Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(7): e2207887120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745790

ABSTRACT

Mammalian voltage-activated L-type Ca2+ channels, such as Ca(v)1.2, control transmembrane Ca2+ fluxes in numerous excitable tissues. Here, we report that the pore-forming α1C subunit of Ca(v)1.2 is reversibly palmitoylated in rat, rabbit, and human ventricular myocytes. We map the palmitoylation sites to two regions of the channel: The N terminus and the linker between domains I and II. Whole-cell voltage clamping revealed a rightward shift of the Ca(v)1.2 current-voltage relationship when α1C was not palmitoylated. To examine function, we expressed dihydropyridine-resistant α1C in human induced pluripotent stem cell-derived cardiomyocytes and measured Ca2+ transients in the presence of nifedipine to block the endogenous channels. The transients generated by unpalmitoylatable channels displayed a similar activation time course but significantly reduced amplitude compared to those generated by wild-type channels. We thus conclude that palmitoylation controls the voltage sensitivity of Ca(v)1.2. Given that the identified Ca(v)1.2 palmitoylation sites are also conserved in most Ca(v)1 isoforms, we propose that palmitoylation of the pore-forming α1C subunit provides a means to regulate the voltage sensitivity of voltage-activated Ca2+ channels in excitable cells.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Rats , Humans , Rabbits , Animals , Myocytes, Cardiac/metabolism , Calcium/metabolism , Lipoylation , Calcium Channels, L-Type/metabolism , Induced Pluripotent Stem Cells/metabolism , Calcium, Dietary , Mammals/metabolism
3.
FEBS J ; 289(20): 6267-6285, 2022 10.
Article in English | MEDLINE | ID: mdl-35633070

ABSTRACT

Post-translational modification of the myofilament protein troponin I by phosphorylation is known to trigger functional changes that support enhanced contraction and relaxation of the heart. We report for the first time that human troponin I can also be modified by SUMOylation at lysine 177. Functionally, TnI SUMOylation is not a factor in the development of passive and maximal force generation in response to calcium, however this modification seems to act indirectly by preventing SUMOylation of other myofilament proteins to alter calcium sensitivity and cooperativity of myofilaments. Utilising a novel, custom SUMO site-specific antibody that recognises only the SUMOylated form of troponin I, we verify that this modification occurs in human heart and that it is upregulated during disease.


Subject(s)
Calcium , Troponin I , Calcium/metabolism , Humans , Lysine/metabolism , Myofibrils/metabolism , Phosphorylation , Sumoylation , Troponin I/genetics , Troponin I/metabolism
4.
Cardiovasc Res ; 118(15): 3112-3125, 2022 12 09.
Article in English | MEDLINE | ID: mdl-35020837

ABSTRACT

AIMS: Cardiac electrophysiological heterogeneity includes: (i) regional differences in action potential (AP) waveform, (ii) AP waveform differences in cells isolated from a single region, (iii) variability of the contribution of individual ion currents in cells with similar AP durations (APDs). The aim of this study is to assess intra-regional AP waveform differences, to quantify the contribution of specific ion channels to the APD via drug responses and to generate a population of mathematical models to investigate the mechanisms underlying heterogeneity in rabbit ventricular cells. METHODS AND RESULTS: APD in ∼50 isolated cells from subregions of the LV free wall of rabbit hearts were measured using a voltage-sensitive dye. When stimulated at 2 Hz, average APD90 value in cells from the basal epicardial region was 254 ± 25 ms (mean ± standard deviation) in 17 hearts with a mean interquartile range (IQR) of 53 ± 17 ms. Endo-epicardial and apical-basal APD90 differences accounted for ∼10% of the IQR value. Highly variable changes in APD occurred after IK(r) or ICa(L) block that included a sub-population of cells (HR) with an exaggerated (hyper) response to IK(r) inhibition. A set of 4471 AP models matching the experimental APD90 distribution was generated from a larger population of models created by random variation of the maximum conductances (Gmax) of 8 key ion channels/exchangers/pumps. This set reproduced the pattern of cell-specific responses to ICa(L) and IK(r) block, including the HR sub-population. The models exhibited a wide range of Gmax values with constrained relationships linking ICa(L) with IK(r), ICl, INCX, and INaK. CONCLUSION: Modelling the measured range of inter-cell APDs required a larger range of key Gmax values indicating that ventricular tissue has considerable inter-cell variation in channel/pump/exchanger activity. AP morphology is retained by relationships linking specific ionic conductances. These interrelationships are necessary for stable repolarization despite large inter-cell variation of individual conductances and this explains the variable sensitivity to ion channel block.


Subject(s)
Ion Channels , Myocytes, Cardiac , Animals , Rabbits , Myocytes, Cardiac/physiology
5.
J Mol Cell Cardiol ; 165: 86-102, 2022 04.
Article in English | MEDLINE | ID: mdl-34999055

ABSTRACT

Cyclic AMP is a ubiquitous second messenger used to transduce intracellular signals from a variety of Gs-coupled receptors. Compartmentalisation of protein intermediates within the cAMP signaling pathway underpins receptor-specific responses. The cAMP effector proteins protein-kinase A and EPAC are found in complexes that also contain phosphodiesterases whose presence ensures a coordinated cellular response to receptor activation events. Popeye domain containing (POPDC) proteins are the most recent class of cAMP effectors to be identified and have crucial roles in cardiac pacemaking and conduction. We report the first observation that POPDC proteins exist in complexes with members of the PDE4 family in cardiac myocytes. We show that POPDC1 preferentially binds the PDE4A sub-family via a specificity motif in the PDE4 UCR1 region and that PDE4s bind to the Popeye domain of POPDC1 in a region known to be susceptible to a mutation that causes human disease. Using a cell-permeable disruptor peptide that displaces the POPDC1-PDE4 complex we show that PDE4 activity localized to POPDC1 modulates cycle length of spontaneous Ca2+ transients firing in intact mouse sinoatrial nodes.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Animals , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Mice , Phosphoric Diester Hydrolases/metabolism , Second Messenger Systems , Signal Transduction
6.
J Physiol ; 597(2): 399-418, 2019 01.
Article in English | MEDLINE | ID: mdl-30412283

ABSTRACT

KEY POINTS: Using 3D direct stochastic optical reconstruction microscopy (dSTORM), we developed novel approaches to quantitatively describe the nanoscale, 3D organization of ryanodine receptors (RyRs) in cardiomyocytes. Complex arrangements of RyR clusters were observed in 3D space, both at the cell surface and within the cell interior, with allocation to dyadic and non-dyadic pools. 3D imaging importantly allowed discernment of clusters overlapping in the z-axis, for which detection was obscured by conventional 2D imaging techniques. Thus, RyR clusters were found to be significantly smaller than previous 2D estimates. Ca2+ release units (CRUs), i.e. functional groupings of neighbouring RyR clusters, were similarly observed to be smaller than earlier reports. Internal CRUs contained more RyRs in more clusters than CRUs on the cell surface, and yielded longer duration Ca2+ sparks. ABSTRACT: Cardiomyocyte contraction is dependent on Ca2+ release from ryanodine receptors (RyRs). However, the precise localization of RyRs remains unknown, due to shortcomings of imaging techniques which are diffraction limited or restricted to 2D. We aimed to determine the 3D nanoscale organization of RyRs in rat cardiomyocytes by employing direct stochastic optical reconstruction microscopy (dSTORM) with phase ramp technology. Initial observations at the cell surface showed an undulating organization of RyR clusters, resulting in their frequent overlap in the z-axis and obscured detection by 2D techniques. Non-overlapping clusters were imaged to create a calibration curve for estimating RyR number based on recorded fluorescence blinks. Employing this method at the cell surface and interior revealed smaller RyR clusters than 2D estimates, as erroneous merging of axially aligned RyRs was circumvented. Functional groupings of RyR clusters (Ca2+ release units, CRUs), contained an average of 18 and 23 RyRs at the surface and interior, respectively, although half of all CRUs contained only a single 'rogue' RyR. Internal CRUs were more tightly packed along z-lines than surface CRUs, contained larger and more numerous RyR clusters, and constituted ∼75% of the roughly 1 million RyRs present in an average cardiomyocyte. This complex internal 3D geometry was underscored by correlative imaging of RyRs and t-tubules, which enabled quantification of dyadic and non-dyadic RyR populations. Mirroring differences in CRU size and complexity, Ca2+ sparks originating from internal CRUs were of longer duration than those at the surface. These data provide novel, nanoscale insight into RyR organization and function across cardiomyocytes.


Subject(s)
Myocytes, Cardiac/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Calcium Signaling/physiology , Imaging, Three-Dimensional , Male , Microscopy, Confocal , Rats, Wistar
7.
Elife ; 72018 10 30.
Article in English | MEDLINE | ID: mdl-30375974

ABSTRACT

Reduced cardiac contractility during heart failure (HF) is linked to impaired Ca2+ release from Ryanodine Receptors (RyRs). We investigated whether this deficit can be traced to nanoscale RyR reorganization. Using super-resolution imaging, we observed dispersion of RyR clusters in cardiomyocytes from post-infarction HF rats, resulting in more numerous, smaller clusters. Functional groupings of RyR clusters which produce Ca2+ sparks (Ca2+ release units, CRUs) also became less solid. An increased fraction of small CRUs in HF was linked to augmented 'silent' Ca2+ leak, not visible as sparks. Larger multi-cluster CRUs common in HF also exhibited low fidelity spark generation. When successfully triggered, sparks in failing cells displayed slow kinetics as Ca2+ spread across dispersed CRUs. During the action potential, these slow sparks protracted and desynchronized the overall Ca2+ transient. Thus, nanoscale RyR reorganization during HF augments Ca2+ leak and slows Ca2+ release kinetics, leading to weakened contraction in this disease.


Subject(s)
Calcium/metabolism , Heart Failure/pathology , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Action Potentials , Animals , Cations, Divalent/metabolism , Disease Models, Animal , Microscopy, Fluorescence , Rats
8.
Cardiovasc Res ; 114(11): 1512-1524, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29668881

ABSTRACT

Aims: In ventricular myocytes from humans and large mammals, the transverse and axial tubular system (TATS) network is less extensive than in rodents with consequently a greater proportion of ryanodine receptors (RyRs) not coupled to this membrane system. TATS remodelling in heart failure (HF) and after myocardial infarction (MI) increases the fraction of non-coupled RyRs. Here we investigate whether this remodelling alters the activity of coupled and non-coupled RyR sub-populations through changes in local signalling. We study myocytes from patients with end-stage HF, compared with non-failing (non-HF), and myocytes from pigs with MI and reduced left ventricular (LV) function, compared with sham intervention (SHAM). Methods and results: Single LV myocytes for functional studies were isolated according to standard protocols. Immunofluorescent staining visualized organization of TATS and RyRs. Ca2+ was measured by confocal imaging (fluo-4 as indicator) and using whole-cell patch-clamp (37°C). Spontaneous Ca2+ release events, Ca2+ sparks, as a readout for RyR activity were recorded during a 15 s period following conditioning stimulation at 2 Hz. Sparks were assigned to cell regions categorized as coupled or non-coupled sites according to a previously developed method. Human HF myocytes had more non-coupled sites and these had more spontaneous activity than in non-HF. Hyperactivity of these non-coupled RyRs was reduced by Ca2+/calmodulin-dependent kinase II (CaMKII) inhibition. Myocytes from MI pigs had similar changes compared with SHAM controls as seen in human HF myocytes. As well as by CaMKII inhibition, in MI, the increased activity of non-coupled sites was inhibited by mitochondrial reactive oxygen species (mito-ROS) scavenging. Under adrenergic stimulation, Ca2+ waves were more frequent and originated at non-coupled sites, generating larger Na+/Ca2+ exchange currents in MI than in SHAM. Inhibition of CaMKII or mito-ROS scavenging reduced spontaneous Ca2+ waves, and improved excitation-contraction coupling. Conclusions: In HF and after MI, RyR microdomain re-organization enhances spontaneous Ca2+ release at non-coupled sites in a manner dependent on CaMKII activation and mito-ROS production. This specific modulation generates a substrate for arrhythmia that appears to be responsive to selective pharmacologic modulation.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Signaling , Cardiomyopathies/metabolism , Heart Failure/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Aged , Animals , Arrhythmias, Cardiac/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomyopathies/physiopathology , Case-Control Studies , Disease Models, Animal , Excitation Contraction Coupling , Female , Heart Failure/physiopathology , Humans , Male , Membrane Potentials , Middle Aged , Mitochondria, Heart/metabolism , Myocardial Contraction , Myocardial Infarction/physiopathology , NADPH Oxidase 2/metabolism , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Sus scrofa , Time Factors , Ventricular Function, Left , Ventricular Remodeling
9.
Front Physiol ; 9: 1773, 2018.
Article in English | MEDLINE | ID: mdl-30618792

ABSTRACT

Contraction of cardiomyocytes is dependent on sub-cellular structures called dyads, where invaginations of the surface membrane (t-tubules) form functional junctions with the sarcoplasmic reticulum (SR). Within each dyad, Ca2+ entry through t-tubular L-type Ca2+ channels (LTCCs) elicits Ca2+ release from closely apposed Ryanodine Receptors (RyRs) in the SR membrane. The efficiency of this process is dependent on the density and macroscale arrangement of dyads, but also on the nanoscale organization of LTCCs and RyRs within them. We presently review accumulating data demonstrating the remarkable plasticity of these structures. Dyads are known to form gradually during development, with progressive assembly of both t-tubules and junctional SR terminals, and precise trafficking of LTCCs and RyRs. While dyads can exhibit compensatory remodeling when required, dyadic degradation is believed to promote impaired contractility and arrythmogenesis in cardiac disease. Recent data indicate that this plasticity of dyadic structure/function is dependent on the regulatory proteins junctophilin-2, amphiphysin-2 (BIN1), and caveolin-3, which critically arrange dyadic membranes while stabilizing the position and activity of LTCCs and RyRs. Indeed, emerging evidence indicates that clustering of both channels enables "coupled gating", implying that nanoscale localization and function are intimately linked, and may allow fine-tuning of LTCC-RyR crosstalk. We anticipate that improved understanding of dyadic plasticity will provide greater insight into the processes of cardiac compensation and decompensation, and new opportunities to target the basic mechanisms underlying heart disease.

10.
Cardiovasc Res ; 108(3): 387-98, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26490742

ABSTRACT

AIMS: In atrial fibrillation (AF), abnormalities in Ca(2+) release contribute to arrhythmia generation and contractile dysfunction. We explore whether ryanodine receptor (RyR) cluster ultrastructure is altered and is associated with functional abnormalities in AF. METHODS AND RESULTS: Using high-resolution confocal microscopy (STED), we examined RyR cluster morphology in fixed atrial myocytes from sheep with persistent AF (N = 6) and control (Ctrl; N = 6) animals. RyR clusters on average contained 15 contiguous RyRs; this did not differ between AF and Ctrl. However, the distance between clusters was significantly reduced in AF (288 ± 12 vs. 376 ± 17 nm). When RyR clusters were grouped into Ca(2+) release units (CRUs), i.e. clusters separated by <150 nm, CRUs in AF had more clusters (3.43 ± 0.10 vs. 2.95 ± 0.02 in Ctrl), which were more dispersed. Furthermore, in AF cells, more RyR clusters were found between Z lines. In parallel experiments, Ca(2+) sparks were monitored in live permeabilized myocytes. In AF, myocytes had >50% higher spark frequency with increased spark time to peak (TTP) and duration, and a higher incidence of macrosparks. A computational model of the CRU was used to simulate the morphological alterations observed in AF cells. Increasing cluster fragmentation to the level observed in AF cells caused the observed changes, i.e. higher spark frequency, increased TTP and duration; RyR clusters dispersed between Z-lines increased the occurrence of macrosparks. CONCLUSION: In persistent AF, ultrastructural reorganization of RyR clusters within CRUs is associated with overactive Ca(2+) release, increasing the likelihood of propagating Ca(2+) release.


Subject(s)
Atrial Fibrillation/metabolism , Calcium Signaling , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Atrial Fibrillation/physiopathology , Computer Simulation , Disease Models, Animal , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Atria/ultrastructure , Kinetics , Microscopy, Confocal , Models, Cardiovascular , Models, Molecular , Myocytes, Cardiac/ultrastructure , Protein Conformation , Ryanodine Receptor Calcium Release Channel/ultrastructure , Sheep , Structure-Activity Relationship
11.
Cold Spring Harb Protoc ; 2015(5): 490-7, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25934930

ABSTRACT

This protocol describes the measurement of Ca(2+) sparks in intact myocytes by using a Ca(2+)-sensitive dye and imaging using laser scanning confocal microscopy. It takes advantage of spontaneous Ca(2+)-release events-sparks-using them as a measure of the activity of ryanodine receptors (RyRs). Two methodologies are described: One requires that cardiomyocytes be stimulated, preferably under voltage clamp by depolarizing pulses, until steady-state is reached, and then stimulation is stopped and Ca(2+) sparks are recorded. The second requires that cells be permeabilized and bathed in a solution to load the cell with Ca(2+) sufficient to elicit Ca(2+) sparks, but not Ca(2+) waves. These are then analyzed offline to quantify spark frequency and morphology. The advantages and disadvantages of each approach are discussed.


Subject(s)
Calcium/analysis , Cytological Techniques/methods , Fluorescent Dyes/metabolism , Microscopy, Confocal/methods , Myocytes, Cardiac/chemistry , Patch-Clamp Techniques/methods , Staining and Labeling/methods , Myocytes, Cardiac/metabolism
12.
Cold Spring Harb Protoc ; 2015(5): 498-503, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25934931

ABSTRACT

The decline of an intracellular calcium ([Ca(2+)]i) transient during a single excitation-contraction coupling (ECC) cycle reflects the combined activity of the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) pump and the sarcolemmal Na(+)-Ca(2+) exchanger (NCX), along with minor contributions of the plasma membrane Ca(2+)-ATPase and mitochondrial Ca(2+) uniporter, in removing Ca(2+) from the cytosol. A traditional approach for assessing the individual components is to fit the decline of the [Ca(2+)]i transient evoked during electrical stimulation with an exponential. This reflects mostly the SERCA-dependent rate of uptake, which can be properly deduced after correcting for a component of NCX removal. As NCX function is an important determinant of the membrane potential as well as the Ca(2+) balance, we present here several detailed protocols for assessing NCX function. As the reversal potential and the amplitudes of the current are highly dependent on the prevailing concentrations of Na(+) and Ca(2+), we show how NCX function can be assessed under highly controlled conditions, with Ca(2+) and Na(+) clamped, as well as under more physiological conditions, with freely changing Ca(2+) and Na(+).


Subject(s)
Calcium/analysis , Cytological Techniques/methods , Metabolic Networks and Pathways , Myocytes, Cardiac/chemistry , Calcium/metabolism , Electrophysiological Phenomena , Myocytes, Cardiac/metabolism
13.
Cold Spring Harb Protoc ; 2015(5): 431-3, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25934941

ABSTRACT

In cardiac myocytes, Ca(2+) release from the sarcoplasmic reticulum (SR) Ca(2+) store through the opening of ryanodine receptors (RyRs) is the major source of Ca(2+) for activation of myofilaments and contraction. Over the past 20 years, tools have become available to study this release process in detail, allowing new insights into the regulation of SR Ca(2+) release and RyR function. To assess these processes, we recommend and here review a systematic approach that evaluates the essential transport mechanisms and Ca(2+) fluxes in isolated single cardiac myocytes by using fluorescent Ca(2+) indicators and whole-cell recording of membrane voltage and ionic currents under voltage clamp. The approach includes an assessment of the L-type Ca(2+) current as a trigger for opening of RyRs and release of SR Ca(2+), of the SR Ca(2+) content, of intrinsic properties of RyRs, and of Ca(2+)-removal systems.


Subject(s)
Calcium/metabolism , Cytological Techniques/methods , Myocytes, Cardiac/physiology , Optical Imaging/methods , Patch-Clamp Techniques/methods , Muscle Contraction , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Staining and Labeling/methods
15.
Cold Spring Harb Protoc ; 2015(4): 398-402, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834258

ABSTRACT

Here, we describe a method for characterizing the L-type Ca(2+) current, ICaL, which is a major trigger for Ca(2+) release from the sarcoplasmic reticulum (SR). The protocol includes measuring ICaL amplitude and voltage dependence and the elicited SR Ca(2+) release. The procedure for measuring ICaL activity is performed using solutions (internal and external) and voltage control such that other ionic currents are eliminated. The resultant relationship between the Ca(2+) current and the associated internal [Ca(2+)]i transient is a first approach for evaluating coupling gain. We discuss which parameters are most appropriate for this analysis and how an evaluation of gain needs to be further explored by measuring the SR Ca(2+) content.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Dogs , Mice , Sus scrofa
16.
Cold Spring Harb Protoc ; 2015(4): 392-7, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834257

ABSTRACT

In cardiac myocytes, the physiological increase of intracellular calcium, the [Ca(2+)]i transient, elicited during excitation-contraction coupling typically reaches a peak amplitude of up to 1 µm, from a resting value of ∼100 nm, within 50-100 msec, depending on the species. Various conditions will affect the amplitude and rise time of the [Ca(2+)]i transient and, depending on the nature of the Ca(2+) signals under study, a variety of different probes are available for monitoring changes in intracellular Ca(2+). In this protocol, we focus on Fluo-3, which exists in the cytosol in its salt form K5Fluo-3. This form is practically nonfluorescent in the absence of Ca(2+), but the fluorescence increases dramatically on Ca(2+) binding. Although Fluo-3 is a single excitation-emission dye, it has a number of advantages for investigators, including an ideal dissociation constant (Kd) value and high quantum yield, meaning that it can be used at low concentrations that introduce minimal buffering. Here, we describe the basic setup and methodology for recording the global cytosolic [Ca(2+)]i transient with this probe during simultaneous patch-clamp and whole-cell recording of membrane voltage or of ionic currents under voltage clamp.


Subject(s)
Aniline Compounds/metabolism , Calcium/metabolism , Cytological Techniques/methods , Intracellular Space/metabolism , Myocytes, Cardiac/metabolism , Xanthenes/metabolism , Animals , Calibration , Coloring Agents/metabolism , Electrophysiological Phenomena , Fluorescence , Patch-Clamp Techniques
17.
Cold Spring Harb Protoc ; 2015(4): 403-7, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834259

ABSTRACT

Here, we describe a protocol for the reliable measurement of the amount of Ca(2+) in the sarcoplasmic reticulum (SR) Ca(2+) store of cardiac myocytes. The whole-cell patch-clamp method is used to provide controlled loading of the SR during conditioning depolarizing pulses, followed by rapid application of a high dose of caffeine to release all SR Ca(2+) and to prevent Ca(2+) reuptake by the SR. Simultaneous measurement of membrane currents records Ca(2+) extruded through the Na(+)-Ca(2+) exchanger. The integral of the caffeine-induced Na(+)-Ca(2+) exchange current is then used as a measure of the SR Ca(2+). Derived measurements include the Ca(2+) buffering capacity and measurement of fractional release as an indicator of coupling gain. Caveats, advantages, and disadvantages of this method and alternative methods are discussed.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques/methods , Sarcoplasmic Reticulum/metabolism , Animals , Buffers
18.
Circ Res ; 113(11): 1242-52, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24081880

ABSTRACT

RATIONALE: In ventricular myocytes of large mammals with low T-tubule density, a significant number of ryanodine receptors (RyRs) are not coupled to the sarcolemma; cardiac remodeling increases noncoupled RyRs. OBJECTIVE: Our aim was to test the hypothesis that coupled and noncoupled RyRs have distinct microdomain-dependent modulation. METHODS AND RESULTS: We studied single myocytes from pig left ventricle. The T-tubule network was analyzed in 3-dimension (3D) to measure distance to membrane of release sites. The rising phase of the Ca(2+) transient was correlated with proximity to the membrane (confocal imaging, whole-cell voltage-clamp, K5fluo-4 as Ca(2+) indicator). Ca(2+) sparks after stimulation were thus identified as resulting from coupled or noncoupled RyRs. We used high-frequency stimulation as a known activator of Ca(2+)/calmodulin-dependent kinase II. Spark frequency increased significantly more in coupled than in noncoupled RyRs. This specific modulation of coupled RyRs was abolished by the Ca(2+)/calmodulin-dependent kinase II blockers autocamtide-2-related inhibitory peptide and KN-93, but not by KN-92. Colocalization of Ca(2+)/calmodulin-dependent kinase II and RyR was not detectably different for coupled and noncoupled sites, but the F-actin disruptor cytochalasin D prevented the specific modulation of coupled RyRs. NADPH oxidase 2 inhibition by diphenyleneiodonium or apocynin, or global reactive oxygen species scavenging, also prevented coupled RyR modulation. During stimulated Ca(2+) transients, frequency-dependent increase of the rate of Ca(2+) rise was seen in coupled RyR regions only and abolished by autocamtide-2-related inhibitory peptide. After myocardial infarction, selective modulation of coupled RyR was lost. CONCLUSIONS: Coupled RyRs have a distinct modulation by Ca(2+)/calmodulin-dependent kinase II and reactive oxygen species, dependent on an intact cytoskeleton and consistent with a local Ca(2+)/reactive oxygen species microdomain, and subject to modification with disease.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Membrane Microdomains/physiology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcolemma/physiology , Animals , Calcium/metabolism , Disease Models, Animal , Imaging, Three-Dimensional , Microscopy, Confocal , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Reactive Oxygen Species/metabolism , Sarcoplasmic Reticulum/metabolism , Swine
19.
Circ Res ; 113(5): 527-38, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23825358

ABSTRACT

RATIONALE: Synchronized release of Ca²âº into the cytosol during each cardiac cycle determines cardiomyocyte contraction. OBJECTIVE: We investigated synchrony of cytosolic [Ca²âº] decay during diastole and the impact of cardiac remodeling. METHODS AND RESULTS: Local cytosolic [Ca²âº] transients (1-µm intervals) were recorded in murine, porcine, and human ventricular single cardiomyocytes. We identified intracellular regions of slow (slowCaR) and fast (fastCaR) [Ca²âº] decay based on the local time constants of decay (TAUlocal). The SD of TAUlocal as a measure of dyssynchrony was not related to the amplitude or the timing of local Ca²âº release. Stimulation of sarcoplasmic reticulum Ca²âº ATPase with forskolin or istaroxime accelerated and its inhibition with cyclopiazonic acid slowed TAUlocal significantly more in slowCaR, thus altering the relationship between SD of TAUlocal and global [Ca²âº] decay (TAUglobal). Na⁺/Ca²âº exchanger inhibitor SEA0400 prolonged TAUlocal similarly in slowCaR and fastCaR. FastCaR were associated with increased mitochondrial density and were more sensitive to the mitochondrial Ca²âº uniporter blocker Ru360. Variation in TAUlocal was higher in pig and human cardiomyocytes and higher with increased stimulation frequency (2 Hz). TAUlocal correlated with local sarcomere relengthening. In mice with myocardial hypertrophy after transverse aortic constriction, in pigs with chronic myocardial ischemia, and in end-stage human heart failure, variation in TAUlocal was increased and related to cardiomyocyte hypertrophy and increased mitochondrial density. CONCLUSIONS: In cardiomyocytes, cytosolic [Ca²âº] decay is regulated locally and related to local sarcomere relengthening. Dyssynchronous intracellular [Ca²âº] decay in cardiac remodeling and end-stage heart failure suggests a novel mechanism of cellular contractile dysfunction.


Subject(s)
Calcium Signaling/physiology , Heart Failure/physiopathology , Heart Ventricles/cytology , Myocytes, Cardiac/physiology , Ventricular Remodeling/physiology , Aniline Compounds/pharmacology , Animals , Calcium Signaling/drug effects , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Colforsin/pharmacology , Cytosol/metabolism , Diastole , Electric Stimulation , Etiocholanolone/analogs & derivatives , Etiocholanolone/pharmacology , Humans , Hypertrophy , Hypertrophy, Left Ventricular/physiopathology , Indoles/pharmacology , Mice , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenyl Ethers/pharmacology , Ruthenium Compounds/pharmacology , Sarcomeres/ultrastructure , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Sus scrofa , Swine
20.
Exp Physiol ; 98(1): 134-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22689442

ABSTRACT

Reducing the open probability of the ryanodine receptor (RyR) has been proposed to have beneficial effects in heart failure. We investigated whether conditional FKBP12.6 overexpression at the time of myocardial infarction (MI) could improve cardiac remodelling and cell Ca(2+) handling. Wild-type (WT) mice and mice overexpressing FKBP12.6 (Tg) were studied on average 7.5 ± 0.2 weeks after MI and compared with sham-operated mice for in vivo, myocyte function and remodelling. At baseline, unloaded cell shortening in Tg was not different from WT. The [Ca(2+)](i) transient amplitude was similar, but sarcoplasmic reticulum (SR) Ca(2+) content was larger in Tg, suggesting reduced fractional release. Spontaneous spark frequency was similar despite the increased SR Ca(2+) content, consistent with a reduced RyR channel open probability in Tg. After MI, left ventricular dilatation and myocyte hypertrophy were present in both groups, but more pronounced in Tg. Cell shortening amplitude was unchanged with MI in WT, but increased with MI in Tg. The amplitude of the [Ca(2+)](i) transient was not affected by MI in either genotype, but time to peak was increased; this was most pronounced in Tg. The SR Ca(2+) content and Na(+)- Ca(2+) exchanger function were not affected by MI. Spontaneous spark frequency was increased significantly after MI in Tg, and larger than in WT (at 4 Hz, 2.6 ± 0.4 sparks (100 µm)(-1) s(-1) in Tg MI versus 1.6 ± 0.2 sparks (100 µm)(-1) s(-1) in WT MI; P < 0.05). We conclude that FKPB12.6 overexpression can effectively reduce RyR open probability with maintained cardiomyocyte contraction. However, this approach appears insufficient to prevent and reduce post-MI remodelling, indicating that additional pathways may need to be targeted.


Subject(s)
Myocardial Infarction/physiopathology , Tacrolimus Binding Proteins/biosynthesis , Ventricular Remodeling/drug effects , Animals , Calcium/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocytes, Cardiac/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism , Tacrolimus Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...