Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 155(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37191672

ABSTRACT

Connexin-43 (Cx43) is the most abundant protein forming gap junction channels (GJCs) in cardiac ventricles. In multiple cardiac pathologies, including hypertrophy and heart failure, Cx43 is found remodeled at the lateral side of the intercalated discs of ventricular cardiomyocytes. Remodeling of Cx43 has been long linked to spontaneous ventricular arrhythmia, yet the mechanisms by which arrhythmias develop are still debated. Using a model of dystrophic cardiomyopathy, we previously showed that remodeled Cx43 function as aberrant hemichannels (non-forming GJCs) that alter cardiomyocyte excitability and, consequently, promote arrhythmias. Here, we aim to evaluate if opening of remodeled Cx43 can serve as a general mechanism to alter cardiac excitability independent of cellular dysfunction associated with a particular cardiomyopathy. To address this issue, we used a genetically modified Cx43 knock-in mouse (S3A) that promotes cardiac remodeling of Cx43 protein without apparent cardiac dysfunction. Importantly, when S3A mice were subjected to cardiac stress using the ß-adrenergic agonist isoproterenol (Iso), they displayed acute and severe arrhythmias, which were not observed in WT mice. Pretreatment of S3A mice with the Cx43 hemichannel blocker, Gap19, prevented Iso-induced abnormal electrocardiographic behavior. At the cellular level, when compared with WT, Iso-treated S3A cardiomyocytes showed increased membrane permeability, greater plasma membrane depolarization, and Ca2+ overload, which likely caused prolonged action potentials, delayed after depolarizations, and triggered activity. All these cellular dysfunctions were also prevented by Cx43 hemichannel blockers. Our results support the notion that opening of remodeled Cx43 hemichannels, regardless of the type of cardiomyopathy, is sufficient to mediate cardiac-stress-induced arrhythmogenicity.


Subject(s)
Cardiomyopathies , Connexin 43 , Mice , Animals , Connexin 43/genetics , Connexin 43/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/metabolism , Gap Junctions , Ion Channels/metabolism , Isoproterenol
2.
Am J Physiol Heart Circ Physiol ; 323(5): H983-H995, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36206047

ABSTRACT

Dilated cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD), an inherited degenerative disease of the cardiac and skeletal muscle caused by absence of the protein dystrophin. We showed one hallmark of DMD cardiomyopathy is the dysregulation of cardiac gap junction channel protein connexin-43 (Cx43). Proper Cx43 localization and function at the cardiac intercalated disc (ID) is regulated by post-translational phosphorylation of Cx43-carboxy-terminus residues S325/S328/S330 (pS-Cx43). Concurrently, Cx43 traffics along microtubules (MTs) for targeted delivery to the ID. In DMD hearts, absence of dystrophin results in a hyperdensified and disorganized MT cytoskeleton, yet the link with pS-Cx43 remains unaddressed. To gain insight into the relationship between MTs and pS-Cx43, DMD mice (mdx) and pS-Cx43-deficient (mdxS3A) mice were treated with an inhibitor of MT polymerization, colchicine (Colch). Colch treatment protected mdx, not mdxS3A mice, against Cx43 remodeling, improved MT directionality, and enhanced pS-Cx43/tubulin interaction. Likewise, severe arrhythmias were prevented in isoproterenol-stressed mdx, not mdxS3A mice. Furthermore, MT directionality was improved in pS-Cx43-mimicking mdx (mdxS3E). Mdxutr+/- and mdxutr+/-S3A mice, lacking one copy of dystrophin homolog utrophin, displayed enhanced cardiac fibrosis and reduced lifespan compared with mdxutr+/-S3E; and Colch treatment corrected cardiac fibrosis in mdxutr+/- but not mdxutr+/-S3A. Collectively, the data suggest that improved MT directionality reduces Cx43 remodeling and that pS-Cx43 is necessary and sufficient to regulate MT organization, which plays crucial role in correcting cardiac dysfunction in DMD mice. Thus, identification of novel organizational mechanisms acting on pS-Cx43-MT will help develop novel cardioprotective therapies for DMD cardiomyopathy.NEW & NOTEWORTHY We found that colchicine administration to Cx43-phospho-deficient dystrophic mice fails to protect against Cx43 remodeling. Conversely, Cx43-phospho-mimic dystrophic mice display a normalized MT network. We envision a bidirectional regulation whereby correction of the dystrophic MTs leads to correction of Cx43 remodeling, which in turn leads to further correction of the MTs. Our findings suggest a link between phospho-Cx43 and MTs that provides strong foundations for novel therapeutics in DMD cardiomyopathy.


Subject(s)
Cardiomyopathies , Muscular Dystrophy, Duchenne , Mice , Animals , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Dystrophin/genetics , Connexin 43/genetics , Connexin 43/metabolism , Mice, Inbred mdx , Disease Models, Animal , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/prevention & control , Cardiomyopathies/genetics , Cardiomyopathies/prevention & control , Microtubules/metabolism , Colchicine , Fibrosis
3.
J Clin Invest ; 130(4): 1713-1727, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31910160

ABSTRACT

Aberrant expression of the cardiac gap junction protein connexin-43 (Cx43) has been suggested as playing a role in the development of cardiac disease in the mdx mouse model of Duchenne muscular dystrophy (DMD); however, a mechanistic understanding of this association is lacking. Here, we identified a reduction of phosphorylation of Cx43 serines S325/S328/S330 in human and mouse DMD hearts. We hypothesized that hypophosphorylation of Cx43 serine-triplet triggers pathological Cx43 redistribution to the lateral sides of cardiomyocytes (remodeling). Therefore, we generated knockin mdx mice in which the Cx43 serine-triplet was replaced with either phospho-mimicking glutamic acids (mdxS3E) or nonphosphorylatable alanines (mdxS3A). The mdxS3E, but not mdxS3A, mice were resistant to Cx43 remodeling, with a corresponding reduction of Cx43 hemichannel activity. MdxS3E cardiomyocytes displayed improved intracellular Ca2+ signaling and a reduction of NADPH oxidase 2 (NOX2)/ROS production. Furthermore, mdxS3E mice were protected against inducible arrhythmias, related lethality, and the development of cardiomyopathy. Inhibition of microtubule polymerization by colchicine reduced both NOX2/ROS and oxidized CaMKII, increased S325/S328/S330 phosphorylation, and prevented Cx43 remodeling in mdx hearts. Together, these results demonstrate a mechanism of dystrophic Cx43 remodeling and suggest that targeting Cx43 may be a therapeutic strategy for preventing heart dysfunction and arrhythmias in DMD patients.


Subject(s)
Calcium Signaling , Cardiomyopathies/metabolism , Connexin 43/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Connexin 43/genetics , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Microtubules/genetics , Microtubules/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism
4.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31751316

ABSTRACT

Patients with Duchenne muscular dystrophy (DMD) commonly present with severe ventricular arrhythmias that contribute to heart failure. Arrhythmias and lethality are also consistently observed in adult Dmdmdx mice, a mouse model of DMD, after acute ß-adrenergic stimulation. These pathological features were previously linked to aberrant expression and remodeling of the cardiac gap junction protein connexin43 (Cx43). Here, we report that remodeled Cx43 protein forms Cx43 hemichannels in the lateral membrane of Dmdmdx cardiomyocytes and that the ß-adrenergic agonist isoproterenol (Iso) aberrantly activates these hemichannels. Block of Cx43 hemichannels or a reduction in Cx43 levels (using Dmdmdx Cx43+/- mice) prevents the abnormal increase in membrane permeability, plasma membrane depolarization, and Iso-evoked electrical activity in these cells. Additionally, Iso treatment promotes nitric oxide (NO) production and S-nitrosylation of Cx43 hemichannels in Dmdmdx heart. Importantly, inhibition of NO production prevents arrhythmias evoked by Iso. We found that NO directly activates Cx43 hemichannels by S-nitrosylation of cysteine at position 271. Our results demonstrate that opening of remodeled and S-nitrosylated Cx43 hemichannels plays a key role in the development of arrhythmias in DMD mice and that these channels may serve as therapeutic targets to prevent fatal arrhythmias in patients with DMD .


Subject(s)
Arrhythmias, Cardiac/pathology , Connexin 43/metabolism , Muscular Dystrophy, Duchenne/complications , Myocytes, Cardiac/pathology , Adrenergic beta-Agonists/toxicity , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Membrane Permeability , Connexin 43/genetics , Cysteine/metabolism , Disease Models, Animal , Dystrophin/genetics , Evoked Potentials/drug effects , Humans , Isoproterenol/toxicity , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Duchenne/genetics , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Oocytes , Patch-Clamp Techniques , Xenopus laevis
5.
J Neurosci ; 39(31): 6067-6080, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31127000

ABSTRACT

The cold- and menthol-sensitive transient receptor potential melastatin 8 (TRPM8) channel is important for both physiological temperature detection and cold allodynia. Activation of G-protein-coupled receptors (GPCRs) by proinflammatory mediators inhibits these channels. It was proposed that this inhibition proceeds via direct binding of Gαq to the channel. TRPM8 requires the plasma membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2 or PIP2] for activity. However, it was claimed that a decrease in cellular levels of this lipid upon receptor activation does not contribute to channel inhibition. Here, we show that supplementing the whole-cell patch pipette with PI(4,5)P2 reduced inhibition of TRPM8 by activation of Gαq-coupled receptors in mouse dorsal root ganglion (DRG) neurons isolated from both sexes. Stimulating the same receptors activated phospholipase C (PLC) and decreased plasma membrane PI(4,5)P2 levels in these neurons. PI(4,5)P2 also reduced inhibition of TRPM8 by activation of heterologously expressed muscarinic M1 receptors. Coexpression of a constitutively active Gαq protein that does not couple to PLC inhibited TRPM8 activity, and in cells expressing this protein, decreasing PI(4,5)P2 levels using a voltage-sensitive 5'-phosphatase induced a stronger inhibition of TRPM8 activity than in control cells. Our data indicate that, upon GPCR activation, Gαq binding reduces the apparent affinity of TRPM8 for PI(4,5)P2 and thus sensitizes the channel to inhibition induced by decreasing PI(4,5)P2 levels.SIGNIFICANCE STATEMENT Increased sensitivity to heat in inflammation is partially mediated by inhibition of the cold- and menthol-sensitive transient receptor potential melastatin 8 (TRPM8) ion channels. Most inflammatory mediators act via G-protein-coupled receptors that activate the phospholipase C pathway, leading to the hydrolysis of phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. How receptor activation by inflammatory mediators leads to TRPM8 inhibition is not well understood. Here, we propose that direct binding of Gαq both reduces TRPM8 activity and sensitizes the channel to inhibition by decreased levels of its cofactor, PI(4,5)P2 Our data demonstrate the convergence of two downstream effectors of receptor activation, Gαq and PI(4,5)P2 hydrolysis, in the regulation of TRPM8.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , TRPM Cation Channels/metabolism , Animals , Female , Ganglia, Spinal/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL
6.
Neuromuscul Disord ; 28(4): 361-372, 2018 04.
Article in English | MEDLINE | ID: mdl-29477453

ABSTRACT

Duchenne muscular dystrophy (DMD) associated cardiomyopathy remains incurable. Connexin 43 (Cx43) is upregulated and remodeled in the hearts of mdx mice, a mouse model of DMD. Hearts from Wild Type, mdx, and mdx:Cx43(+/-) mice were studied before (4-6 months) and after (10-15 months) the onset of cardiomyopathy to assess the impact of decreasing Cx43 levels on cardiac pathology in dystrophic mice. Increased connexin 43 protein levels in mdx hearts were not observed in mdx:Cx43(+/-) hearts. Cx43 remodeling in mdx hearts was attenuated in mdx:Cx43(+/-) hearts. At time-point 4-6 months, isolated cardiomyocytes from mdx hearts displayed enhanced ethidium bromide uptake, augmented intracellular calcium signals and increased production of reactive oxygen species. These pathological features were improved in mdx:Cx43(+/-) cardiomyocytes. Isoproterenol-challenged mdx:Cx43(+/-) mice did not show arrhythmias or acute lethality observed in mdx mice. Likewise, isoproterenol-challenged mdx:Cx43(+/-) isolated hearts were also protected from arrhythmogenesis. At time-point 10-15 months, mdx:Cx43(+/-) mice showed decreased cardiac fibrosis and improved ventricular function, relative to mdx mice. These results suggest that normalization of connexin 43 protein levels in mdx mice reduces overall cardiac pathology.


Subject(s)
Calcium/metabolism , Cardiomyopathies/metabolism , Connexin 43/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Cardiomyopathies/pathology , Disease Models, Animal , Female , Male , Mice, Transgenic , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology
7.
Cardiovasc Res ; 114(1): 90-102, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29036556

ABSTRACT

Aims: Duchenne muscular dystrophy (DMD) is an inherited devastating muscle disease with severe and often lethal cardiac complications. Emerging evidence suggests that the evolution of the pathology in DMD is accompanied by the accumulation of mitochondria with defective structure and function. Here, we investigate whether defects in the housekeeping autophagic pathway contribute to mitochondrial and metabolic dysfunctions in dystrophic cardiomyopathy. Methods and results: We employed various biochemical and imaging techniques to assess mitochondrial structure and function as well as to evaluate autophagy, and specific mitochondrial autophagy (mitophagy), in hearts of mdx mice, an animal model of DMD. Our results indicate substantial structural damage of mitochondria and a significant decrease in ATP production in hearts of mdx animals, which developed cardiomyopathy. In these hearts, we also detected enhanced autophagy but paradoxically, mitophagy appeared to be suppressed. In addition, we found decreased levels of several proteins involved in the PINK1/PARKIN mitophagy pathway as well as an insignificant amount of PARKIN protein phosphorylation at the S65 residue upon induction of mitophagy. Conclusions: Our results suggest faulty mitophagy in dystrophic hearts due to defects in the PINK1/PARKIN pathway.


Subject(s)
Autophagy , Cardiomyopathies/enzymology , Mitochondria, Heart/enzymology , Mitophagy , Muscular Dystrophy, Duchenne/complications , Myocytes, Cardiac/enzymology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cellular Senescence , Disease Models, Animal , Mice, Inbred mdx , Microtubule-Associated Proteins/metabolism , Mitochondria, Heart/ultrastructure , Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/ultrastructure , Phosphorylation , Signal Transduction
8.
Stem Cells ; 35(3): 597-610, 2017 03.
Article in English | MEDLINE | ID: mdl-27734557

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by the loss of the protein dystrophin, leading to muscle fragility, progressive weakening, and susceptibility to mechanical stress. Although dystrophin-negative mdx mouse models have classically been used to study DMD, phenotypes appear mild compared to patients. As a result, characterization of muscle pathology, especially in the heart, has proven difficult. We report that injection of mdx embryonic stem cells (ESCs) into Wild Type blastocysts produces adult mouse chimeras with severe DMD phenotypes in the heart and skeletal muscle. Inflammation, regeneration and fibrosis are observed at the whole organ level, both in dystrophin-negative and dystrophin-positive portions of the chimeric tissues. Skeletal and cardiac muscle function are also decreased to mdx levels. In contrast to mdx heterozygous carriers, which show no significant phenotypes, these effects are even observed in chimeras with low levels of mdx ESC incorporation (10%-30%). Chimeric mice lack typical compensatory utrophin upregulation, and show pathological remodeling of Connexin-43. In addition, dystrophin-negative and dystrophin-positive isolated cardiomyocytes show augmented calcium response to mechanical stress, similar to mdx cells. These global effects highlight a novel role of mdx ESCs in triggering muscular dystrophy even when only low amounts are present. Stem Cells 2017;35:597-610.


Subject(s)
Aging/pathology , Chimera/metabolism , Embryonic Stem Cells/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myocardium/pathology , Animals , Calcium/metabolism , Connexin 43/metabolism , Dystrophin/metabolism , Female , Heart Function Tests , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Myocytes, Cardiac/metabolism , Regeneration
9.
Cardiovasc Res ; 108(3): 324-34, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26503985

ABSTRACT

AIMS: Nicotinamide adenine dinucleotide oxidases (NOXs) are important contributors to cellular oxidative stress in the cardiovascular system. The NOX2 isoform is upregulated in numerous disorders, including dystrophic cardiomyopathy, where it drives the progression of the disease. However, mechanisms underlying NOX2 overexpression are still unknown. We investigated the role of microRNAs (miRs) in the regulation of NOX2 expression. METHODS AND RESULTS: Duchenne muscular dystrophy (DMD) was used as a model of cardiomyopathy. After screening with miRNA target prediction databases and following qRT-PCR analysis, we found drastic downregulation of miR-448-3p in hearts of mdx mice, an animal model of DMD. The downregulation correlated with overexpression of the Ncf1 gene, encoding the NOX2 regulatory subunit p47(phox). Specificity of Ncf1 targeting by miR-448-3p was validated by luciferase reporter assay. Silencing of miR-448-3p in wild-type mice had a dramatic effect on cellular and functional properties of cardiac muscle as assessed by western blotting, qRT-PCR, confocal imaging, echocardiography, and histology. Acute treatment of mice with LNA-miR-448 inhibitors led to increased Ncf1 expression, abnormally elevated reactive oxygen species (ROS) production and exacerbated Ca(2+) signalling in cardiomyocytes, reminiscent of features previously observed in dystrophic cardiac cells. In addition, chronic inhibition of miR-448-3p resulted in dilated cardiomyopathy and arrhythmia, hallmarks of dystrophic cardiomyopathy. CONCLUSIONS: Our studies suggest that downregulation of miR-448-3p leads to the increase in the expression of Ncf1 gene and p47(phox) protein, as well as to the substantial increase in NOX2-derived ROS production. Cellular oxidative stress subsequently triggers events that finally culminate in cardiac tissue damage and development of cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/enzymology , MicroRNAs/metabolism , Myocardium/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Calcium Signaling , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Silencing , Genetic Predisposition to Disease , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred mdx , MicroRNAs/genetics , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Myocardium/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phenotype , Time Factors , Transfection , Ventricular Remodeling
10.
Cell Calcium ; 58(2): 186-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25975620

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive muscle disease with severe cardiac complications. It is believed that cellular oxidative stress and augmented Ca(2+) signaling drives the development of cardiac pathology. Some mitochondrial and metabolic dysfunctions have also been reported. Here we investigate cellular mechanisms responsible for impaired mitochondrial metabolism in dystrophic cardiomyopathy at early stages of the disease. We employed electrophysiological and imaging techniques to study mitochondrial structure and function in cardiomyocytes from mdx mice, an animal model of DMD. Here we show that mitochondrial matrix was progressively oxidized in myocytes isolated from mdx mice. Moreover, an abrupt increase in workload resulted in significantly more pronounced oxidation of mitochondria in dystrophic cells. Electron micrographs revealed a gradually increased number of damaged mitochondria in mdx myocytes. Degradation in mitochondrial structure was correlated with progressive increase in mitochondrial Ca(2+) sequestration and mitochondrial depolarization, despite a substantial and persistent elevation in resting cytosolic sodium levels. Treatment of mdx cells with cyclosporine A, an inhibitor of mitochondrial permeability transition pore (mPTP), shifted both resting and workload-dependent mitochondrial redox state to the levels recorded in control myocytes. It also significantly reduced workload dependent depolarization of mitochondrial membrane in dystrophic cardiomyocytes. Overall, our studies highlight age dependent deterioration of mitochondrial function in dystrophic cardiomyocytes, which seems to be associated with excessive opening of mPTP due to oxidative stress and cellular Ca(2+) overload.


Subject(s)
Mitochondria, Heart/metabolism , Muscular Dystrophy, Duchenne/pathology , Animals , Calcium/metabolism , Cells, Cultured , Cyclosporine/pharmacology , Cytosol/metabolism , Disease Models, Animal , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NAD/metabolism , Oxidative Stress/drug effects , Sodium/metabolism
11.
Biophys J ; 107(12): 2815-2827, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25517148

ABSTRACT

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.


Subject(s)
Calcium Signaling , Myocytes, Cardiac/metabolism , Oxidative Stress , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
12.
Biochim Biophys Acta ; 1833(4): 866-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22960642

ABSTRACT

In cardiac muscle, a number of posttranslational protein modifications can alter the function of the Ca(2+) release channel of the sarcoplasmic reticulum (SR), also known as the ryanodine receptor (RyR). During every heartbeat RyRs are activated by the Ca(2+)-induced Ca(2+) release mechanism and contribute a large fraction of the Ca(2+) required for contraction. Some of the posttranslational modifications of the RyR are known to affect its gating and Ca(2+) sensitivity. Presently, research in a number of laboratories is focused on RyR phosphorylation, both by PKA and CaMKII, or on RyR modifications caused by reactive oxygen and nitrogen species (ROS/RNS). Both classes of posttranslational modifications are thought to play important roles in the physiological regulation of channel activity, but are also known to provoke abnormal alterations during various diseases. Only recently it was realized that several types of posttranslational modifications are tightly connected and form synergistic (or antagonistic) feed-back loops resulting in additive and potentially detrimental downstream effects. This review summarizes recent findings on such posttranslational modifications, attempts to bridge molecular with cellular findings, and opens a perspective for future work trying to understand the ramifications of crosstalk in these multiple signaling pathways. Clarifying these complex interactions will be important in the development of novel therapeutic approaches, since this may form the foundation for the implementation of multi-pronged treatment regimes in the future. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Subject(s)
Calcium Signaling/physiology , Excitation Contraction Coupling/physiology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Ion Channel Gating/physiology , Myocytes, Cardiac/cytology , Phosphorylation , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism
13.
J Mol Cell Cardiol ; 58: 217-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23261966

ABSTRACT

Dilated cardiomyopathy is a serious and almost inevitable complication of Duchenne Muscular Dystrophy, a devastating and fatal disease of skeletal muscle resulting from the lack of functional dystrophin, a protein linking the cytoskeleton to the extracellular matrix. Ultimately, it leads to congestive heart failure and arrhythmias resulting from both cardiac muscle fibrosis and impaired function of the remaining cardiomyocytes. Here we summarize findings obtained in several laboratories, focusing on cellular mechanisms that result in degradation of cardiac functions in dystrophy.


Subject(s)
Cardiomyopathy, Dilated/genetics , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/pathology , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/physiopathology , Dystrophin/deficiency , Dystrophin/metabolism , Heart Failure/complications , Heart Failure/metabolism , Heart Failure/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/physiopathology , Myocytes, Cardiac/metabolism
14.
Cardiovasc Res ; 97(4): 666-75, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23263329

ABSTRACT

AIMS: Duchenne muscular dystrophy (DMD) is a muscle disease with serious cardiac complications. Changes in Ca(2+) homeostasis and oxidative stress were recently associated with cardiac deterioration, but the cellular pathophysiological mechanisms remain elusive. We investigated whether the activity of ryanodine receptor (RyR) Ca(2+) release channels is affected, whether changes in function are cause or consequence and which post-translational modifications drive disease progression. METHODS AND RESULTS: Electrophysiological, imaging, and biochemical techniques were used to study RyRs in cardiomyocytes from mdx mice, an animal model of DMD. Young mdx mice show no changes in cardiac performance, but do so after ∼8 months. Nevertheless, myocytes from mdx pups exhibited exaggerated Ca(2+) responses to mechanical stress and 'hypersensitive' excitation-contraction coupling, hallmarks of increased RyR Ca(2+) sensitivity. Both were normalized by antioxidants, inhibitors of NAD(P)H oxidase and CaMKII, but not by NO synthases and PKA antagonists. Sarcoplasmic reticulum Ca(2+) load and leak were unchanged in young mdx mice. However, by the age of 4-5 months and in senescence, leak was increased and load was reduced, indicating disease progression. By this age, all pharmacological interventions listed above normalized Ca(2+) signals and corrected changes in ECC, Ca(2+) load, and leak. CONCLUSION: Our findings suggest that increased RyR Ca(2+) sensitivity precedes and presumably drives the progression of dystrophic cardiomyopathy, with oxidative stress initiating its development. RyR oxidation followed by phosphorylation, first by CaMKII and later by PKA, synergistically contributes to cardiac deterioration.


Subject(s)
Cardiomyopathies/metabolism , Muscular Dystrophy, Duchenne/complications , Protein Processing, Post-Translational , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Disease Progression , Dystrophin/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Myocardium/pathology , Myocytes, Cardiac/metabolism , NADPH Oxidases/physiology , Oxidation-Reduction , Reactive Oxygen Species/metabolism
15.
J Biol Chem ; 288(4): 2103-9, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23223241

ABSTRACT

Functional coupling between inositol (1,4,5)-trisphosphate receptor (IP(3)R) and ryanodine receptor (RyR) represents a critical component of intracellular Ca(2+) signaling in many excitable cells; however, the role of this mechanism in skeletal muscle remains elusive. In skeletal muscle, RyR-mediated Ca(2+) sparks are suppressed in resting conditions, whereas application of transient osmotic stress can trigger activation of Ca(2+) sparks that are restricted to the periphery of the fiber. Here we show that onset of these spatially confined Ca(2+) sparks involves interaction between activation of IP(3)R and RyR near the sarcolemmal membrane. Pharmacological prevention of IP(3) production or inhibition of IP(3)R channel activity abolishes stress-induced Ca(2+) sparks in skeletal muscle. Although genetic ablation of the type 2 IP(3)R does not appear to affect Ca(2+) sparks in skeletal muscle, specific silencing of the type 1 IP(3)R leads to ablation of stress-induced Ca(2+) sparks. Our data indicate that membrane-delimited signaling involving cross-talk between IP(3)R1 and RyR1 contributes to Ca(2+) spark activation in skeletal muscle.


Subject(s)
Calcium/metabolism , Gene Expression Regulation , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/chemistry , Calcium Signaling , Mice , Microscopy, Confocal/methods , Models, Biological , Models, Genetic , Osmosis , Patch-Clamp Techniques , Plasmids/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
16.
Am J Physiol Heart Circ Physiol ; 297(6): H1992-2003, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19783774

ABSTRACT

Duchenne muscular dystrophy represents a severe inherited disease of striated muscle. It is caused by a mutation of the dystrophin gene and characterized by a progressive loss of skeletal muscle function. Most patients also develop a dystrophic cardiomyopathy, resulting in dilated hypertrophy and heart failure, but the cellular mechanisms leading to the deterioration of cardiac function remain elusive. In the present study, we tested whether defective excitation-contraction (E-C) coupling contributes to impaired cardiac performance. "E-C coupling gain" was determined in cardiomyocytes from control and dystrophin-deficient mdx mice. To this end, L-type Ca2+ currents (ICaL) were measured with the whole cell patch-clamp technique, whereas Ca2+ transients were simultaneously recorded with confocal imaging of fluo-3. Initial findings indicated subtle changes of E-C coupling in mdx cells despite matched Ca2+ loading of the sarcoplasmic reticulum (SR). However, lowering the extracellular Ca2+ concentration, a maneuver used to unmask latent E-C coupling problems, was surprisingly much better tolerated by mdx myocytes, suggesting a hypersensitive E-C coupling mechanism. Challenging the SR Ca2+ release by slow elevations of the intracellular Ca2+ concentration resulted in Ca2+ oscillations after a much shorter delay in mdx cells. This is consistent with an enhanced Ca2+ sensitivity of the SR Ca2+-release channels [ryanodine receptors (RyRs)]. The hypersensitivity could be normalized by the introduction of reducing agents, indicating that the elevated cellular ROS generation in dystrophy underlies the abnormal RyR sensitivity and hypersensitive E-C coupling. Our data suggest that in dystrophin-deficient cardiomyocytes, E-C coupling is altered due to potentially arrhythmogenic changes in the Ca2+ sensitivity of redox-modified RyRs.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Cardiomyopathy, Dilated/etiology , Muscular Dystrophy, Duchenne/complications , Myocardial Contraction , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Disease Progression , Free Radical Scavengers/pharmacology , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Membrane Potentials , Mice , Mice, Inbred mdx , Microscopy, Confocal , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Patch-Clamp Techniques , Reactive Oxygen Species/metabolism , Reducing Agents/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/drug effects , Sodium/metabolism , Time Factors
17.
Pflugers Arch ; 458(5): 915-28, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19387681

ABSTRACT

Muscular dystrophies are among the most severe inherited muscle diseases. The genetic defect is a mutation in the gene for dystrophin, a cytoskeletal protein which protects muscle cells from mechanical damage. Mechanical stress, applied as osmotic shock, elicits an abnormal surge of Ca(2+) spark-like events in skeletal muscle fibers from dystrophin deficient (mdx) mice. Previous studies suggested a link between changes in the intracellular redox environment and appearance of Ca(2+) sparks in normal mammalian skeletal muscle. Here, we tested whether the exaggerated Ca(2+) responses in mdx fibers are related to oxidative stress. Localized intracellular and mitochondrial Ca(2+) transients, as well as ROS production, were assessed with confocal microscopy. The rate of basal cellular but not mitochondrial ROS generation was significantly higher in mdx cells. This difference was abolished by pre-incubation of mdx fibers with an inhibitor of NAD(P)H oxidase. In addition, immunoblotting showed a significantly stronger expression of NAD(P)H oxidase in mdx muscle, suggesting a major contribution of this enzyme to oxidative stress in mdx fibers. Osmotic shock produced an abnormal and persistent Ca(2+) spark activity, which was suppressed by ROS-reducing agents and by inhibitors of NAD(P)H oxidase. These Ca(2+) signals resulted in mitochondrial Ca(2+) accumulation in mdx fibers and an additional boost in cellular and mitochondrial ROS production. Taken together, our results indicate that the excessive ROS production and the simultaneous activation of abnormal Ca(2+) signals amplify each other, finally culminating in a vicious cycle of damaging events, which may contribute to the abnormal stress sensitivity in dystrophic skeletal muscle.


Subject(s)
Calcium Signaling/physiology , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Calcium Signaling/drug effects , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Free Radicals/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria/metabolism , Muscular Dystrophy, Duchenne/physiopathology , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Osmotic Pressure/physiology , Reactive Nitrogen Species/metabolism
18.
Methods ; 46(3): 183-93, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18848990

ABSTRACT

The ryanodine receptors (RyRs) are intracellular Ca2+ release channels of the sarcoplasmic reticulum (SR) involved in many cellular responses, including muscle excitation-contraction coupling. Multiple biochemical and biophysical methods are available to study RyR functions. However, most of them are somewhat limited because they can only be used to examine channels which are purified from the SR and no longer in their natural environment. In this review we discuss optical methods for studying RyR functions in situ. We describe several techniques for the investigation of local (microscopic) intracellular Ca2+ signals (a.k.a Ca2+ sparks) by means of confocal microscopy and flash photolysis of caged compounds. We discuss how these studies can and will continue to contribute to our understanding of RyR function in physiological and pathological conditions.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Acetates/pharmacology , Algorithms , Animals , Calcium Signaling/drug effects , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Ethylenediamines/pharmacology , Hydroquinones/pharmacology , Microscopy, Confocal/methods , Muscle, Skeletal/cytology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques/methods , Phenylacetates/pharmacology , Sarcoplasmic Reticulum/metabolism
19.
Cell ; 133(1): 53-65, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18394989

ABSTRACT

Mice with a malignant hyperthermia mutation (Y522S) in the ryanodine receptor (RyR1) display muscle contractures, rhabdomyolysis, and death in response to elevated environmental temperatures. We demonstrate that this mutation in RyR1 causes Ca(2+) leak, which drives increased generation of reactive nitrogen species (RNS). Subsequent S-nitrosylation of the mutant RyR1 increases its temperature sensitivity for activation, producing muscle contractures upon exposure to elevated temperatures. The Y522S mutation in humans is associated with central core disease. Many mitochondria in the muscle of heterozygous Y522S mice are swollen and misshapen. The mutant muscle displays decreased force production and increased mitochondrial lipid peroxidation with aging. Chronic treatment with N-acetylcysteine protects against mitochondrial oxidative damage and the decline in force generation. We propose a feed-forward cyclic mechanism that increases the temperature sensitivity of RyR1 activation and underlies heat stroke and sudden death. The cycle eventually produces a myopathy with damaged mitochondria.


Subject(s)
Death, Sudden/etiology , Heat Stroke/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Hot Temperature , Humans , Malignant Hyperthermia/metabolism , Mice , Mitochondria/metabolism , Muscle, Skeletal/pathology , Nitrosation , Oxidative Stress , Reactive Nitrogen Species , Reactive Oxygen Species
20.
J Physiol ; 586(1): 197-210, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17974587

ABSTRACT

Ca(2+) sparks, localized elevations in cytosolic [Ca(2+)], are rarely detected in intact adult mammalian skeletal muscle under physiological conditions. However, they have been observed in permeabilized cells and in intact fibres subjected to stresses, such as osmotic shock and strenuous exercise. Our previous studies indicated that an excess in cellular reactive oxygen species (ROS) generation over the ROS scavenging capabilities could be one of the up-stream causes of Ca(2+) spark appearance in permeabilized muscle fibres. Here we tested whether the cytosolic ROS balance is compromised in intact skeletal muscle fibres that underwent osmotic shock and whether this misbalance contributes to unmasking Ca(2+) sparks. Spontaneous Ca(2+) sparks and the rate of ROS generation were assessed with single photon confocal microscopy and fluorescent indicators fluo-4, CM-H(2)DCFDA and MitoSOX Red. Osmotic shock produced spontaneous Ca(2+) sparks and a concomitant significant increase in ROS production. Preincubation of muscle cells with ROS scavengers (e.g. MnTBAP, Mn-cpx 3, TIRON) nearly eliminated Ca(2+) sparks. In addition, inhibitors of NAD(P)H oxidase (DPI and apocynin) significantly reduced ROS production and suppressed the appearance of Ca(2+) sparks. Taken together, the data suggest that ROS contribute to the abnormal Ca(2+) spark activity in mammalian skeletal muscle subjected to osmotic stress and also indicate that NAD(P)H oxidase is a possible source of ROS. We propose that ROS-dependent Ca(2+) sparks are an important component of adaptive/maladaptive muscle responses under various pathological conditions such as eccentric stretch, osmotic changes during ischaemia and reperfusion, and some muscle diseases.


Subject(s)
Calcium Signaling/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Reactive Oxygen Species/metabolism , Animals , Calcium/metabolism , Free Radical Scavengers/pharmacology , Male , Mice , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , NADPH Oxidases/metabolism , Osmotic Pressure , Reactive Nitrogen Species/metabolism , Xanthine Oxidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL