected to Ca2+ overload caused by the following: 1) exposure to low K+ solutions, 2) periods of high frequency electrical stimulation, 3) exposures to isoproterenol, or 4) caffeine. At room temperatures, 50-100 nM xanthohumol reduced the rate of relaxation of electrically- or caffeine-triggered Ca2+transients, without suppressing ICa, but this effect was small and reversed by isoproterenol at physiologic temperatures. Xanthohumol also suppressed the Ca2+ content of the SR and its rate of recirculation. The stabilizing effects of xanthohumol on the frequency of spontaneously triggered Ca2+ sparks and waves combined with its antioxidant properties, and lack of significant effects on Na+ and Ca2+ channels, may provide this compound with clinically desirable antiarrhythmic properties.


Asunto(s)
Antiarrítmicos/farmacología , Señalización del Calcio/efectos de los fármacos , Flavonoides/farmacología , Ventrículos Cardíacos/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Propiofenonas/farmacología , Animales , Antiarrítmicos/metabolismo , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Flavonoides/metabolismo , Isoproterenol/farmacología , Masculino , Miocitos Cardíacos/metabolismo , Propiofenonas/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Temperatura
16.
Cell Calcium ; 59(2-3): 98-107, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26725479

RESUMEN

Derivation of cardiomyocyte cell lines from human fibroblasts (induced pluripotent stem cells, iPSCs) has made it possible not only to investigate the electrophysiological and Ca(2+) signaling properties of these cells, but also to determine the altered electrophysiological and Ca(2+)-signaling profiles of such cells lines derived from patients expressing mutation-inducing pathologies. This approach has the potential of generating in vitro human models of cardiovascular diseases where cellular pathology can be investigated in detail and possibly specific pharmacotherapy developed. Although this approach has been applied to a number of mutations in channel proteins that cause arrhythmias, there are only few detailed reports addressing Ca(2+) signaling pathologies beyond measurements of Ca(2+) transients in intact non-voltage clamped cells. Unfortunately, full understanding of Ca(2+) signaling pathologies remains elusive, not only because of the plethora of Ca(2+) signaling proteins defects that cause arrhythmias and cardiomyopathies, but also because detailed functional properties of Ca(2+) signaling proteins are difficult to obtain. Catecholaminergic polymorphic ventricular tachycardia (CPVT1) is a malignant inherited arrhythmogenic disorder predominantly caused by mutations in the cardiac ryanodine receptor (RyR2). Thus far over 150 mutations in RyR2 have been identified that appear to cause this arrhythmia, a number of which have been expressed and studied in transgenic mice or cell-line models. The development of human iPSC-technology makes it possible to create human heart cell-lines carrying these mutations, making detailed identification of Ca(2+) signaling defects and its specific pharmacotherapy possible. In this review we shall first briefly summarize the essential characteristics of the mammalian cardiac Ca(2+) signaling, then compare them to Ca(2+) signaling phenotypes of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) and to those of rat neonatal cardiomyocytes, and categorize the possible variance in Ca(2+) signaling defects caused by different CPVT-inducing mutations as expressed in hiPSC-CMs.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Taquicardia Ventricular/metabolismo , Humanos , Taquicardia Ventricular/patología
17.
Sci Rep ; 5: 15978, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26527405

RESUMEN

Faithful reporting of temporal patterns of intracellular Ca(2+) dynamics requires the working range of indicators to match the signals. Current genetically encoded calmodulin-based fluorescent indicators are likely to distort fast Ca(2+) signals by apparent saturation and integration due to their limiting fluorescence rise and decay kinetics. A series of probes was engineered with a range of Ca(2+) affinities and accelerated kinetics by weakening the Ca(2+)-calmodulin-peptide interactions. At 37 °C, the GCaMP3-derived probe termed GCaMP3fast is 40-fold faster than GCaMP3 with Ca(2+) decay and rise times, t1/2, of 3.3 ms and 0.9 ms, respectively, making it the fastest to-date. GCaMP3fast revealed discreet transients with significantly faster Ca(2+) dynamics in neonatal cardiac myocytes than GCaMP6f. With 5-fold increased two-photon fluorescence cross-section for Ca(2+) at 940 nm, GCaMP3fast is suitable for deep tissue studies. The green fluorescent protein serves as a reporter providing important novel insights into the kinetic mechanism of target recognition by calmodulin. Our strategy to match the probe to the signal by tuning the affinity and hence the Ca(2+) kinetics of the indicator is applicable to the emerging new generations of calmodulin-based probes.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Señalización del Calcio , Calmodulina/genética , Células Cultivadas , Fluorescencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Espacio Intracelular/metabolismo , Cinética , Microscopía Confocal , Mutación , Miocitos Cardíacos/citología , Ratas
18.
Cell Calcium ; 57(5-6): 321-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25746147

RESUMEN

The quintessential property of developing cardiomyocytes is their ability to beat spontaneously. The mechanisms underlying spontaneous beating in developing cardiomyocytes are thought to resemble those of adult heart, but have not been directly tested. Contributions of sarcoplasmic and mitochondrial Ca(2+)-signaling vs. If-channel in initiating spontaneous beating were tested in human induced Pluripotent Stem cell-derived cardiomyocytes (hiPS-CM) and rat Neonatal cardiomyocytes (rN-CM). Whole-cell and perforated-patch voltage-clamping and 2-D confocal imaging showed: (1) both cell types beat spontaneously (60-140/min, at 24°C); (2) holding potentials between -70 and 0mV had no significant effects on spontaneous pacing, but suppressed action potential formation; (3) spontaneous pacing at -50mV activated cytosolic Ca(2+)-transients, accompanied by in-phase inward current oscillations that were suppressed by Na(+)-Ca(2+)-exchanger (NCX)- and ryanodine receptor (RyR2)-blockers, but not by Ca(2+)- and If-channels blockers; (4) spreading fluorescence images of cytosolic Ca(2+)-transients emanated repeatedly from preferred central cellular locations during spontaneous beating; (5) mitochondrial un-coupler, FCCP at non-depolarizing concentrations (∼50nM), reversibly suppressed spontaneous pacing; (6) genetically encoded mitochondrial Ca(2+)-biosensor (mitycam-E31Q) detected regionally diverse, and FCCP-sensitive mitochondrial Ca(2+)-uptake and release signals activating during INCX oscillations; (7) If-channel was absent in rN-CM, but activated only negative to -80mV in hiPS-CM; nevertheless blockers of If-channel failed to alter spontaneous pacing.


Asunto(s)
Señalización del Calcio/fisiología , Frecuencia Cardíaca/fisiología , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Línea Celular , Células Cultivadas , Humanos , Ratones , Modelos Animales , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Ratas , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Intercambiador de Sodio-Calcio/efectos de los fármacos , Intercambiador de Sodio-Calcio/fisiología
19.
Cell Calcium ; 56(3): 133-46, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24994483

RESUMEN

I(Ca)-gated Ca(2+) release (CICR) from the cardiac SR is the main mechanism mediating the rise of cytosolic Ca(2+), but the extent to which mitochondria contribute to the overall Ca(2+) signaling remains controversial. To examine the possible role of mitochondria in Ca(2+) signaling, we developed a low affinity mitochondrial Ca(2+) probe, mitycam-E31Q (300-500 MOI, 48-72h) and used it in conjunction with Fura-2AM to obtain simultaneous TIRF images of mitochondrial and cytosolic Ca(2+) in cultured neonatal rat cardiomyocytes. Mitycam-E31Q staining of adult feline cardiomyocytes showed the typical mitochondrial longitudinal fluorescent bandings similar to that of TMRE staining, while neonatal rat cardiomyocytes had a disorganized tubular or punctuate appearance. Caffeine puffs produced rapid increases in cytosolic Ca(2+) while simultaneously measured global mitycam-E31Q signals decreased more slowly (increased mitochondrial Ca(2+)) before decaying to baseline levels. Similar, but oscillating mitycam-E31Q signals were seen in spontaneously pacing cells. Withdrawal of Na(+) increased global cytosolic and mitochondrial Ca(2+) signals in one population of mitochondria, but unexpectedly decreased it (release of Ca(2+)) in another mitochondrial population. Such mitochondrial Ca(2+) release signals were seen not only during long lasting Na(+) withdrawal, but also when Ca(2+) loaded cells were exposed to caffeine-puffs, and during spontaneous rhythmic beating. Thus, mitochondrial Ca(2+) transients appear to activate with a delay following the cytosolic rise of Ca(2+) and show diversity in subpopulations of mitochondria that could contribute to the plasticity of mitochondrial Ca(2+) signaling.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Calmodulina/genética , Fura-2/análogos & derivados , Mitocondrias Cardíacas/metabolismo , Sondas Moleculares , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Citosol/metabolismo , Fluorescencia , Células HEK293 , Humanos , Sondas Moleculares/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal
20.
Psychopharmacology (Berl) ; 231(9): 2009-18, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24241791

RESUMEN

RATIONALE: Large conductance calcium-activated potassium (BK(Ca) or K(Ca)1.1) channels are well-known molecular targets for the action of alcohol and therefore may play an important role in the pathogenesis of alcohol withdrawal syndrome. OBJECTIVES: We evaluate the modifications of total outward K⁺ currents and protein expression of BK(Ca) channels α-subunit in inferior colliculus (IC) neurons obtained from controls and rats subjected to alcohol withdrawal associated with enhanced susceptibility to seizures. METHODS: Outward K⁺ currents and BK(Ca) channel proteins were measured using the whole cell configuration of patch clamp techniques and Western blot analysis, respectively. RESULTS: Total outward K⁺ current density was significantly reduced in IC neurons at 24 and 48 h during the alcohol withdrawal period when the susceptibility to seizures was maximal and absent, respectively. The iberiotoxin-sensitive (BK(Ca)) current density and conductance also were significantly reduced at 24 h following alcohol withdrawal. Consistent with functional data, the levels of protein expression of α-subunit associated with BK(Ca) channels also was significantly reduced in IC neurons at 24 and 48 h following alcohol withdrawal. CONCLUSIONS: The downregulation of BK(Ca) channels outlasts the finite period of elevated susceptibility to alcohol withdrawal seizures. These findings indicate that BK(Ca) channels, per se, may not be fundamentally important for the generation of alcohol withdrawal seizures.


Asunto(s)
Trastornos Relacionados con Alcohol/metabolismo , Colículos Inferiores/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Neuronas/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Convulsiones por Abstinencia de Alcohol/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Depresores del Sistema Nervioso Central/metabolismo , Regulación hacia Abajo , Etanol/farmacología , Colículos Inferiores/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Potasio/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...