Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
J Mol Cell Cardiol ; 170: 1-14, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35644481

RESUMO

Mutations of the RyR2 are channelopathies that can predispose to life threatening catecholaminergic polymorphic ventricular tachycardias (CPVTs) during exercise or stress. However, the cellular and molecular mechanisms that are causal for the arrhythmias downstream of the ß-adrenergic receptor (ß-AR) activation are not defined. They may be specific and different for each particular RyR2 mutation. Obvious possibilities are the phosphorylation of the mutated RyR2s or the stimulation of the SR Ca2+ pump (SERCA), which could increase SR Ca2+ loading. Potentially arrhythmogenic Ca2+ signals, such as Ca2+ waves, were recorded and analyzed from WT and RyR2R420Q+/- mouse cardiomyocytes with confocal microscopy after field stimulation at 1 Hz. In RyR2R420Q+/- cardiomyocytes we found a higher occurrence and frequency of Ca2+ waves, particularly upon ß-AR stimulation with isoproterenol. This was accompanied by a shorter latency to the first spontaneous wave. Wave velocity from raw traces, as well as amplitude and decay time constant (τ) analyzed in de-skewed traces were comparable in both cell types. To obtain further insight into the role of the SERCA we selectively stimulated SERCA in permeabilized myocytes using Fab fragments of a PLB antibody (2D12). Surprisingly, SERCA stimulation alone resulted in considerably higher wave frequencies than when mimicking ß-AR stimulation with cAMP, particularly in RyR2R420Q+/- cardiomyocytes. This may be a consequence of some protective SR Ca2+ unloading resulting from the SR Ca2+ leak via phosphorylated RyR2s in cAMP. Spark-to-spark recovery analysis suggested a remarkably higher Ca2+ release sensitivity in RyR2R420Q+/- cells, both in control and upon ß-AR stimulation. Together these findings suggest that the fine balance between SR Ca2+ loading via SERCA and the Ca2+ leak via mutated and phosphorylated RyR2s is an important determinant for the overall cellular arrhythmogenicity prevailing in the RyR2R420Q+/- myocytes.


Assuntos
Miócitos Cardíacos , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Isoproterenol/farmacologia , Camundongos , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
2.
Chem Commun (Camb) ; 58(17): 2826-2829, 2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35112125

RESUMO

Monitoring and manipulation of ionized intracellular calcium concentrations within intact, living cells using optical probes with organic chromophores is a core method for cell physiology. Since all these probes have multiple negative charges, they must be smuggled through the plasma membrane in a transiently neutral form, with intracellular esterases used to deprotect the masked anions. Here we explore the ability of the synthetically easily accessible n-butyl ester protecting group to deliver amphipathic cargoes to the cytosol. We show that the size of the caging chromophore conditions the ability of intracellular probe delivery and esterase charge unmasking.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Esterases/metabolismo , Corantes Fluorescentes/metabolismo , Miócitos Cardíacos/metabolismo , Cálcio/química , Membrana Celular/química , Citosol/química , Esterases/química , Corantes Fluorescentes/química , Humanos , Estrutura Molecular , Miócitos Cardíacos/química , Tamanho da Partícula
3.
J Physiol ; 599(16): 3841-3852, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34245001

RESUMO

KEY POINTS: In cardiac myocytes, subcellular local calcium release signals, calcium sparks, are recruited to form each cellular calcium transient and activate the contractile machinery. Abnormal timing of recovery of sparks after their termination may contribute to arrhythmias. We developed a method to interrogate recovery of calcium spark trigger probabilities and their amplitude over time using two-photon photolysis of a new ultra-effective caged calcium compound. The findings confirm the utility of the technique to define an elevated sensitivity of the calcium release mechanism in situ and to follow hastened recovery of spark trigger probabilities in a mouse model of an inherited cardiac arrhythmia, which was used for validation. Analogous methods are likely to be applicable to investigate other microscopic subcellular signalling systems in a variety of cell types. ABSTRACT: In cardiac myocytes Ca2+ -induced Ca2+ release (CICR) from the sarcoplasmic reticulum (SR) through ryanodine receptors (RyRs) governs activation of contraction. Ca2+ release occurs via subcellular Ca2+ signalling events, Ca2+ sparks. Local recovery of Ca2+ release depends on both SR refilling and restoration of Ca2+ sensitivity of the RyRs. We used two-photon (2P) photolysis of the ultra-effective caged Ca2+ compound BIST-2EGTA and laser-scanning confocal Ca2+ imaging to probe refractoriness of local Ca2+ release in control conditions and in the presence of cAMP or low-dose caffeine (to stimulate CICR) or cyclopiazonic acid (CPA; to slow SR refilling). Permeabilized cardiomyocytes were loaded with BIST-2EGTA and rhod-2. Pairs of short 2P photolytic pulses (1 ms, 810 nm) were applied with different intervals to test Ca2+ release amplitude recovery and trigger probability for the second spark in a pair. Photolytic and biological events were distinguished by classification with a self-learning support vector machine (SVM) algorithm. In permeabilized myocytes data recorded in the presence of CPA showed a lower probability of triggering a second spark compared to control or cAMP conditions. Cardiomyocytes from a mouse model harbouring the arrhythmogenic RyRR420Q mutation were used for further validation and revealed a higher Ca2+ sensitivity of CICR. This new 2P approach provides composite information of Ca2+ release amplitude and trigger probability recovery reflecting both SR refilling and restoration of CICR and RyR Ca2+ sensitivity. It can be used to measure the kinetics of local CICR recovery, alterations of which may be related to premature heart beats and arrhythmias.


Assuntos
Cálcio , Retículo Sarcoplasmático , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Camundongos , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
6.
J Physiol ; 598(6): 1131-1150, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943206

RESUMO

KEY POINTS: Increased protein phosphatase 1 (PP-1) activity has been found in end stage human heart failure. Although PP-1 has been extensively studied, a detailed understanding of its role in the excitation-contraction coupling mechanism, in normal and diseased hearts, remains elusive. The present study investigates the functional effect of the PP-1 activity on local Ca2+ release events in ventricular cardiomyocytes, by using an activating peptide (PDP3) for the stimulation of the endogenous PP-1 protein. We report that acute de-phosphorylation may increase the sensitivity of RyR2 channels to Ca2+ in situ, and that the RyR2-serine2808 phosphorylation site may mediate such a process. Our approach unmasks the functional importance of PP-1 in the regulation of RyR2 activity, suggesting a potential role in the generation of a pathophysiological sarcoplasmic reticulum Ca2+ leak in the diseased heart. ABSTRACT: Changes in cardiac ryanodine receptor (RyR2) phosphorylation are considered to be important regulatory and disease related post-translational protein modifications. The extent of RyR2 phosphorylation is mainly determined by the balance of the activities of protein kinases and phosphatases, respectively. Increased protein phosphatase-1 (PP-1) activity has been observed in heart failure, although the regulatory role of this enzyme on intracellular Ca2+ handling remains poorly understood. To determine the physiological and pathophysiological significance of increased PP-1 activity, we investigated how the PP-1 catalytic subunit (PP-1c) alters Ca2+ sparks in permeabilized cardiomyocytes and we also applied a PP-1-disrupting peptide (PDP3) to specifically activate endogenous PP-1, including the one anchored on the RyR2 macromolecular complex. We compared wild-type and transgenic mice in which the usually highly phosphorylated site RyR2-S2808 has been ablated to investigate its involvement in RyR2 modulation (S2808A+/+ ). In wild-type myocytes, PP-1 increased Ca2+ spark frequency by two-fold, followed by depletion of the sarcoplasmic reticulum Ca2+ store. Similarly, PDP3 transiently increased spark frequency and decreased sarcoplasmic reticulum Ca2+ load. RyR2 Ca2+ sensitivity, which was assessed by Ca2+ spark recovery analysis, was increased in the presence of PDP3 compared to a negative control peptide. S2808A+/+ cardiomyocytes did not respond to both PP-1c and PDP3 treatment. Our results suggest an increased Ca2+ sensitivity of RyR2 upon de-phosphorylation by PP-1. Furthermore, we have confirmed the S2808 site as a target for PP-1 and as a potential link between RyR2s modulation and the cellular response.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Miócitos Cardíacos , Proteína Fosfatase 1/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
7.
Methods Mol Biol ; 1929: 53-71, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30710267

RESUMO

An increase in the cytosolic Ca2+ concentration triggers the contraction in cardiomyocytes. In these cells sarcoplasmic reticulum (SR) is the major source of Ca2+, and the release from this store is mediated by the ryanodine receptors (RyRs). These receptors are regulated by cytosolic and intra-SR [Ca2+]. The cytosolic Ca2+ regulation is well established, but there are some limitations to determine indirectly the intra-SR Ca2+ concentration and understand its role in the RyRs regulation. Therefore, the interest to directly measure the free intra-SR Ca2+ concentration ([Ca2+]SR) has led to the application of a low-affinity Ca2+ indicator (Fluo-5N AM) to follow changes of [Ca2+]SR in cardiomyocytes. However the loading of this AM-ester dye into the SR has remained a challenge in freshly isolated mouse cardiomyocytes. Here, we describe an optimized protocol to measure changes of [Ca2+]SR in mouse cardiomyocytes using fluorescent Ca2+ indicators and confocal microscopy. The application of this protocol allows to evaluate directly intra-SR Ca2+ in real time in various mouse models of cardiac disease, including transgenic animals harboring mutants of RyRs or other Ca2+ signaling proteins.


Assuntos
Cálcio/análise , Miócitos Cardíacos/citologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , Animais , Sinalização do Cálcio , Retículo Endoplasmático/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Mutação , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
8.
Biophys J ; 116(3): 383-394, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30638961

RESUMO

Analysis of Ca2+ signals obtained in various cell types (i.e., cardiomyocytes) is always a tradeoff between acquisition speed and signal/noise ratio of the fluorescence signal. This becomes especially apparent during fast two- or three-dimensional confocal imaging when local intracellular fluorescence signals originating from Ca2+ release from intracellular Ca2+ stores (e.g., sarcoplasmic reticulum) need to be examined. Mathematical methods have been developed to remedy a high noise level by fitting each pixel with a transient function to "denoise" the image. So far, current available analytical approaches have been impaired by a number of constraints (e.g., inability to fit local, concurrent, and consecutive events) and the limited ability to customize implementation. Here, we suggest a, to our knowledge, novel approach for detailed analysis of subcellular micro-Ca2+ events based on pixel-by-pixel denoising of confocal frame- and line-scan images. The algorithm enables spatiotemporally overlapping events (e.g., a Ca2+ spark occurring during the decaying phase of a Ca2+ wave) to be extracted so that various types of Ca2+ events can be detected at a pixel time level of precision. The method allows a nonconstant baseline to be estimated for each pixel, foregoing the need to subtract fluorescence background or apply self-ratio methods before image analysis. Furthermore, by using a clustering algorithm, identified single-pixel events are grouped into "physiologically relevant" Ca2+ signaling events spanning multiple pixels (sparks, waves, puffs, transients, etc.), from which spatiotemporal event parameters (e.g., full duration at half maximal amplitude, full width at half maximal amplitude, amplitude, wave speed, rise, and decay times) can be easily extracted. The method was implemented with cross-platform open source software, providing a comprehensive and easy-to-use graphical user interface enabling rapid line-scan images and rapid frame-scan image sequences (up to 150 frames/s) to be analyzed and repetitive Ca2+ events (Ca2+ sparks and Ca2+ puffs) originating from clusters of Ca2+ release channels located in the sarcoplasmic reticulum membrane (ryanodine receptors and inositol 1,4,5-trisphosphate receptors) of isolated cardiomyocytes to be examined with a high level of precision.


Assuntos
Cálcio/metabolismo , Microscopia Confocal , Algoritmos , Animais , Automação , Sinalização do Cálcio , Citosol/metabolismo , Processamento de Imagem Assistida por Computador , Masculino , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo
9.
J Gen Physiol ; 151(2): 131-145, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30541771

RESUMO

During physical exercise or stress, the sympathetic system stimulates cardiac contractility via ß-adrenergic receptor (ß-AR) activation, resulting in protein kinase A (PKA)-mediated phosphorylation of the cardiac ryanodine receptor RyR2. PKA-dependent "hyperphosphorylation" of the RyR2 channel has been proposed as a major impairment that contributes to progression of heart failure. However, the sites of PKA phosphorylation and their phosphorylation status in cardiac diseases are not well defined. Among the known RyR2 phosphorylation sites, serine 2030 (S2030) remains highly controversial as a site of functional impact. We examined the contribution of RyR2-S2030 to Ca2+ signaling and excitation-contraction coupling (ECC) in a transgenic mouse with an ablated RyR2-S2030 phosphorylation site (RyR2-S2030A+/+). We assessed ECC gain by using whole-cell patch-clamp recordings and confocal Ca2+ imaging during ß-ARs stimulation with isoproterenol (Iso) and consistent SR Ca2+ loading and L-type Ca2+ current (I Ca) triggering. Under these conditions, ECC gain is diminished in mutant compared with WT cardiomyocytes. Resting Ca2+ spark frequency (CaSpF) with Iso is also reduced by mutation of S2030. In permeabilized cells, when SR Ca2+ pump activity is kept constant (using 2D12 antibody against phospholamban), cAMP does not change CaSpF in S2030A+/+ myocytes. Using Ca2+ spark recovery analysis, we found that mutant RyR Ca2+ sensitivity is not enhanced by Iso application, contrary to WT RyRs. Furthermore, ablation of RyR2-S2030 prevents acceleration of Ca2+ waves and increases latency to the first spontaneous Ca2+ release after a train of stimulations during Iso treatment. Together, these results suggest that phosphorylation at S2030 may represent an important step in the modulation of RyR2 activity during ß-adrenergic stimulation and a potential target for the development of new antiarrhythmic drugs.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Isoproterenol/farmacologia , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Serina/metabolismo , Animais , Sinalização do Cálcio , Células Cultivadas , Acoplamento Excitação-Contração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/química
10.
J Mol Cell Cardiol ; 119: 87-95, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29715473

RESUMO

AIMS: In cardiac muscle, phosphorylation of the RyRs is proposed to increase their Ca2+ sensitivity. This mechanism could be arrhythmogenic via facilitation of spontaneous Ca2+ waves. Surprisingly, the level of Ca2+ inside the SR needed to initiate such waves has been reported to increase upon ß-adrenergic stimulation, an observation which cannot be easily reconciled with elevated Ca2+ sensitivity of the RyRs. We tested the hypothesis that this change of Ca2+ wave threshold could occur indirectly, subsequent to SERCA stimulation. METHODS AND RESULTS: Cytosolic and intra-SR Ca2+ waves were simultaneously recorded with confocal line-scan imaging in intact and permeabilized mouse cardiomyocytes using Rhod-2 and Fluo-5-N, respectively. We analyzed changes of several Ca2+ signaling parameters during specific SERCA stimulation by ochratoxin A (OTA), jasmonate or the Fab fragment of a phospholamban antibody. SERCA stimulation resulted in a substantial increase of the threshold for Ca2+ wave initiation. Faster Ca2+ transient decay and SR refilling confirmed SERCA acceleration. CONCLUSIONS: These results suggest that isolated SERCA stimulation can elevate the intra-SR threshold for the generation of Ca2+ waves, independently of RyR phosphorylation. Simultaneously, fractional Ca2+ release and wave amplitudes are reduced. Thus, SERCA stimulation appears to exert a negative feed-back on the Ca2+-induced Ca2+ release mechanisms sustaining the waves. Thereby, it may be profoundly antiarrhythmic. This may be clinically relevant when therapies are applied to stimulate the SERCA activity (e.g. SERCA overexpression with gene therapy, future small molecule SERCA stimulators).


Assuntos
Sinalização do Cálcio/genética , Miocárdio/enzimologia , Miócitos Cardíacos/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ocratoxinas/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo
11.
Biochemistry ; 57(26): 3976-3986, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29791142

RESUMO

The human zinc transporter SLC39A2, also known as ZIP2, was shown to mediate zinc transport that could be inhibited at pH <7.0 and stimulated by HCO3-, suggesting a Zn2+/HCO3- cotransport mechanism [Gaither, L. A., and Eide, D. J. (2000) J. Biol. Chem. 275, 5560-5564]. In contrast, recent experiments in our laboratory indicated that the functional activity of ZIP2 increases at acidic pH [Franz, M. C., et al. (2014) J. Biomol. Screening 19, 909-916]. The study presented here was therefore designed to reexamine the findings about the pH dependence and to extend the functional characterization of ZIP2. Our current results show that ZIP2-mediated transport is modulated by extracellular pH but independent of the H+ driving force. Also, in our experiments, ZIP2-mediated transport is not modulated by extracellular HCO3-. Moreover, a high extracellular [K+], which induces depolarization, inhibited ZIP2-mediated transport, indicating that the transport mechanism is voltage-dependent. We also show that ZIP2 mediates the uptake of Cd2+ ( Km ∼ 1.57 µM) in a pH-dependent manner ( KH+ ∼ 66 nM). Cd2+ transport is inhibited by extracellular [Zn2+] (IC50 ∼ 0.32 µM), [Cu2+] (IC50 ∼ 1.81 µM), and to a lesser extent [Co2+], but not by [Mn2+] or [Ba2+]. Fe2+ is not transported by ZIP2. Accordingly, the substrate selectivity of ZIP2 decreases in the following order: Zn2+ > Cd2+ ≥ Cu2+ > Co2+. Altogether, we propose that ZIP2 is a facilitated divalent metal ion transporter that can be modulated by extracellular pH and membrane potential. Given that ZIP2 expression has been reported in acidic environments [Desouki, M. M., et al. (2007) Mol. Cancer 6, 37; Inoue, Y., et al. (2014) J. Biol. Chem. 289, 21451-21462; Tao, Y. T., et al. (2013) Mol. Biol. Rep. 40, 4979-4984], we suggest that the herein described H+-mediated regulatory mechanism might be important for determining the velocity and direction of the transport process.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Bicarbonatos/metabolismo , Células HEK293 , Humanos , Transporte de Íons/fisiologia , Metais
12.
BMC Complement Altern Med ; 17(1): 117, 2017 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-28219356

RESUMO

BACKGROUND: This study explored Bacopa monnieri, a medicinal Ayurvedic herb, as a cardioprotectant against ischemia/reperfusion injury using cardiac function and coronary flow as end-points. METHODS: In normal isolated rat hearts, coronary flow, left ventricular developed pressure, heart rate, and functional recovery were measured using the Langendorff preparation. Hearts were perfused with either (i) Krebs-Henseleit (normal) solution, (control), or with 30, 100 µg/ml B. monnieri ethanolic extract (30 min), or (ii) with normal solution or extract for 10 min preceding no-perfusion ischemia (30 min) followed by reperfusion (30 min) with normal solution. Infarct volumes were measured by triphenyltetrazolium staining. L-type Ca2+-currents (ICa, L) were measured by whole-cell patching in HL-1 cells, a mouse atrial cardiomyocyte cell line. Cytotoxicity of B. monnieri was assessed in rat isolated ventricular myocytes by trypan blue exclusion. RESULTS: In normally perfused hearts, B. monnieri increased coronary flow by 63 ± 13% (30 µg/ml) and 216 ± 21% (100 µg/ml), compared to control (5 ± 3%) (n = 8-10, p < 0.001). B. monnieri treatment preceding ischemia/reperfusion improved left ventricular developed pressure by 84 ± 10% (30 µg/ml), 82 ± 10% (100 µg/ml) and 52 ± 6% (control) compared to pre- ischemia/reperfusion. Similarly, functional recovery showed a sustained increase. Moreover, B. monnieri (100 µg/ml) reduced the percentage of infarct size from 51 ± 2% (control) to 25 ± 2% (n = 6-8, p < 0.0001). B. monnieri (100 µg/ml) reduced ICa, L by 63 ± 4% in HL-1 cells. Ventricular myocyte survival decreased at higher concentrations (50-1000 µg/ml) B. monnieri. CONCLUSIONS: B. monnieri improves myocardial function following ischemia/reperfusion injury through recovery of coronary blood flow, contractile force and decrease in infarct size. Thus this may lead to a novel cardioprotectant strategy.


Assuntos
Bacopa , Coração/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico , Fluxo Sanguíneo Regional/efeitos dos fármacos , Animais , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiopatologia , Coração/fisiopatologia , Frequência Cardíaca , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Técnicas In Vitro , Masculino , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/patologia , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Ratos Wistar , Pressão Ventricular
13.
Cell Calcium ; 60(5): 331-340, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27431464

RESUMO

To initiate the contraction of cardiomyocytes, Ca2+ is released from the SR to the cytosol via ryanodine receptors (RyRs), which are activated by the Ca2+-induced Ca2+ release mechanism (CICR). The activity of RyRs is regulated by both, cytosolic and SR luminal Ca2+. Deregulation of the CICR, by dysfunctional SR Ca2+ release or uptake, is frequently associated with cardiac pathologies (e.g. arrhythmias, CPVT, heart failure). Recently, the interest to directly measure changes of the free Ca2+ concentration within the SR ([Ca2+]SR) has led to the application of low affinity Ca2+ indicators (mag-fluo-4, Fluo-5N) to follow changes of [Ca2+]SR in cardiomyocytes from some species. However, direct measurement of Ca2+ signals from the SR have not been possible in freshly isolated mouse cardiomyocytes. Here, we show a new protocol optimized to measure changes of [Ca2+]SR in mouse cardiomyocytes using fluorescent Ca2+ indicators and confocal microscopy. The application of this protocol permits the design of experimental studies with direct evaluation of SR Ca2+ in real time in various mouse models of cardiac disease, including transgenic animals harboring mutants of RyRs or other Ca2+ signaling proteins. The technique, in combination with these models, will help to understand how these diseases and mutations affect Ca2+ signals within the SR and the Ca2+ sensitivity of the RyRs for cytosolic and SR luminal Ca2+, thereby contributing to arrhythmias or weak heart beat.


Assuntos
Cálcio/análise , Miócitos Cardíacos/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Camundongos , Microscopia Confocal , Miócitos Cardíacos/citologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
14.
J Am Chem Soc ; 138(11): 3687-93, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-26974387

RESUMO

We have designed a nitroaromatic photochemical protecting group that absorbs visible light in the violet-blue range. The chromophore is a dinitro derivative of bisstyrylthiophene (or BIST) that absorbs light very effectively (ε440 = 66,000 M(-1) cm(-1) and two-photon cross section of 350 GM at 775 nm). We developed a "caged calcium" molecule by conjugation of BIST to a Ca(2+) chelator that upon laser flash photolysis rapidly releases Ca(2+) in <0.2 ms. Using the patch-clamp method the optical probe, loaded with Ca(2+), was delivered into acutely isolated mouse cardiac myocytes, where either one- and two-photon uncaging of Ca(2+) induced highly local or cell-wide physiological Ca(2+) signaling events.


Assuntos
Compostos de Cálcio/química , Tiofenos/química , Compostos de Cálcio/síntese química , Quelantes/química , Ácido Egtázico/química , Luz , Processos Fotoquímicos , Tiofenos/síntese química
15.
JACC Basic Transl Sci ; 1(6): 472-493, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29707678

RESUMO

The mechanisms controlling differentiation in adult cardiac precursor cells (CPCs) are still largely unknown. In this study, CPCs isolated from the human heart were found to produce predominantly smooth muscle cells but could be redirected to the cardiomyocyte fate by transient activation followed by inhibition of NOTCH signaling. NOTCH inhibition repressed MIR-143/145 expression, and blocked smooth muscle differentiation. Expression of the microRNAs is under control of CARMEN, a long noncoding RNA associated with an enhancer located in the MIR-143/145 locus and target of NOTCH signaling. The CARMEN/MIR-145/143 axis represents, therefore, a promising target to favor production of cardiomyocytes in cell replacement therapies.

17.
J Physiol ; 593(6): 1495-507, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25772298

RESUMO

KEY POINTS: Refractoriness of calcium release in heart cells is altered in several disease states, but the physiological mechanisms that regulate this process are incompletely understood. We examined refractoriness of calcium release in mouse ventricular myocytes and investigated how activation of different intracellular signalling pathways influenced this process. We found that refractoriness of calcium release is abbreviated by stimulation of the 'fight-or-flight' response, and that simultaneous activation of multiple intracellular signalling pathways contributes to this response. Data obtained under several conditions at the subcellular, microscopic level were consistent with results obtained at the cellular level. The results provide insight into regulation of cardiac calcium release and how alterations to this process may increase arrhythmia risk under different conditions. ABSTRACT: Time-dependent refractoriness of calcium (Ca(2+)) release in cardiac myocytes is an important factor in determining whether pro-arrhythmic release patterns develop. At the subcellular level of the Ca(2+) spark, recent studies have suggested that recovery of spark amplitude is controlled by local sarcoplasmic reticulum (SR) refilling whereas refractoriness of spark triggering depends on both refilling and the sensitivity of the ryanodine receptor (RyR) release channels that produce sparks. Here we studied regulation of Ca(2+) spark refractoriness in mouse ventricular myocytes by examining how ß-adrenergic stimulation influenced sequences of Ca(2+) sparks originating from individual RyR clusters. Our protocol allowed us to separately measure recovery of spark amplitude and delays between successive sparks, and data were interpreted quantitatively through simulations with a stochastic mathematical model. We found that, compared with spark sequences measured under control conditions: (1) ß-adrenergic stimulation with isoproterenol (isoprenaline) accelerated spark amplitude recovery and decreased spark-to-spark delays; (2) activating protein kinase A (PKA) with forskolin accelerated amplitude recovery but did not affect spark-to-spark delays; (3) inhibiting PKA with H89 retarded amplitude recovery and increased spark-to-spark delays; (4) preventing phosphorylation of the RyR at serine 2808 with a knock-in mouse prevented the decrease in spark-to-spark delays seen with ß-adrenergic stimulation; (5) inhibiting either PKA or Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) during ß-adrenergic stimulation prevented the decrease in spark-to-spark delays seen without inhibition. The results suggest that activation of either PKA or CaMKII is sufficient to speed SR refilling, but activation of both kinases appears necessary to observe increased RyR sensitivity. The data provide novel insight into ß-adrenergic regulation of Ca(2+) release refractoriness in mouse myocytes.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Cálcio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Ventrículos do Coração/citologia , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
18.
Cardiovasc Res ; 106(1): 153-62, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25616416

RESUMO

AIMS: Duchenne muscular dystrophy (DMD), a degenerative pathology of skeletal muscle, also induces cardiac failure and arrhythmias due to a mutation leading to the lack of the protein dystrophin. In cardiac cells, the subsarcolemmal localization of dystrophin is thought to protect the membrane from mechanical stress. The absence of dystrophin results in an elevated stress-induced Ca2+ influx due to the inadequate functioning of several proteins, such as stretch-activated channels (SACs). Our aim was to investigate whether transient receptor potential vanilloid channels type 2 (TRPV2) form subunits of the dysregulated SACs in cardiac dystrophy. METHODS AND RESULTS: We defined the role of TRPV2 channels in the abnormal Ca2+ influx of cardiomyocytes isolated from dystrophic mdx mice, an established animal model for DMD. In dystrophic cells, western blotting showed that TRPV2 was two-fold overexpressed. While normally localized intracellularly, in myocytes from mdx mice TRPV2 channels were translocated to the sarcolemma and were prominent along the T-tubules, as indicated by immunocytochemistry. Membrane localization was confirmed by biotinylation assays. Furthermore, in mdx myocytes pharmacological modulators suggested an abnormal activity of TRPV2, which has a unique pharmacological profile among TRP channels. Confocal imaging showed that these compounds protected the cells from stress-induced abnormal Ca2+ signals. The involvement of TRPV2 in these signals was confirmed by specific pore-blocking antibodies and by small-interfering RNA ablation of TRPV2. CONCLUSION: Together, these results establish the involvement of TRPV2 in a stretch-activated calcium influx pathway in dystrophic cardiomyopathy, contributing to the defective cellular Ca2+ handling in this disease.


Assuntos
Canais de Cálcio/fisiologia , Cardiomiopatias/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Miócitos Cardíacos/patologia , Estresse Mecânico , Canais de Cátion TRPV/fisiologia , Animais , Cálcio/metabolismo , Cardiomiopatias/patologia , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/patologia , Osmose/fisiologia , Sarcolema/metabolismo , Transdução de Sinais/fisiologia
19.
Biophys J ; 107(12): 2815-2827, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25517148

RESUMO

Cellular oxidative stress, associated with a variety of common cardiac diseases, is well recognized to affect the function of several key proteins involved in Ca(2+) signaling and excitation-contraction coupling, which are known to be exquisitely sensitive to reactive oxygen species. These include the Ca(2+) release channels of the sarcoplasmic reticulum (ryanodine receptors or RyR2s) and the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). Oxidation of RyR2s was found to increase the open probability of the channel, whereas CaMKII can be activated independent of Ca(2+) through oxidation. Here, we investigated how oxidative stress affects RyR2 function and SR Ca(2+) signaling in situ, by analyzing Ca(2+) sparks in permeabilized mouse cardiomyocytes under a broad range of oxidative conditions. The results show that with increasing oxidative stress Ca(2+) spark duration is prolonged. In addition, long and very long-lasting (up to hundreds of milliseconds) localized Ca(2+) release events started to appear, eventually leading to sarcoplasmic reticulum (SR) Ca(2+) depletion. These changes of release duration could be prevented by the CaMKII inhibitor KN93 and did not occur in mice lacking the CaMKII-specific S2814 phosphorylation site on RyR2. The appearance of long-lasting Ca(2+) release events was paralleled by an increase of RyR2 oxidation, but also by RyR-S2814 phosphorylation, and by CaMKII oxidation. Our results suggest that in a strongly oxidative environment oxidation-dependent activation of CaMKII leads to RyR2 phosphorylation and thereby contributes to the massive prolongation of SR Ca(2+) release events.


Assuntos
Sinalização do Cálcio , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
20.
Cardiovasc Res ; 100(3): 392-401, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23963842

RESUMO

AIMS: During ß-adrenergic receptor (ß-AR) stimulation, phosphorylation of cardiomyocyte ryanodine receptors by protein kinases may contribute to an increased diastolic Ca(2+) spark frequency. Regardless of prompt activation of protein kinase A during ß-AR stimulation, this appears to rely more on activation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), by a not yet identified signalling pathway. The goal of the present study was to identify and characterize the mechanisms which lead to CaMKII activation and elevated Ca(2+) spark frequencies during ß-AR stimulation in single cardiomyocytes in diastolic conditions. METHODS AND RESULTS: Confocal imaging revealed that ß-AR stimulation increases endogenous NO production in cardiomyocytes, resulting in NO-dependent activation of CaMKII and a subsequent increase in diastolic Ca(2+) spark frequency. These changes of spark frequency could be mimicked by exposure to the NO donor GSNO and were sensitive to the CaMKII inhibitors KN-93 and AIP. In vitro, CaMKII became nitrosated and its activity remained increased independent of Ca(2+) in the presence of GSNO, as assessed with biochemical assays. CONCLUSIONS: ß-AR stimulation of cardiomyocytes may activate CaMKII by a novel direct pathway involving NO, without requiring Ca(2+) transients. This crosstalk between two established signalling pathways may contribute to arrhythmogenic diastolic Ca(2+) release and Ca(2+) waves during adrenergic stress, particularly in combination with cardiac diseases. In addition, NO-dependent activation of CaMKII is likely to have repercussions in many cellular signalling systems and cell types.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Diástole , Ativação Enzimática , Cobaias , Miócitos Cardíacos/enzimologia , Doadores de Óxido Nítrico/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA