Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807195

ABSTRACT

(1) Background: The exact mechanism(s) underlying pathological changes in a heart in transition to hypertrophy and failure are not yet fully understood. However, alterations in cardiac energy metabolism seem to be an important contributor. We characterized an in vitro model of adrenergic stimulation-induced cardiac hypertrophy for studying metabolic, structural, and functional changes over time. Accordingly, we investigated whether metabolic interventions prevent cardiac structural and functional changes; (2) Methods: Primary rat cardiomyocytes were treated with phenylephrine (PE) for 16 h, 24 h, or 48 h, whereafter hypertrophic marker expression, protein synthesis rate, glucose uptake, and contractile function were assessed; (3) Results: 24 h PE treatment increased expression of hypertrophic markers, phosphorylation of hypertrophy-related signaling kinases, protein synthesis, and glucose uptake. Importantly, the increased glucose uptake preceded structural and functional changes, suggesting a causal role for metabolism in the onset of PE-induced hypertrophy. Indeed, PE treatment in the presence of a PAN-Akt inhibitor or of a GLUT4 inhibitor dipyridamole prevented PE-induced increases in cellular glucose uptake and ameliorated PE-induced contractile alterations; (4) Conclusions: Pharmacological interventions, forcing substrate metabolism away from glucose utilization, improved contractile properties in PE-treated cardiomyocytes, suggesting that targeting glucose uptake, independent from protein synthesis, forms a promising strategy to prevent hypertrophy and hypertrophy-induced cardiac dysfunction.


Subject(s)
Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Animals , Animals, Newborn , Cells, Cultured , Energy Metabolism , Glucose/metabolism , Muscle Contraction/drug effects , Muscle Contraction/physiology , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Phosphorylation , Rats , Signal Transduction/drug effects
2.
Int J Mol Sci ; 21(4)2020 Feb 23.
Article in English | MEDLINE | ID: mdl-32102213

ABSTRACT

The diabetic heart is characterized by a shift in substrate utilization from glucose to lipids, which may ultimately lead to contractile dysfunction. This substrate shift is facilitated by increased translocation of lipid transporter CD36 (SR-B2) from endosomes to the sarcolemma resulting in increased lipid uptake. We previously showed that endosomal retention of CD36 is dependent on the proper functioning of vacuolar H+-ATPase (v-ATPase). Excess lipids trigger CD36 translocation through inhibition of v-ATPase function. Conversely, in yeast, glucose availability is known to enhance v-ATPase function, allowing us to hypothesize that glucose availability, via v-ATPase, may internalize CD36 and restore contractile function in lipid-overloaded cardiomyocytes. Increased glucose availability was achieved through (a) high glucose (25 mM) addition to the culture medium or (b) adenoviral overexpression of protein kinase-D1 (a kinase mediating GLUT4 translocation). In HL-1 cardiomyocytes, adult rat and human cardiomyocytes cultured under high-lipid conditions, each treatment stimulated v-ATPase re-assembly, endosomal acidification, endosomal CD36 retention and prevented myocellular lipid accumulation. Additionally, these treatments preserved insulin-stimulated GLUT4 translocation and glucose uptake as well as contractile force. The present findings reveal v-ATPase functions as a key regulator of cardiomyocyte substrate preference and as a novel potential treatment approach for the diabetic heart.


Subject(s)
Lipid Metabolism , Lipids/adverse effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/pharmacology , Animals , Biological Transport/drug effects , CD36 Antigens/metabolism , Endosomes/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Induced Pluripotent Stem Cells , Insulin Resistance , Lipid Accumulation Product , Male , Myocardial Contraction/drug effects , Phosphotransferases/metabolism , Rats , Sarcolemma/metabolism , Triglycerides/metabolism
3.
J Electrocardiol ; 49(2): 124-31, 2016.
Article in English | MEDLINE | ID: mdl-26803554

ABSTRACT

Mechanisms underlying atrial remodeling toward atrial fibrillation (AF) are incompletely understood. We induced AF in 16 pigs by 6weeks of rapid atrial pacing (RAP, 600bpm) using a custom-built, telemetrically controlled pacemaker. AF evolution was monitored three times per week telemetrically in unstressed, conscious animals. We established a dose-response relationship between RAP duration and occurrence of sustained AF >60minutes. Left atrial (LA) dilatation was present already at 2weeks of RAP. There was no evidence of left ventricular heart failure after 6weeks of RAP. As a proof-of-principle, arterial hypertension was induced in 5/16 animals by implanting desoxycorticosterone acetate (DOCA, an aldosterone-analog) subcutaneously to accelerate atrial remodeling. RAP+DOCA resulted in increased AF stability with earlier onset of sustained AF and accelerated anatomical atrial remodeling with more pronounced LA dilatation. This novel porcine model can serve to characterize effects of maladaptive stimuli or protective interventions specifically during early AF.


Subject(s)
Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Disease Models, Animal , Pacemaker, Artificial , Prostheses and Implants , Telemetry/instrumentation , Animals , Equipment Design , Equipment Failure Analysis , Female , Swine , Telemetry/methods
4.
J Mol Cell Cardiol ; 89(Pt B): 326-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26454162

ABSTRACT

Beat-to-beat variability of ventricular repolarization (BVR) has been proposed as a strong predictor of Torsades de Pointes (TdP). BVR is also observed at the myocyte level, and a number of studies have shown the importance of calcium handling in influencing this parameter. The chronic AV block (CAVB) dog is a model of TdP arrhythmia in cardiac hypertrophy, and myocytes from these animals show extensive remodeling, including of Ca(2+) handling. This remodeling process also leads to increased BVR. We aimed to determine the role that (local) Ca(2+) handling plays in BVR. In isolated LV myocytes an exponential relationship was observed between BVR magnitude and action potential duration (APD) at baseline. Inhibition of Ca(2+) release from sarcoplasmic reticulum (SR) with thapsigargin resulted in a reduction of [Ca(2+)]i, and of both BVR and APD. Increasing ICaL in the presence of thapsigargin restored APD but BVR remained low. In contrast, increasing ICaL with preserved Ca(2+) release increased both APD and BVR. Inhibition of Ca(2+) release with caffeine, as with thapsigargin, reduced BVR despite maintained APD. Simultaneous inhibition of Na(+)/Ca(2+) exchange and ICaL decreased APD and BVR to similar degrees, whilst increasing diastolic Ca(2+). Buffering of Ca(2+) transients with BAPTA reduced BVR for a given APD to a greater extent than buffering with EGTA, suggesting subsarcolemmal Ca(2+) transients modulated BVR to a larger extent than the cytosolic Ca(2+) transient. In conclusion, BVR in hypertrophied dog myocytes, at any APD, is strongly dependent on SR Ca(2+) release, which may act through modulation of the l-type Ca(2+) current in a subsarcolemmal microdomain.


Subject(s)
Atrioventricular Block/metabolism , Atrioventricular Block/physiopathology , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Heart Rate , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Animals , Caffeine/pharmacology , Calcium Signaling/drug effects , Chronic Disease , Dogs , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Sarcoplasmic Reticulum/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism
5.
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
6.
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
7.
Europace ; 17(10): 1457-66, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26364316

ABSTRACT

Atrial fibrillation (AF) is the most common sustained arrhythmia in the general population. As an age-related arrhythmia AF is becoming a huge socio-economic burden for European healthcare systems. Despite significant progress in our understanding of the pathophysiology of AF, therapeutic strategies for AF have not changed substantially and the major challenges in the management of AF are still unmet. This lack of progress may be related to the multifactorial pathogenesis of atrial remodelling and AF that hampers the identification of causative pathophysiological alterations in individual patients. Also, again new mechanisms have been identified and the relative contribution of these mechanisms still has to be established. In November 2010, the European Union launched the large collaborative project EUTRAF (European Network of Translational Research in Atrial Fibrillation) to address these challenges. The main aims of EUTRAF are to study the main mechanisms of initiation and perpetuation of AF, to identify the molecular alterations underlying atrial remodelling, to develop markers allowing to monitor this processes, and suggest strategies to treat AF based on insights in newly defined disease mechanisms. This article reports on the objectives, the structure, and initial results of this network.


Subject(s)
Atrial Fibrillation/diagnosis , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Remodeling , Translational Research, Biomedical/trends , Cooperative Behavior , Electrocardiography , Europe , Humans
8.
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
9.
Adv Exp Med Biol ; 961: 375-83, 2013.
Article in English | MEDLINE | ID: mdl-23224896

ABSTRACT

The Na(+)/Ca(2+) exchanger (NCX) is a key regulator of intracellular Ca(2+) in cardiac myocytes, predominantly contributing to Ca(2+) removal during the diastolic relaxation process but also modulating excitation-contraction coupling. NCX is preferentially located in the T-tubules and can be close to or within the dyad, where L-type Ca(2+) channels face ryanodine receptors (RyRs), the Ca(2+) release channels of the sarcoplasmic reticulum. However, especially in larger animals, not all RyRs are in dyads or adjacent to T-tubules, and a substantial fraction of Ca(2+) release from the sarcoplasmic reticulum thus occurs at distance from NCX. This chapter deals with the functional consequences of NCX location and how NCX can modulate diastolic and systolic Ca(2+) events. The loss of T-tubules and the effects on RyR function and NCX modulation are explored, as well as quantitative measurement of local Ca(2+) gradients at the level of the dyadic space.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium/metabolism , Animals , Calcium Channels, L-Type/genetics , Humans , Muscle Proteins/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/genetics , Sodium-Calcium Exchanger/genetics
10.
J Physiol ; 590(18): 4423-46, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22547631

ABSTRACT

In cardiac cells, Ca(2+) release flux (J(rel)) via ryanodine receptors (RyRs) from the sarcoplasmic reticulum (SR) has a complex effect on the action potential (AP). Coupling between J(rel) and the AP occurs via L-type Ca(2+) channels (I(Ca)) and the Na(+)/Ca(2+) exchanger (I(NCX)). We used a combined experimental and modelling approach to study interactions between J(rel), I(Ca) and I(NCX) in porcine ventricular myocytes.We tested the hypothesis that during normal uniform J(rel), the interaction between these fluxes can be represented as occurring in two myoplasmic subcompartments for Ca(2+) distribution, one (T-space) associated with RyR and enclosed by the junctional portion of the SR membrane and corresponding T-tubular portion of the sarcolemma, the other (M-space) encompassing the rest of the myoplasm. I(Ca) and I(NCX) were partitioned into subpopulations in the T-space and M-space sarcolemma. We denoted free Ca(2+) concentrations in T-space and M-space Ca(t) and Ca(m), respectively. Experiments were designed to allow separate measurements of I(Ca) and I(NCX) as a function of J(rel). Inclusion of T-space in themodel allowed us to reproduce in silico the following important experimental results: (1) hysteresis of I(NCX) dependence on Ca(m); (2) delay between peak I(NCX) and peak Ca(m) during caffeine application protocol; (3) delay between I(NCX) and Ca(m) during Ca(2+)-induced-Ca(2+)-release; (4) rapid I(Ca) inactivation (within 2 ms) due to J(rel), with magnitude graded as a function of the SR Ca(2+) content; (5) time delay between I(Ca) inactivation due to J(rel) and Ca(m). Partition of 25% NCX in T-space and 75% in M-space provided the best fit to the experimental data. Measured Ca(m) and I(Ca) or I(NCX) were used as input to the model for estimating Ca(t). The actual model-computed Ca(t), obtained by simulating specific experimental protocols, was used as a gold standard for comparison. The model predicted peak Ca(t) in the range of 6­25 µM, with time to equilibrium of Ca(t) with Ca(m) of ~350 ms. These Ca(t) values are in the range of LCC and RyR sensitivity to Ca(2+). An increase of the SR Ca(2+) load increased the time to equilibrium. The I(Ca)-based estimation method was most accurate during the ascending phase of Ca(t). The I(NCX)-based method provided a good estimate for the descending phase of Ca(t). Thus, application of both methods in combination provides the best estimate of the entire Ca(t) time course.


Subject(s)
Calcium/physiology , Models, Cardiovascular , Myocytes, Cardiac/physiology , Animals , Calcium Channels, L-Type/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology , Sodium-Calcium Exchanger/physiology , Swine
11.
Basic Res Cardiol ; 107(6): 304, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23095853

ABSTRACT

Connexin mimetic peptides (CxMPs), such as Gap26 and Gap27, are known as inhibitors of gap junction channels but evidence is accruing that these peptides also inhibit unapposed/non-junctional hemichannels (HCs) residing in the plasma membrane. We used voltage clamp studies to investigate the effect of Gap26/27 at the single channel level. Such an approach allows unequivocal identification of HC currents by their single channel conductance that is typically ~220 pS for Cx43. In HeLa cells stably transfected with Cx43 (HeLa-Cx43), Gap26/27 peptides inhibited Cx43 HC unitary currents over minutes and increased the voltage threshold for HC opening. By contrast, an elevation of intracellular calcium ([Ca(2+)](i)) to 200-500 nM potentiated the unitary HC current activity and lowered the voltage threshold for HC opening. Interestingly, Gap26/27 inhibited the Ca(2+)-potentiated HC currents and prevented lowering of the voltage threshold for HC opening. Experiments on isolated pig ventricular cardiomyocytes, which display strong endogenous Cx43 expression, demonstrated voltage-activated unitary currents with biophysical properties of Cx43 HCs that were inhibited by small interfering RNA targeting Cx43. As observed in HeLa-Cx43 cells, HC current activity in ventricular cardiomyocytes was potentiated by [Ca(2+)](i) elevation to 500 nM and was inhibited by Gap26/27. Our results indicate that under pathological conditions, when [Ca(2+)](i) is elevated, Cx43 HC opening is promoted in cardiomyocytes and CxMPs counteract this effect.


Subject(s)
Calcium/metabolism , Connexin 43/metabolism , Ion Channels/metabolism , Myocytes, Cardiac/metabolism , Animals , HeLa Cells , Humans , Membrane Potentials , Patch-Clamp Techniques , Peptides , Swine
12.
J Physiol ; 589(Pt 10): 2569-83, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21486798

ABSTRACT

During Ca²âº release from the sarcoplasmic reticulum triggered by Ca²âº influx through L-type Ca²âº channels (LTCCs), [Ca²âº] near release sites ([Ca²âº]nrs) temporarily exceeds global cytosolic [Ca²âº]. [Ca²âº]nrs can at present not be measured directly but the Na+/Ca2+ exchanger (NCX) near release sites and LTCCs also experience [Ca²âº]nrs. We have tested the hypothesis that ICaL and INCX could be calibrated to report [Ca²âº]nrs and would report different time course and values for local [Ca²âº]. Experiments were performed in pig ventricular myocytes (whole-cell voltage-clamp, Fluo-3 to monitor global cytosolic [Ca²âº], 37◦C). [Ca²âº]nrs-dependent inactivation of ICaL during a step to +10 mV peaked around 10 ms. For INCX we computationally isolateda current fraction activated by [Ca²âº]nrs; values were maximal at 10 ms into depolarization. The recovery of [Ca²âº]nrs was comparable with both reporters (>90% within 50 ms). Calibration yielded maximal values for [Ca²âº]nrs between 10 and 15 µmol l⁻¹ with both methods. When applied to a step to less positive potentials (-30 to -20 mV), the time course of [Ca²âº]nrs was slower but peak values were not very different. In conclusion, both ICaL inactivation and INCX activation, using a subcomponent analysis, can be used to report dynamic changes of [Ca²âº]nrs. Absolute values obtained by these different methods are within the same range, suggesting that they are reporting on a similar functional compartment near ryanodine receptors. Comparable [Ca²âº]nrs at +10 mV and -20 mV suggests that, although the number of activated release sites differs at these potentials, local gradients at release sites can reach similar values.


Subject(s)
Membrane Microdomains/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Sodium-Calcium Exchanger/physiology , Aniline Compounds/administration & dosage , Animals , Calcium/analysis , Calcium/metabolism , Calcium/physiology , Calcium Channels, L-Type/physiology , Fluorescent Dyes/administration & dosage , Membrane Potentials/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/physiology , Swine , Ventricular Function , Xanthenes/administration & dosage
13.
ESC Heart Fail ; 8(1): 151-161, 2021 02.
Article in English | MEDLINE | ID: mdl-33251761

ABSTRACT

AIMS: Atrial contractile dysfunction contributes to worse prognosis in hypertensive heart disease (HHD), but the role of cardiomyocyte dysfunction in atrial remodelling in HHD is not well understood. We investigated and compared cellular mechanisms of left (LA) and right atrial (RA) contractile dysfunction in pigs with HHD. METHODS AND RESULTS: In vivo electrophysiological and magnetic resonance imaging studies were performed in control and pigs treated with 11-deoxycorticosterone acetate (DOCA)/high-salt/glucose diet (12 weeks) to induce HHD. HHD leads to significant atrial remodelling and loss of contractile function in LA and a similar trend in RA (magnetic resonance imaging). Atrial remodelling was associated with a higher inducibility of atrial fibrillation but unrelated to changes in atrial refractory period or fibrosis (histology). Reduced atrial function in DOCA pigs was related to reduced contraction amplitude of isolated LA (already at baseline) and RA myocytes (at higher frequencies) due to reduced intracellular Ca release (Fura 2-AM, field stimulation). However, Ca regulation differed in LA and RA cardiomyocytes: LA cardiomyocytes showed reduced sarcoplasmic reticulum (SR) [Ca], whereas in RA, SR [Ca] was unchanged and SR Ca2+ -ATPase activity was increased. Sodium-calcium exchanger (NCX) activity was not significantly altered. We used ORM-10103 (3 µM), a specific NCX inhibitor to improve Ca availability in LA and RA cardiomyocytes from DOCA pigs. Partial inhibition of NCX increased Ca2+ transient amplitude and SR Ca in LA, but not RA cells. CONCLUSIONS: In this large animal model of HHD, atrial remodelling in sinus rhythm in vivo was related to differential LA and RA cardiomyocyte dysfunction and Ca signalling. Selective acute inhibition of NCX improved Ca release in diseased LA cardiomyocytes, suggesting a potential therapeutic approach to improve atrial inotropy in HHD.


Subject(s)
Calcium , Hypertension , Animals , Calcium/metabolism , Heart Atria/diagnostic imaging , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger , Swine
14.
Cardiovasc Res ; 77(2): 315-24, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18006436

ABSTRACT

In the cardiac dyad, sarcolemmal L-type Ca(2+) channels (LCCs) and sarcoplasmic reticulum (SR) Ca(2+) release channels (RyR) are structurally in close proximity. This organization provides for an efficient functional coupling, tuning SR Ca(2+) release for optimal contraction of the myocyte. Given that LCC are regulated by the prevailing [Ca(2+)], this structural organization is the setting for feedback mechanisms and crosstalk. A defective coupling of Ca(2+) influx via LCC to activation of RyR has been implicated in reduced SR Ca(2+) release in heart failure. Both functional changes in LCC properties and structural re-organization of LCC in T-tubules could be involved. LCC are regulated by cytosolic Ca(2+), and crosstalk with SR Ca(2+) handling occurs on a long-term basis, i.e. during steady-state changes in heart rate, on an intermediate-term basis, i.e. on a beat-to-beat basis during sudden rate changes, and on a very short- or immediate-term basis, i.e. during a single heartbeat. We review the properties and consequences of these different feedback mechanisms and the changes in heart failure and cardiac hypertrophy that have thus far been studied.


Subject(s)
Calcium Channels, L-Type/physiology , Cardiomegaly/metabolism , Heart Failure/metabolism , Sarcoplasmic Reticulum/physiology , Animals , Arrhythmias, Cardiac/etiology , Calcium/metabolism , Feedback, Physiological , Heart Rate , Humans , Myocardial Contraction , Ryanodine Receptor Calcium Release Channel/physiology
15.
Heart Rhythm ; 5(3): 438-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18313604

ABSTRACT

BACKGROUND: Several cardiac disorders affect the right ventricle (RV) and left ventricle (LV) equally, but nevertheless, RV vulnerability to conduction slowing and arrhythmias exceeds that of the LV. OBJECTIVE: This study sought to assess the mechanism of dominant RV arrhythmia vulnerability in senescent mice as a model of general reduced myocardial integrity. METHODS: Epicardial ventricular activation mapping was performed on senescent (22 months) and adult (3 months) Langendorff perfused mouse hearts. Arrhythmia inducibility was tested by programmed stimulation. Conduction velocity longitudinal and transversal (CVT) to fiber orientation, conduction heterogeneity, and effective refractory period were determined. Subsequently, hearts were processed for immunohistochemistry, Western blotting, and Sirius red staining. RESULTS: In senescent RV, but not LV, CVT was reduced and wavelength decreased, whereas anisotropic ratio and conduction heterogeneity increased. Arrhythmias, based on anisotropic reentry, were induced in 55% of senescent hearts only and predominantly in RV. In senescent mice, Connexin 43 (Cx43) and Cardiac Sodium Channel (Nav1.5) were decreased and interstitial fibrosis increased comparably in RV and LV. However, in senescent mice, heterogeneously distributed patches of replacement fibrosis were present throughout the entire RV myocardium, but only in midendocardium and subendocardium of LV. Cx43 expression in these areas was disrupted. CONCLUSION: Widespread presence of replacement fibrosis in senescent RV compared with LV, combined with Cx43 and Nav1.5 disruption, potentiate shorter wavelength, conduction slowing, and conduction heterogeneity in RV, resulting in greater vulnerability of senescent RV to arrhythmias.


Subject(s)
Aging/physiology , Arrhythmias, Cardiac/physiopathology , Heart Conduction System/physiopathology , Heart Ventricles/physiopathology , Analysis of Variance , Animals , Blotting, Western , Echocardiography, Doppler , Electrocardiography , Fibrosis , Immunohistochemistry , Linear Models , Mice , Mice, Inbred C57BL
16.
Europace ; 10(12): 1364-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18845561

ABSTRACT

Abnormal calcium (Ca) handling can contribute to arrhythmogenesis directly by triggering abnormal depolarizations and indirectly by modulating action potential time course and duration. Recent data have shown the importance of these mechanisms in rare genetic diseases but also in more common conditions such as heart failure. Modulating Ca release from the sarcoplasmic reticulum via the ryanodine receptor, Ca uptake via sarcoplasmic reticulum Ca ATPase or Ca removal from the cell via the Na/Ca exchanger, are potential approaches to reduce arrhythmias. New tools allow exploring these ideas. The principles underlying this approach and the first results are critically reviewed.


Subject(s)
Anti-Arrhythmia Agents/administration & dosage , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Drug Delivery Systems/methods , Ion Channel Gating/drug effects , Humans , Models, Cardiovascular
17.
Front Physiol ; 9: 1453, 2018.
Article in English | MEDLINE | ID: mdl-30374311

ABSTRACT

Heart failure (HF) is associated with elevated sympathetic tone and mechanical load. Both systems activate signaling transduction pathways that increase cardiac output, but eventually become part of the disease process itself leading to further worsening of cardiac function. These alterations can adversely contribute to electrical instability, at least in part due to the modulation of Ca2+ handling at the level of the single cardiac myocyte. The major aim of this review is to provide a definitive overview of the links and cross talk between ß-adrenergic stimulation, mechanical load, and arrhythmogenesis in the setting of HF. We will initially review the role of Ca2+ in the induction of both early and delayed afterdepolarizations, the role that ß-adrenergic stimulation plays in the initiation of these and how the propensity for these may be altered in HF. We will then go onto reviewing the current data with regards to the link between mechanical load and afterdepolarizations, the associated mechano-sensitivity of the ryanodine receptor and other stretch activated channels that may be associated with HF-associated arrhythmias. Furthermore, we will discuss how alterations in local Ca2+ microdomains during the remodeling process associated the HF may contribute to the increased disposition for ß-adrenergic or stretch induced arrhythmogenic triggers. Finally, the potential mechanisms linking ß-adrenergic stimulation and mechanical stretch will be clarified, with the aim of finding common modalities of arrhythmogenesis that could be targeted by novel therapeutic agents in the setting of HF.

18.
Front Physiol ; 9: 1108, 2018.
Article in English | MEDLINE | ID: mdl-30166973

ABSTRACT

Spontaneous Ca2+-release events (SCaEs) from the sarcoplasmic reticulum play crucial roles in the initiation of cardiac arrhythmias by promoting triggered activity. However, the subcellular determinants of these SCaEs remain incompletely understood. Structural differences between atrial and ventricular cardiomyocytes, e.g., regarding the density of T-tubular membrane invaginations, may influence cardiomyocyte Ca2+-handling and the distribution of cardiac ryanodine receptors (RyR2) has recently been shown to undergo remodeling in atrial fibrillation. These data suggest that the subcellular distribution of Ca2+-handling proteins influences proarrhythmic Ca2+-handling abnormalities. Here, we employ computational modeling to provide an in-depth analysis of the impact of variations in subcellular RyR2 and L-type Ca2+-channel distributions on Ca2+-transient properties and SCaEs in a human atrial cardiomyocyte model. We incorporate experimentally observed RyR2 expression patterns and various configurations of axial tubules in a previously published model of the human atrial cardiomyocyte. We identify an increased SCaE incidence for larger heterogeneity in RyR2 expression, in which SCaEs preferentially arise from regions of high local RyR2 expression. Furthermore, we show that the propagation of Ca2+ waves is modulated by the distance between RyR2 bands, as well as the presence of experimentally observed RyR2 clusters between bands near the lateral membranes. We also show that incorporation of axial tubules in various amounts and locations reduces Ca2+-transient time to peak. Furthermore, selective hyperphosphorylation of RyR2 around axial tubules increases the number of spontaneous waves. Finally, we present a novel model of the human atrial cardiomyocyte with physiological RyR2 and L-type Ca2+-channel distributions that reproduces experimentally observed Ca2+-handling properties. Taken together, these results significantly enhance our understanding of the structure-function relationship in cardiomyocytes, identifying that RyR2 and L-type Ca2+-channel distributions have a major impact on systolic Ca2+ transients and SCaEs.

19.
Eur J Pharmacol ; 818: 278-286, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29066415

ABSTRACT

Na+/Ca2+ exchanger (NCX) is the main Ca2+ transporter in cardiac myocytes. Its inhibition could be expected to exert positive inotropic action by accumulation of cytosolic Ca2+ ([Ca2+]i). However, we have observed only a marginal positive inotropic effect upon selective inhibition of NCX, which was enhanced when forward activity was facilitated. Here we attempted to clarify the underlying mechanism of the limited inotropic action of selective NCX inhibition by a novel inhibitor ORM-10962 on canine ventricular myocytes. 1µM ORM-10962 reduced the Ca2+ content of sarcoplasmic reticulum (SR) when the reverse NCX was favoured, while SR Ca2+ content was increased by ORM-10962 under conditions favouring the forward activity, like elevation of [Ca2+]i. L-type Ca2+ current (ICa) was not affected by 1µM ORM-10962 in the absence of SR Ca2+ release, while ICa was suppressed by ORM-10962 during normal Ca2+ cycling. The apparent degree of forward NCX inhibition was dependent on the elevation of [Ca2+]i, suggesting that an increased driving force of forward NCX can also limit the accumulation of [Ca2+i]. We concluded that in healthy myocardium the possible positive inotropic potential of NCX inhibition is considerably weaker than it was expected earlier by theoretical assumptions. The underlying mechanism may involve the autoregulation of Ca2+ handling and/or the preserved inducibility of forward NCX by high [Ca2+]i. This limitation of selective NCX inhibition seen in undiseased myocardium requires further studies in failing heart, which may allow correct evaluation of the potential therapeutic value of selective NCX inhibitors in the treatment of heart failure.


Subject(s)
Acetamides/pharmacology , Chromans/pharmacology , Heart Ventricles/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Piperidines/pharmacology , Sodium-Calcium Exchanger/antagonists & inhibitors , Animals , Calcium/metabolism , Dogs , Electrophysiological Phenomena/drug effects , Female , Male , Myocytes, Cardiac/cytology , Sarcoplasmic Reticulum/drug effects
20.
Heart Rhythm ; 15(9): 1328-1336, 2018 09.
Article in English | MEDLINE | ID: mdl-29803020

ABSTRACT

BACKGROUND: Arterial hypertension (HT) contributes to progression of atrial fibrillation (AF) via unknown mechanisms. OBJECTIVE: We aimed to characterize electrical and structural changes accounting for increased AF stability in a large animal model of rapid atrial pacing (RAP)-induced AF combined with desoxycorticosterone acetate (DOCA)-induced HT. METHODS: Eighteen pigs were instrumented with right atrial endocardial pacemaker leads and custom-made pacemakers to induce AF by continuous RAP (600 beats/min). DOCA pellets were subcutaneously implanted in a subgroup of 9 animals (AF+HT group); the other 9 animals served as controls (AF group). Final experiments included electrophysiology studies, endocardial electroanatomic mapping, and high-density mapping with epicardial multielectrode arrays. In addition, 3-dimensional computational modeling was performed. RESULTS: DOCA implantation led to secondary HT (median [interquartile range] aortic pressure 109.9 [100-137] mm Hg in AF+HT vs 82.2 [79-96] mm Hg in AF; P < .05), increased AF stability (55.6% vs 12.5% of animals with AF episodes lasting >1 hour; P < .05), concentric left ventricular hypertrophy, atrial dilatation (119 ± 31 cm2 in AF+HT vs 78 ± 23 cm2 in AF; P < .05), and fibrosis. Collagen accumulation in the AF+HT group was mainly found in non-intermyocyte areas (1.62 ± 0.38 cm3 in AF+HT vs 0.96 ± 0.3 cm3 in AF; P < .05). Left and right atrial effective refractory periods, action potential durations, endo- and epicardial conduction velocities, and measures of AF complexity were comparable between the 2 groups. A 3-dimensional computational model confirmed an increase in AF stability observed in the in vivo experiments associated with increased atrial size. CONCLUSION: In this model of secondary HT, higher AF stability after 2 weeks of RAP is mainly driven by atrial dilatation.


Subject(s)
Atrial Fibrillation/physiopathology , Atrial Remodeling , Blood Pressure/physiology , Computer Simulation , Heart Atria/physiopathology , Heart Rate/physiology , Hypertension/complications , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/therapy , Disease Models, Animal , Electrocardiography , Heart Atria/diagnostic imaging , Hypertension/physiopathology , Pacemaker, Artificial , Swine
SELECTION OF CITATIONS
SEARCH DETAIL