Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jan 18.
Published in final edited form as: Circ Res. 2013 Jan 18;112(2):393–405. doi: 10.1161/CIRCRESAHA.111.300496

OXIDATIVE STRESS AND SARCOMERIC PROTEINS

Susan F Steinberg 1
PMCID: PMC3595003  NIHMSID: NIHMS437748  PMID: 23329794

Abstract

Oxidative stress accompanies a wide spectrum of clinically important cardiac disorders, including ischemia/reperfusion, diabetes, and hypertensive heart disease. While reactive oxygen species (ROS) can activate signaling pathways that contribute to ischemic preconditioning and cardioprotection, high levels of ROS induce structural modifications of the sarcomere that impact on pump function and the pathogenesis of heart failure. However, the precise nature of the redox-dependent change in contractility is determined by the source/identity of the oxidant species, the level of oxidative stress, and the chemistry/position of oxidant-induced post-translational modifications on individual proteins within the sarcomere. This review focuses on various ROS-induced post-translational modifications of myofilament proteins (including direct oxidative modifications of myofilament proteins, myofilament protein phosphorylation by ROS-activated signaling enzymes, and myofilament protein cleavage by ROS-activated proteases) that have been implicated in the control of cardiac contractility.

Keywords: Oxidative stress, sarcomere, protein kinase, protease, contraction

INTRODUCTION

Reactive oxygen species (ROS) production increases in the context of many cardiac disorders. ROS-activated mechanisms that contribute to ischemic preconditioning are cardioprotective. However, high levels of ROS production that overwhelm cellular antioxidant defense systems generally produce deleterious changes in contractile performance and lead to adverse cardiac remodeling. Some cardiodepressive actions of ROS have been attributed to the activation of signaling pathways that influence the expression, phosphorylation, or function of calcium regulatory proteins (such as sarcoplasmic reticular Ca-ATPase 2a and ryanodine receptor 2), leading to changes in the magnitude or timing of the calcium transient and an inadequate calcium-induced contractile response 13. However, oxidative stress-induced modifications of contractile proteins - that are not associated with changes in intracellular calcium homeostasis - also may contribute to contractile dysfunction and the evolution of heart failure. This review focuses on ROS-induced structural modifications of the sarcomere – due to direct oxidative modifications of myofilament proteins, myofilament protein phosphorylation by ROS-activated kinases, or myofilament protein cleavage by ROS-activated proteases – that interfere with the transduction of calcium-dependent contractile responses. Modifications that are not the obvious or direct target of a ROS-regulated processes are beyond the scope of this review and are not covered.

Oxidative modifications of sarcomeric proteins

The contractile apparatus consists of a parallel array of interdigitating thick and thin filaments that form the molecular motor that powers cardiac contraction (Fig 1). The thin filament backbone is made up of two helical strands of actin monomers, the elongated tropomyosin molecule that associates end-to-end to form a continuous strand along the actin filament, and troponin complexes (consisting of the calcium-binding cTnC subunit, the inhibitory cTnI subunit, and the tropomyosin-binding cTnT subunit) positioned every 7th actin monomer along the thin filament. The thick filament is formed by two myosin heavy chain (MHC) molecules complexed with two molecules of myosin light chain 1 (MLC-1 or essential light chain) and two molecules of myosin light chain 2 (MLC-2 or regulatory light chain). The smaller light chain proteins are positioned at the myosin lever arm, between the rod portion of the molecule that forms the thick filament backbone and the head region that contains the actin- and nucleotide-binding sites. Cardiac contraction is powered by cyclical interactions between the myosin motor and actin-containing thin filaments – with additional regulation provided by cardiac myosin binding protein-C (cMyBP-C), a large multidomain thick filament protein located in the C-zone of the sarcomere. Titin - the third giant filament protein - runs from the Z disc to the M-band at the center of the sarcomere. Titin plays a role in the structural organization and assembly of myofibrillar proteins and it functions as a molecular scaffold to recruit signaling molecules that influence mechanotransduction. Titin also contains three serially-linked spring-like segments in an elastic I-band (the immunoglobulin-like domains, a Proline, Glutamate, Valine, and Lysine rich PEVK element, and an N2B element) that control the passive tension of the heart. The extensible elements in titin’s I-band region are targets for molecular events (isoform splicing and post-translational modifications) that ‘fine-tune’ titin’s elasticity.

Fig 1.

Fig 1

Schematic showing the arrangement of the major contractile and regulatory proteins in the sarcomere.

Oxidative stress and increased formation of ROS (or reactive nitrogen species) can result in direct chemical oxidation (or nitrosylation) of many contractile proteins, leading to changes in their structural conformation and/or functional activity. Protein oxidation or nitrosylation generally occurs at reactive thiol moieties in cysteine (or to a lesser extent methionine) residues. The reactivity of any particular cysteine residue is determined by the pKa of its thiol moiety; cysteine residues adjacent to basic amino acids such as Arg or Lys, aromatic amino acids, or metal centers have a relatively low pKa (<6.5), are prone to deprotonate, and tend to be more susceptible to oxidation (Fig 2). The reaction between the cysteine thiolate anion and H2O2 results in the formation of sulfenic acid (R-SOH), a relatively unstable structure that typically reacts with other thiol groups to form intra- or intermolecular disulfide bonds. A reaction between the cysteine thiolate anion and NO (S-nitrosylation) or glutathione (S-glutathionylation) leads to the formation of mixed disulfides. The formation of mixed disulfides prevents further more irreversible peroxidation of sulfenic acid to more highly oxidized sulfinic (R-SO2H) or sulfonic (R-SO3H) acid species - that typically are more disruptive to protein structure and function. Other residues also can be targets for oxidative modification. When oxidative stress and superoxide formation is associated with increased formation of NO (for example during early post-ischemic reperfusion or in the context of inflammation, where proinflammatory cytokines increase the expression of inducible nitric oxide synthase 4, 5), a near diffusion-limited reaction results in the formation of peroxynitrite (ONOO-); peroxynitrite is a highly reactive compound that promotes protein tyrosine nitration, the addition of a nitro group (NO2) to the 3-position of the tyrosine phenolic ring. Peroxynitrite also can oxidize Cys residues and promote protein carbonylation, the addition of a carbonyl group to susceptible Lys, Arg, or Pro residues.

Fig 2.

Fig 2

Major redox modifications of cysteine and tyrosine side chains.

Cardiac contraction typically is reduced following treatment with oxidizing agents such as superoxide anion or H2O2 (the more stable reactive species formed endogenously through spontaneous or SOD-catalyzed dismutation of superoxide). Early studies in chemically “skinned” rat cardiac muscle fibers showed that superoxide anion depresses maximal calcium-activated force without changing calcium-sensitivity or influencing rigor contracture in ATP-free solutions 6. These results were interpreted as evidence that superoxide anion acts in a very specific manner to alter some aspect of cross-bridge cycling (rather than some more nonspecific mechanism, for example a proteolytic event that disrupts the structural integrity of the sarcomere). Initial attempts to expose mechanism showed that H2O2 treatment of isolated rat heart leads to the oxidation of thin filament proteins - both cysteinyl oxidation of tropomyosin and cysteinyl oxidation/carbonylation of actin 7, 8. Oxidative modifications of tropomyosin also have been detected - in association with the development of contractile dysfunction - in ischemic microembolized pig hearts and in the early post-myocardial infarction period in mouse hearts 9, 10. Oxidative modifications of actin (and protein kinase C-α) are detected during reperfusion of ischemic rat hearts 11. There is evidence that oxidative modifications of cardiac tropomyosin (at its single cysteine residue at position 190) leads to the formation of dimers that alter tropomyosin’s flexibility and interfere with tropomyosin’s interactions with other thin filament proteins. While some investigators have argued that these structural events contribute to oxidative stress- and heart-failure-dependent changes in contractility 12, this formulation ignores the many other ROS-dependent modifications of sarcomeric proteins that are detected in end stage human heart failure, that correlate with contractile dysfunction, and also may be contributory 8.

Peroxynitrite also decreases maximal force development of the intact heart and it decreases contractility in isolated human ventricular myocytes. Some of the cardiodepressant actions of peroxynitrite have been attributed to an increase in cGMP and the activation of a protein kinase G- (PKG-) dependent pathway that decreases myofilament responsiveness to calcium 13, 14. However, peroxynitrite decreases force generation in isolated rat cardiac trabeculae in association with the nitration of MHC (and the myofibrillar isoform of creatine kinase, an additional target for a post-translational modification that impairs contractility by disrupting myofibrillar energetic mechanisms 15, 16). In vitro studies performed on purified MHC show that peroxynitrite promotes myosin nitration, cysteine oxidation and/or carbonylation at several highly-reactive solvent-exposed sites in the catalytic subfragment-1 (S1) globular head region 17. Functional studies suggest that these redox-induced post-translational modifications (in particular, myosin oxidation at Cys707/Cys697 and myosin carbonylation at Lys84, which sits at a domain interface in close proximity to the reactive Cys707/Cys697 residues) lead to a partial unfolding of the myosin subfragment-1, enhanced susceptibility to proteolytic cleavage by trypsin, and changes in Mg-ATPase activity (both increased intrinsic Mg2+-ATPase activity and decreased actin-stimulated Mg2+-ATPase activity 15, 1719). However, there is reason to interpret the results of studies performed on purified myosin preparations in solution with caution, since some oxidative modifications of myosin (for example Cys707 oxidation) are not detected in more physiologically relevant preparations (i.e., in isolated cardiac myofibrils), where incorporation of myosin into the myofilament lattice leads to decreased cysteine reactivity 20, 21. In this regard, studies in an aging rat heart model identify myosin nitration at Tyr114, Tyr116, Tyr134, and Tyr142 and pharmacologic studies suggest that peroxynitrite decreases force generation by increasing myosin carbonylation; these studies conclude that nitration is not contributory and cysteine oxidation may actually be protective, since cysteine residues might act as peroxynitrite scavengers and prevent the peroxynitrite-induced modifications elsewhere in the protein that disrupt functional activity 17, 22). The singular focus on myosin as the primary target of oxidative modifications also may be misguided, since ischemia/reperfusion injury leads to a decrease in maximum force per cross-sectional area and a decrease in rate of tension redevelopment in association with S-glutathionylation of actin in rat heart 23; pro-oxidants such as glutathione+H2O2 or glutathione+diamide induce a high level of α-actin (not myosin) S-glutathionylation in isolated human cardiac myofibrils 20. Actin S-glutathionylation at Cys374 (a site at the physiologically labile C-terminus) slows the kinetics of α-actin polymerization in vitro, destabilizes actin filaments in vivo, influences actin’s role as a myosin binding partner in the sarcomere, and decreases contractility; substitution of a glutathionylated form of actin for unmodified actin decreases maximal actomyosin-S1 ATPase activity 24.

Oxidative modifications of other sarcomeric proteins also have been identified. Peroxynitrite treatment or aging has been linked to increased nitration of tyrosine residues in a range of sarcomeric proteins, including cardiac troponin I (cTnI), cardiac troponin T (cTnT), MHC, myosin light chain, tropomyosin, cMyBP-C, actin desmin, and α-actinin 22, 25,26. Studies in human cardiomyocytes link α-actinin nitration to changes in cellular ultrastructure (disruption of the myofibrillar cross-striation pattern) and a defect in contractile function (reduced isometric force generation) 13. The less compliant titin N2B isoform also has been characterized as a redox-sensor. Titin is co-expressed in the heart as N2BA and N2B isoforms that arise through alternative splicing of the transcript of a single gene. The principal difference between titin N2BA and N2B isoforms is in the length of their elastic I-band spring segment; N2B has a relatively short I-band segment and is very stiff, whereas N2BA has a longer I-band region and is more compliant. The shorter titin N2B isoform contains six cysteine residues that form one or more disulfide bonds under oxidizing conditions; disulfide bonding decreases the extensibility of N2B and leads to an increase cardiac stiffness 27.

While oxidizing agents such as H2O2, superoxide, or peroxynitrite typically reduce force generation in skinned muscle preparations, nitroxyl (HNO, an electron reduction product of NO that displays very distinct chemistry and reactivity) acts in an antithetical fashion to increase force generation by increasing myofilament calcium sensitivity 28. HNO reacts chiefly with cysteine thiols, forming either a N-hydroxlsulfenamide (RSNHOH) or (if there is a second cysteine in close proximity) inter- or intramolecular disulfide bonds (Fig 2). A recent study mapped HNO-dependent redox modifications in the sarcomere to strategically located cysteine thiols in actin, tropomyosin, MHC, and MLC1. The HNO-dependent formation of actin-tropomyosin dimers (due to disulfide bridging between Cys257 in actin and Cys190 in tropomyosin) is predicted to tether tropomyosin to a position that is more permissive for Ca2+-induced myofilament activation, thereby increasing contractility. The HNO-dependent formation of dimers between MHC and Cys81 in MLC1 is predicted to enhance myofilament calcium sensitivity and would also improve cardiac contractility 29. These recently identified redox-dependent modifications of myofilament proteins that enhance force generation represent promising targets for novel classes of inotropic agents that could be developed for the therapy of heart failure.

REDOX-REGULATION OF MYOFILAMENT PROTEIN PHOSPHORYLATION

Cardiac contraction must be dynamically regulated on a beat-to-beat basis to accommodate to changes in hemodynamic load and to respond to neurohumeral stresses. Much of this control is accomplished by signal-regulated protein kinases (or phosphatases) that regulate the phosphorylation state of strategically located Ser or Thr residues in various myofilament proteins (i.e., myofibrillar protein phosphorylation is almost exclusively on Ser/Thr - and not Tyr - residues). Of note, many protein kinases that contribute to mechanical or neurohumoral control of cardiac contraction also are regulated by oxidative stress. This section focuses on phosphorylation events on the thin filament proteins cTnI and cTnT, the thick filament accessory protein cMyBP-C, and titin that are targets for redox-regulated enzymes.

Redox-regulation of thin filament protein phosphorylation

cTnI is the inhibitory component of the troponin complex that functions to ‘fine-tune’ myofilament function to hemodynamic load; cTnI contains three well described phosphorylation clusters at Ser23/Ser24, Ser43/Ser45, and Thr144. cTnI phosphorylation at Ser23/Ser24 (in the N-terminal region unique to cardiac TnI) is generally attributed to the beta-adrenergic receptor pathway involving protein kinase A (PKA) 30. cTnI-Ser23/Ser24 phosphorylation accelerates the off-rate for calcium binding to cTnC, leading to a faster rate of cardiac relaxation (which is crucial to accommodate the beta-adrenergic receptor dependent positive chronotropic response). PKA is a heterotetramer enzyme consisting of two catalytic (C) subunits that are maintained in an inactive conformation by two cAMP-binding regulatory (R) subunits. cAMP activates PKA by binding to the R subunits; this interaction leads to the dissociation of the enzyme complex and frees the C subunit to phosphorylate target substrates. PKA holoenzymes are classified as type I or II based upon the identity of the R subunit (RI or RII) in the enzyme complex. Cardiomyocytes co-express both PKAI and PKAII enzymes that display distinct biochemical properties and subcellular localization patterns; PKAII is primarily recovered in the particulate cell fraction (in association with membrane scaffolding proteins, or A-kinase Anchoring Proteins [AKAPs]) whereas the type I PKA holoenzyme is recovered primarily as a cytosolic enzyme. While RI and RII subunits share similar domain organization, there is genetic and biochemical evidence that RI and RII are not functionally non-redundant. In particular, PKAI functions as a redox-activated enzyme (Table 2). RI subunits contain a pair of redox-sensitive cysteine thiols within the N-terminal AKAP binding region of the protein; these redox-sensitive cysteine thiols are not present in RII. The redox-sensitive cysteine thiols in RI form interprotein disulfide dimers that stabilize a conformation that binds AKAP proteins with higher affinity 31. In cardiomyocytes, this is detected as a redox-dependent increase in PKAI binding to α-MHC, which has been characterized as a putative AKAP in the myofilament fraction 32. In theory, RI dimerization might also control binding to cTnT, another myofilament protein recently identified as a sarcomeric AKAP 33, but this has not been considered. Since the PKAI holoenzyme is activated by substrate-induced sensitization to cAMP (i.e. it displays activity at low cAMP concentrations that do not support activation of type II PKA), the redox-dependent redistribution of PKAI to the sarcomere could allow for the phosphorylation of cTnI (and other sarcomeric substrates such as cMyBP-C, see below) and an increased cardiac contractility under conditions that are not associated with a beta-adrenergic receptor-dependent increase in cAMP 32.

TABLE 2.

OXIDATIVE MODIFICATIONS OF PROTEIN KINASES

Kinase Post-Translational Modification Functional effects Ref
PKA RI subunit oxidation ↑ RI binding to AKAPs (α-MHC)
↑ PKAI kinase activity
32
Catalytic domain Cys199 -S-glutathionylation ↓kinase activity 3436
PKG1α Oxidation of Cys42 in the homodimerization domain ↑ affinity for substrates
↑ cGMP-independent catalytic activity
38
PKC Oxidation of C1 domain Cys residues ↓ autoinhibition, ↑ kinase activity 59
Calpain-dependent cleavage – release of a constitutively active catalytic domain fragment ↑ PKCα catalytic activity, ↑ PKCδ-dependent phosphorylation of 14-3-3 68, 8587
Oxidation of a conserved activation loop Cys ↓ kinase activity 36, 59
Src-dependent phosphorylation of PKCδ at Y311 Altered substrate specificity-acquisition of cTnI-T144 kinase activity 40
PKD c-Abl- and Src-dependent phosphorylations of PKD at Tyr463 and Tyr95 that relieve autoinhibition, promote PKCδ-dependent PKD phosphorylation at Ser744/Ser748. ↑ kinase activity 50, 51
CaMKII Met281/Met282 oxidation ↑ Ca2+-independent catalytic activity 76
ASK-1 Mechanisms that disrupt a C-terminal interaction with 14-3-3: Dephosphorylation of Ser967 at the ASK-1 C-terminus or phosphorylation of 14-3-3 by ROS-regulated kinases (PKD, MST-1, catalytic fragment of PKCδ).
Mechanisms that disrupt an N-terminal interaction with Trx-1 (Trx-1 oxidation)
↑ kinase activity 6668
MST-1 Caspase-dependent cleavage of an autoinhibitory domain ↑ kinase activity 60

The presence of distinct PKAI and PKAII activation mechanisms at the sarcomere allows for dynamic and nuanced control of myofilament function in response to various physiologic and pathologic stimuli. However, a redox-dependent mechanism that activates PKAI (via the RI subunit) may be counterbalanced by oxidative modifications involving a strategic located cysteine residue in the PKA catalytic subunit (at position 199 in the activation loop); S-glutathionylation at Cys199 (or the formation of internal disulfide between Cys199 and Cys343) leads to a decrease in kinase activity 34, 35. Structural models suggest that the redox-dependent decrease in catalytic activity is due to a steric effect and reduced affinity for substrate 34, but there is biochemical evidence that the cysteine thiol modification also decreases catalytic activity indirectly by facilitating the dephosphorylation of an adjacent threonine residue in the activation loop (a post-translational modification that is required for kinase activity 36).

Most studies have focused on cTnI-Ser23/Ser24 phosphorylation as a post-translational modification regulated by PKA, but this site also is a target for phosphorylation by other ROS-regulated Ser/Thr kinases. For example, autocrine/paracrine stimuli that activate the NO/cGMP pathway can promote cTnI-Ser23/Ser24 phosphorylation by protein kinase G (PKG) 37. PKG1α (a major PKG isoform in cardiomyocytes) contains a reactive cysteine at position 42 in the N-terminal homodimerization domain that abuts in the enzyme homodimer; oxidative stress leads to the formation of interprotein disulfide bonds that increase the enzyme’s affinity for substrate and leads to a high level of cGMP-independent PKG1α catalytic activity. The N-terminus of PKG1β (the other major PKG splice variant in cardiomyocytes) does not contain a reactive cysteine at this position and is not activated by oxidative stress 38. The redox-dependent mechanism for PKG1α activation appears to be important in the vasculature, where it provides for stimulus-specific mechanisms to control vasodilatation in response to NO and oxidative stress 39; the functional consequences of a redox-dependent PKG1α activation mechanism in cardiomyocytes warrant further study.

Other redox-regulated signaling enzymes that can function as cTnI-Ser23/Ser24 kinases include protein kinase D (PKD), p90 ribosomal S6 kinase (RSK), and certain isoforms of protein kinase C (PKC) 4044 (Table 2). PKD is a signal-regulated Ser/Thr kinase that phosphorylates sarcomeric proteins (cTnI, cMyBP-C) and regulates cardiac contractility; PKD also activates signal transduction pathways that regulate gene expression and contribute to cardiac hypertrophy 45. The canonical pathway for PKD activation involves the growth factor receptor-dependent hydrolysis of membrane phosphoinositides leading to the formation of diacylglycerol and the co-localization of PKD with allosterically-activated PKC isoforms at diacyglycerol-enriched membranes; this facilitates PKC-dependent trans-phosphorylation of PKD at Ser744/Ser748 (two highly conserved serine residues in the activation loop that regulate catalytic activity). The activated PKD enzyme then phosphorylates target substrates - typically at LxRxxpS consensus phosphorylation motifs 46. In this regard, it is interesting to note that PKD displays a high level of in vitro cTnI-Ser23/Ser24 catalytic activity, although rodent (PVRRRs23s24) and human (PIRRRS23S24) cTnI sequences diverge somewhat from an optimal PKD consensus phosphorylation motif. However, there is ample evidence that PKD can phosphorylate substrates (such as c-Jun, β-catenin, and type IIα PI4P kinase) that do not conform to a LxRxxpS/T motif and there are hints in the literature that the flexibility of target substrate recognition may be enhanced during oxidative stress (due to the somewhat different ROS-dependent mechanism for PKD activation 4749). PKD is activated during oxidative stress via a mechanism involving c-Abl, which phosphorylates PKD at Tyr463 in its autoinhibitory pleckstrin homology domain 50. This induces a conformational change that relieves autoinhibition and permits Src-dependent PKD phosphorylation at Tyr95 51. Since the phospho-tyrosine at position 95 is a consensus-binding motif for the C2 domain of PKCδ, this leads to a docking interaction between PKD and PKCδ and PKCδ-dependent PKD phosphorylation at Ser744/Ser748. Stimulus-specific differences in PKD activation mechanisms (in response to growth factor receptors and during oxidative stress) have been linked to distinct functional responses in the vasculature; the prediction that the activation mode might also dictate the in vivo actions of PKD in cardiomyocytes has not been considered. Rather, studies to date show that endothelin-1 receptors recruit a PKD-dependent mechanism that promotes cTnI-Ser23/Ser24 phosphorylation, decreases myofilament Ca2+ sensitivity, and enhances contraction in adult cardiomyocytes 4244, but the endothelin-1 receptor-dependent increase in PKD1 activity does not couple to changes in cTnI-Ser23/Ser24 phosphorylation in cultured neonatal rat cardiomyocytes 52. These divergent results suggest that stimulus-, age-, or disease-dependent differences in the cellular signaling machinery might influence PKD’s signaling repertoire (and PKD-dependent control of contraction) in cardiomyocytes.

cTnI contains additional phosphorylation sites at Ser43/Ser45 and Thr144 that are traditionally viewed as a target for PKC 5356. While the functional importance of Ser43/Ser45 phosphorylation remains uncertain, Thr144 is strategically positioned in the inhibitory region of cTnI where it can regulate calcium sensitivity and cross-bridge cycling rates. Thr144 phosphorylation has been attributed to PKCβ or the Tyr311-phosphorylated form of PKCδ (a form of PKCδ that accumulates during oxidative stress 57, 58). Of note, cTnI is phosphorylated by PKCδ in a stimulus-specific manner 40 PKCδ phosphorylates cTnI exclusively at Ser23/Ser24 when it is allosterically activated by lipid cofactors. However, oxidative stress activates Src which phosphorylates PKCδ at Tyr311; the Tyr311-phosphorylated form of PKCδ displays a high level of Thr144 kinase activity – it executes coordinate cTnI phosphorylations at Ser23/Ser24 and Thr144 40. This distinct cTnI phosphorylation pattern (i.e., involving a dual phosphorylation at Ser23/Ser24 and Thr144) is functionally important, since cTnI-Thr144 phosphorylation alone has little effect on force generation or calcium sensitivity; Thr144 phosphorylation becomes functionally important in a Ser23/Ser24-phosphorylated background, where it prevents calcium desensitization due to cTnI-Ser23/Ser24 phosphorylation 57. While these studies focus on a very specific ROS-dependent mechanism that ‘fine tunes’ the enzymology of PKCδ other redox modifications play a more general role to regulate PKC activity. For example, the lipid-binding C1 domain (that is conserved module in the regulatory domain of all phorbol ester-sensitive PKCs) contains redox-sensitive cysteine residues; oxidative modifications at these sites lead to conformational changes that relieve autoinhibition and induce a high level of cofactor-independent catalytic activity. Redox-modifications of the highly conserved cysteine residues in the catalytic domain activation loop have an opposite effect and disrupt PKC catalytic activity59.

Mst1 (mammalian sterile 20-like kinase 1) is a pro-apoptotic kinase that is activated via autophosphorylation and caspase-dependent cleavage of its autoinhibitory domain. Mst1 is activated in the context of ischemia/reperfusion injury and it contributes to adverse cardiac remodeling 60. Recent studies indicate that Mst1 interacts with and phosphorylates cTnI; phosphorylation has been mapped primarily to Thr32 (with some additional phosphorylation at Thr52, and Thr130 at Thr144). Mst1 also phosphorylates cTnT, but only when it is incorporated into the troponin complex; Mst1 does not phosphorylate free cTnT. There is evidence that the Mst1-dependent phosphorylation induces a conformational change in cTnI that alters its binding affinity for cTnT and cTnC 61.

cTnT is another thin filament proteins that contains phosphorylation clusters at Thr197/Ser201/Thr206 and Ser278/Thr287 - although studies to date suggest that T206 is the only phosphorylation site that directly influences contractility. cTnT-Thr206 phosphorylation has been attributed to PKC or Raf-1 (but not PKA or PKG) and is implicated as a mechanism that desensitized the myofilament response to calcium, decreases actomyosin Mg-ATPase, and depressed cardiac contractility 62, 63. cTnT also is phosphorylated by apoptosis signal-regulated kinase-1 (ASK-1), a ROS-regulated stress activated MAPK kinase kinase that is abundant in cardiomyocytes 64, 65. ASK-1 contains a central kinase domain flanked by regulatory domains that engage in intermolecular interactions that limit ASK-1 catalytic activity. The interaction between the Ser967-phosphorylated C-terminal regulatory domain of ASK-1 and 14-3-3 proteins – and the interaction between the ASK-1 N-terminal regulatory domain and reduced thioredoxin-1 – clamps ASK-1 in a configuration that maintains low basal catalytic activity. ASK-1 is activated during oxidative stress as a result of molecular events that disrupt these intermolecular interactions, including the ROS-dependent increase in the activity of cellular phosphatases that dephosphorylate the ASK-1 C-terminal Ser967 residue, 14-3-3 protein phosphorylation by ROS-regulated kinases (such as PKD, Mst family kinases, or the catalytic fragment of PKCδ 6668), or Trx-1 oxidation. ASK-1 activation has been linked to the activation of JNK or NF-κB pathways that influence apoptotic/necrotic cell death and adverse cardiac remodeling 64, 65. However, the activated form of ASK-1 also is detected in the sarcomere, where it phosphorylates cTnT 69. ASK-1 activation leads to decreased cardiomyocyte contractility, but the link between cTnT phosphorylation and the cardiodepressant actions of ASK-1 remain uncertain, both because ASK-1 phosphorylates cTnT at Thr197 and Ser201 –not Thr206 (the site that has been implicated in the control of thin filament function 63) and the activated form of ASK-1 also decreases the amplitude of Ca2+ transient (providing an alternate mechanism to explain the decrease in cardiomyocyte contractility 69).

Redox-regulation of Thick filament protein phosphorylation

Cardiac myosin binding protein-C (cMyBP-C) is a thick filament protein that is required for sarcomeric integrity, the regulation of cardiac contraction, and cardioprotection. cMyBP-C contains multiple phosphorylation sites in a linker region located between the Ig-like C1 and C2 domains in the N-terminal myosin-binding region of the protein (a region unique to the cardiac isoform of MyBP-C). An interaction between this region of cMyBP-C and the myosin subfragment 2 (S2) domain (a region close to the lever arm) influences thick filament packing and the kinetics of cross-bridge cycling; phosphorylation disrupts this interaction and accelerates cross-bridge kinetics. The three best-characterized cMyBP-C phosphorylation sites in this region are at RRTSer273, RR(I/T)Ser282, and LKKRDSer302; these three serine residues are flanked by sequences that support phosphorylation by basophilic kinases (and Ser302 resides in an optimal phosphorylation motif for PKD 70). Early studies established that all three sites are phosphorylated by PKA and that PKA also targets an additional in vitro phosphorylation site at Ser307 71,72. However, current literature suggests that phosphorylation is regulated in a hierarchical manner and that individual sites on cMyBP-C are differentially phosphorylated by PKC, PKD, RSK, and Ca2+- and calmodulin-dependent protein kinase II (CaMKII). In particular, Ser282 is a phosphoacceptor site for PKA, CaMKII, or RSK; phosphorylation at this site primes cMyBP-C for subsequent PKA-, PKC-, CaMKII- or PKD-dependent phosphorylation at Ser302 (and PKA- or PKC-dependent phosphorylation at Ser273 71, 73). Mutagenesis studies suggest cMyBP-C phosphorylation at Ser282 is sufficient to accelerate cross-bridge cycle kinetics (i.e., that under certain circumstances this post-translational modification can result in functional changes even without an increase in cMyBP-C phosphorylation at Ser302 or cTnI phosphorylation at Ser22/Ser23). However, there also is evidence that the physiologic control of cardiac contractile function requires reversible phosphorylations at all three sites 73, 74 and that electrical field stimulation leads to an increase in Ca2+-activated contractility at least in part through a mechanism involving PKD-dependent cMyBP-C phosphorylation at Ser302 75.

The ROS-dependent mechanisms that activate PKA, PKC, and PKD - that might underlie a redox-dependent increase in cMyBP-C phosphorylation - were considered in previous sections. CaMKII also has been characterized as a ROS-activated protein kinase (Table 2). CaMKII functions as dodecameric enzyme that is comprised of individual monomers containing three key structural elements: an association domain that controls assembly of the holoenzyme, a kinase domain that phosphorylates target substrates, and a regulatory domain containing an autoinhibitory motif that regulates catalytic activity. Stimuli that increase intracellular calcium and promote Ca2+/CaM binding to CaMKII induce a conformational change that relieves autoinhibition. With prolonged increases in intracellular calcium, CaMKII executes an intersubunit phosphorylation at Thr287 in the autoinhibitory domain that prevents re-association of the regulatory and catalytic domains and confers Ca2+-independent catalytic activity. Recent studies indicate that the methionine residues at positions 281/282 in CaMKII’s autoinhibitory domain (adjacent to the Thr287 phosphorylation site) are targets for oxidative modifications 76. Oxidation at these sites leads to a high level of Ca2+/CaM-independent CaMKII activity. Since oxidized and autophosphorylated forms of CaMKII share many cellular actions, a role for the redox-activated form of CaMKII as a cMyBP-C-Ser282 kinase is plausible and warrants future consideration.

Redox-regulation of Titin phosphorylation

Changes in titin isoform expression during development and in disease provide a mechanism to regulate cardiac stiffness on a relatively long time scale. The relatively high elastic recoil of the perinatal heart is attributable to a low N2BA/N2B ratio, whereas an increase in the N2BA/N2B ratio in chronic heart failure leads to a decrease in passive tension. The spring-like segments in titin’s elastic I-band also are targets for phosphorylation events that lead to more dynamic changes in cardiac elasticity. The serially-linked spring-like segments in titin’s I band are differentially phosphorylated by PKA/PKG and PKC. PKA and PKG both phosphorylate a single serine residue at position 469 in the N2B segment, leading to a decrease in passive tension 77, 78. Since this residue is conserved in human cardiac N2BA and N2B isoforms, this post-translational modification constitutes a general mechanism to regulate cardiac stiffness. PKCα phosphorylates cardiac and skeletal muscle titin isoforms primarily at different serine residues (Ser11878 and Ser12022) in the PEVK domain; phosphorylation in the PEVK domain has an antithetical effect to increase passive tension 79. Phosphorylation sites in other regions of the titin protein that do not regulate mechanical function also have been identified; some have speculated that these post-translational modifications may regulate docking interactions and influence titin’s role as a molecular scaffold.

MYOFILAMENT PROTEIN CLEAVAGE

Cardiac injury and oxidative stress also can lead to the degradation of sarcomeric proteins. Early studies showed that cTnI degradation is a prominent feature of ischemic damage, that degraded forms of cTnI remain associated with the myofilament lattice, and that cTnI cleavage may contribute to ischemia-induced changes in force generation and myofibrillar calcium sensitivity 80, 81. Some studies attribute myofilament protein degradation to μ-calpain, a calcium-dependent myofibril-associated protease that is activated in ischemic cardiomyocytes 82. There is evidence that cTnI is degraded to progressively smaller cleavage products with increasingly severe or prolonged intervals of ischemia/reperfusion injury. A brief/mild episode of ischemia/reperfusion injury leads to the conversion of cTnI (a 210 amino acid protein) to a smaller degradation product (residues 1-193) that forms covalent complexes with cleaved forms of cTnT and cTnC 83. More severe ischemia/reperfusion injury leads to further degradation of cTnI and the accumulation of shorter catalytic fragments (consisting of residues 63 193 and 73 193) that lack the N-terminal PKA phosphorylation sites and do not form these covalent complexes. Some studies suggest that cTnI may be protected from this form of proteolytic degradation by PKA-dependent phosphorylation of cTnI at Ser23/Ser24 82, 83. cTnT also appears to be vulnerable to calpain-dependent proteolytic cleavage with even very brief episodes of ischemia/reperfusion injury. Calpain cleaves cTnT at a site that removes the NH2-terminal modulatory domain, leaving a conformationally altered cTnT core structure (residues 72-291) that displays altered binding to cTnI, cTnC and Tm 84. Finally, MLC1 also is degraded during prolonged/severe episodes of ischemia/reperfusion injury; this also contributes to a decrease in force generation and calcium sensitivity 81.

While most studies have focused on calpain-mediated proteolytic events that are localized to the sarcomere, calpain could in theory influence contractile function by proteolytically activating protein kinases that phosphorylate myofibrillar proteins 85. For example, calpain cleaves PKCα at the V3 hinge region, freeing the C-terminal catalytic domain from the autoinhibitory constraints imposed by the N-terminal regulatory domain. There is recent evidence that the PKCα catalytic domain fragment displays a high level of constitutive activity; it acts as a ‘rogue’ kinase to phosphorylate cellular substrates – including those that are not (or are only weakly) phosphorylated by full-length PKCα. Receptor-independent proteolytic activation mechanisms are not specific for PKCα – since calpain cleaves other PKC isoforms 86, 87 and other Ser/Thr kinases such as PKD 88. A role for unregulated/mislocalized catalytic domain fragments generated during oxidative stress, as mediators of pathological cardiac remodeling and changes in contractile performance, has not been considered.

Calpain may not be the only (or even the primary) mediator of sarcomeric protein breakdown in the ischemic heart, since pro-apoptotic stimuli and oxidative stress also increase the activity of other proteolytic enzymes. For example, caspase-3 is activated by pro-apoptotic stimuli and it cleaves actin, α-actinin, and cTnT. Caspase-3 cleaves cTnT at a consensus site at DFD↓D97, but only when the protein is incorporated into the myofilament lattice; caspase-3 does not cleave free cTnT 89. Functional studies link caspase-3 treatment of skinned fiber bundles to defects in force/Ca2+ relationships and myofibrillar ATPase activity. These results suggest that caspase-induced myofilament protein breakdown may contribute to mechanical dysfunction and the evolution of heart failure 89. However, the importance of caspase-3 as a general mediator of myofibrillar protein breakdown in the setting oxidative stress remains uncertain, since caspase-3 contains a redox-sensitive catalytic cysteine (Cys163); oxidative modifications (S-glutathionylation or S-nitrosylation) at this site have been linked to a decrease in caspase-3 activity 9092.

Matrix metalloproteinase-2 (MMP-2, an abundant MMP in cardiomyocytes and many other cell types) also has recently emerged as a functionally important redox-activated endopeptidase that cleaves sarcomeric proteins. In fact, some have argued that at least some proteolytic events previously attributed to calpain may actually be mediated by MMP2, since [a] calpain and MMP-2 cleave many common substrates, [b] many inhibitors (MDL-28170, ALLN, ALLM, and PD-150606) that have been used to define the cardiac actions of calpain are also effective inhibitors of MMP-2 93, and [c] degraded forms of sarcomeric proteins such as cTnI are not detected in transgenic mice with cardiac-specific calpain overexpression 94. MMPs are zinc-dependent endoproteinases that are synthesized as latent, inactive zymogens that are maintained in an inactive state by an interaction between a cysteine thiol in the propeptide domain and the Zn2+-containing catalytic domain. MMP-2 is activated in the pericellular or extracellular compartment by upstream proteases (such as MMP-14) that cleave the inhibitory pro-peptide domain and expose the active site. This leads to the degradation of extracellular matrix and underlies MMP-2 widely recognized roles in tissue remodeling (including embryogenesis, angiogenesis, myocardial infarction, and various forms of wound healing). However, there is recent evidence that the highly conserved Cys in the propeptide domain is a target for oxidative modifications (specifically, peroxynitrite-dependent S-glutathiolation); an oxidative modification at this site disrupts the intramolecular autoinhibitory interaction and provides a non-proteolytic mechanism to activate MMP-2 95. The redox-activated form of MMP-2 is recovered in the sarcomere, where it anchors to proteolytic targets such as cTnI and MLC-1. MMP-2 cleaves cTnI, MLC-1, and MLC-2 during ischemia/reperfusion injury; some studies suggest that these sarcomeric protein cleavage events contribute to oxidative stress-dependent defects in cardiac contractility 9698. Moreover, there is increasing evidence that the controls of redox-induced events in the sarcomere can be rather elaborate and multi-factorial, since MLC-1 and MLC-2 are primed for MMP-2-dependent degradation by redox-induced post-translational modifications. For example, MMP-2-dependent cleavage of MLC-1 (at Y189↓E190 in its accessible C-terminus) is enhanced by a peroxynitrite-induced increase in MLC-1-Tyr78/Tyr190 nitration and Cys81 nitrosylation 97,99. Similarly, MMP-2-dependent cleavage of MLC-2 is facilitated by nitration at Tyr150 98. Finally, there is evidence that MMP-2 localizes to the Z-disk where it might play a role in Z-disk assembly and the maintenance of sarcomeric integrity by binding and cleaving α-actinin and titin 100, 101.

CONCLUSIONS, CAVEATS, AND FUTURE DIRECTIONS

This review summarizes recent advances in our knowledge of ROS-regulated post-translational modifications in sarcomeric proteins. The lengthy list and spectrum of the redox-regulated events summarized in Table 1 is a testament to recent advances in methodologies for proteomic profiling and the growing recognition that redox biology plays a fundamentally important role in the control of cardiac contraction. While there is considerable evidence that many protein redox modifications lead to functionally important changes in sarcomeric protein structure, stability, interactivity, and/or activity, our current understanding of the redox-dependent mechanisms that control contractility in vivo in the intact heart remains rather rudimentary in large part because biochemical studies have focused primarily on redox-dependent modifications on single purified contractile proteins or preparations that contain selected components of the contractile apparatus; the large size and limited solubility of many myofibrillar proteins makes some types of biochemical analysis quite challenging. Extrapolations from these more reductionist systems to the in vivo context may be misleading for several reasons. First, the conformation and/or exposed surfaces of a contractile protein may be altered by interactions with binding partners in the myofilament lattice in a manner that influences the accessibility of post-translational modification sites and either facilitates or prevents reactivity. Second, ROS-dependent modifications of sarcomeric proteins seldom occur in isolation – and structural modifications of one protein can have far-reaching effects on molecular interactions between sarcomeric proteins elsewhere in the complex. Hence, the ensemble effects of all post-translational modifications in the sarcomere determine the nature of the ROS-induced change in cardiac contractility in vivo in the intact heart. Finally, generalizations regarding ROS-dependent changes in cardiac contraction ignore the fact that oxidative stress represents a spectrum of responses that depend on the precise chemical nature of the oxidant species and level/severity of oxidative stress; this review provides numerous examples of oxidant species and ROS-activated enzymes that trigger different (in some cases diametrically opposite) effects on cardiac contractility. The complexities inherent in these redox-regulated mechanisms that control pump function present both challenges and opportunities for the development of more specific therapeutic strategies for heart disease.

TABLE 1.

ROS-INDUCED MODIFICATIONS OF CARDIAC SARCOMERIC PROTEINS

Protein Phosphorylation Ref Functional Effect Oxidation Functional effects Ref
cTnI Ser23/Ser24
PKA, PKC, PKG, PKD, p90RSK
30, 37
4044
↑ Ca2+ dissociation from TnC
↓ myofilament Ca2+ sensitivity
↑ rate of relaxation
Tyr nitration 25, 26
Ser43/Ser45
Thr144
PKCβ
MST-1
Y311-phosphorylated PKCδ
57, 58
61
40
↑ myofilament Ca2+ sensitivity
Altered cTnI conformation and cTnI binding to cTnT/cTnC
Thr32/Thr52/Thr130
MST-1
61
cTnT Ser206
PKC
Raf-1
63
62
↓ maximal force
↓ myofilament Ca2+ sensitivity
Tyr nitration 25, 26
Thr197/Ser201
PKC
ASK-1
63
69
Ser274/Thr287
PKC
63
Tm Cys190 Oxidation ↓ contractile function
Formation of Tm dimers
↓ binding to actin
↓ formation of actin-TM complexes
710, 12
HNO-dependent Cys190 Oxidation Formation of dimers Cys257 in actin
Tethers Tm to a position permissive for Ca2+- induced myofilament activation
29
Cys190 Carbonylation 8
Tyr nitration ↓ contractile function 25, 26
MHC Cys697/Cys707 Oxidation ↓ Maximal force 1719
HNO-dependent Cys oxidation Dimerization with Cys81 in MLC1
↑myofilament Ca2+ sensitivity
↑Contractility
29
Lys84 carbonylation 17
Nitration
Tyr114, Tyr116, Tyr134, Tyr142
↓ contractile function 15, 17, 22
MLC1 Tyr78/Tyr190 nitration
Cys81 nitrosylation
↑ degradation by MMP-2
↓ contractility
25, 99
MLC2 Tyr152 nitration ↑ degradation by MMP-2
↓ contractility
98
cMyBP-C Ser282
PKA, PKC, CaMK, PKD
Ser302
PKA, PKC, PKD
Ser273
PKA,PKC
30,31
75
Primes MyBP-C for subsequent phosphorylation at Ser302 and Ser273
Accelerates cross-bridge cycle kinetics
Tyr nitration 25, 26
Actin Cys374 oxidation ↓Tm-actin binding
↓ maximal force
↓ contractile function
↑ F-actin depolymerization
↓ Myosin ATPase activity
↓Actin filament sliding velocity
7, 8, 11
Cys374 glutathionylation ↓ α-actin polymerization kinetics
Destabilizes actin filaments
Decreases contractility
20, 23, 24
Cys257 oxidation by HNO Formation of dimers Cys190 in Tm
Tethers Tm to a position that is permissive for Ca2+-induced myofilament activation
↑ contractility
29
Carbonylation ↓ contractile function 8
Tyr nitration ↓ contractile function 25
α-Actinin Tyr nitration Deterioration of cross-striated pattern
↓ longitudinal force transmission
13, 25, 26
Titin Ser469
PKA
PKG
77, 78 ↓ passive tension Cys oxidation S-S bond formation that decreases the extensibility of titin N2B
↑ cardiac stiffness
27
Ser11878/Ser12022
PKCα
79 ↑ passive tension

Acknowledgments

SOURCES OF FUNDING: This work was supported by USPHS NHLBI grants HL 77860 and HL112388.

Non-standard Abbreviations

AKAP

A-kinase anchoring proteins

ASK-1

apoptosis signal-regulated kinase-1

CaMKII

Ca2+- and calmodulin-dependent protein kinase II

cMyBP

cardiac myosin binding protein

cTnC

cardiac troponin C

cTnI

cardiac troponin I

cTnT

cardiac troponin T

HNO

nitroxyl

MHC

myosin heavy chain

MLC-1

myosin light chain 1 (or essential light chain)

MLC-2

myosin light chain 2 (or regulatory light chain)

MMP

matrix metalloproteinase

Mst1

mammalian sterile 20-like kinase 1

NO

nitric oxide

ONOO-

peroxynitrite

PKA

protein kinase A

PKC

protein kinase C

PKD

protein kinase D

PKG

Protein kinase G

PTM

Post-translational modification

RNS

reactive nitrogen species

ROS

reactive oxygen species

Tm

Tropomyosin

Trx-1

thioredoxin-1

Footnotes

DISCLOSURES: none

References

  • 1.Xu L, Eu JP, Meissner G, Stamler JS. Activation of the cardiac calcium release channel (ryanodine receptor) by poly-S-nitrosylation. Science. 1998;279:234–237. doi: 10.1126/science.279.5348.234. [DOI] [PubMed] [Google Scholar]
  • 2.Adachi T, Weisbrod RM, Pimentel DR, Ying J, Sharov VS, Schoneich C, Cohen RA. S-glutathiolation by peroxynitrite activates serca during arterial relaxation by nitric oxide. Nat Med. 2004;10:1200–1207. doi: 10.1038/nm1119. [DOI] [PubMed] [Google Scholar]
  • 3.Lancel S, Zhang J, Evangelista A, Trucillo MP, Tong X, Siwik DA, Cohen RA, Colucci WS. Nitroxyl activates SERCA in cardiac myocytes via glutathiolation of cysteine 674. Circ Res. 2009;104:720–723. doi: 10.1161/CIRCRESAHA.108.188441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ferdinandy P, Danial H, Ambrus I, Rothery RA, Schulz R. Peroxynitrite is a major contributor to cytokine-induced myocardial contractile failure. Circ Res. 2000;87:241–247. doi: 10.1161/01.res.87.3.241. [DOI] [PubMed] [Google Scholar]
  • 5.Zweier JL, Fertmann J, Wei G. Nitric oxide and peroxynitrite in postischemic myocardium. Antioxidants & redox signaling. 2001;3:11–22. doi: 10.1089/152308601750100443. [DOI] [PubMed] [Google Scholar]
  • 6.MacFarlane NG, Miller DJ. Depression of peak force without altering calcium sensitivity by the superoxide anion in chemically skinned cardiac muscle of rat. Circ Res. 1992;70:1217–1224. doi: 10.1161/01.res.70.6.1217. [DOI] [PubMed] [Google Scholar]
  • 7.Canton M, Neverova I, Menabo R, Van Eyk J, Di Lisa F. Evidence of myofibrillar protein oxidation induced by postischemic reperfusion in isolated rat hearts. Am J Physiol Heart Circ Physiol. 2004;286:H870–877. doi: 10.1152/ajpheart.00714.2003. [DOI] [PubMed] [Google Scholar]
  • 8.Canton M, Menazza S, Sheeran FL, Polverino de Laureto P, Di Lisa F, Pepe S. Oxidation of myofibrillar proteins in human heart failure. J Am Coll Cardiol. 2011;57:300–309. doi: 10.1016/j.jacc.2010.06.058. [DOI] [PubMed] [Google Scholar]
  • 9.Canton M, Skyschally A, Menabo R, Boengler K, Gres P, Schulz R, Haude M, Erbel R, Di Lisa F, Heusch G. Oxidative modification of tropomyosin and myocardial dysfunction following coronary microembolization. Eur Heart J. 2006;27:875–881. doi: 10.1093/eurheartj/ehi751. [DOI] [PubMed] [Google Scholar]
  • 10.Avner BS, Shioura KM, Scruggs SB, Grachoff M, Geenen DL, Helseth DL, Jr, Farjah M, Goldspink PH, John Solaro R. Myocardial infarction in mice alters sarcomeric function via post-translational protein modification. Mol Cell Biochem. 2012;363:203–215. doi: 10.1007/s11010-011-1172-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Eaton P, Byers HL, Leeds N, Ward MA, Shattock MJ. Detection, quantitation, purification, and identification of cardiac proteins S-thiolated during ischemia and reperfusion. The Journal of biological chemistry. 2002;277:9806–9811. doi: 10.1074/jbc.M111454200. [DOI] [PubMed] [Google Scholar]
  • 12.Williams DL, Jr, Swenson CA. Disulfide bridges in tropomyosin. Effect on ATPase activity of actomyosin. European journal of biochemistry / FEBS. 1982;127:495–499. [PubMed] [Google Scholar]
  • 13.Borbely A, Toth A, Edes I, Virag L, Papp JG, Varro A, Paulus WJ, van der Velden J, Stienen GJ, Papp Z. Peroxynitrite-induced α-actinin nitration and contractile alterations in isolated human myocardial cells. Cardiovasc Res. 2005;67:225–233. doi: 10.1016/j.cardiores.2005.03.025. [DOI] [PubMed] [Google Scholar]
  • 14.Brunner F, Wolkart G. Peroxynitrite-induced cardiac depression: Role of myofilament desensitization and cgmp pathway. Cardiovasc Res. 2003;60:355–364. doi: 10.1016/j.cardiores.2003.08.001. [DOI] [PubMed] [Google Scholar]
  • 15.Mihm MJ, Yu F, Reiser PJ, Bauer JA. Effects of peroxynitrite on isolated cardiac trabeculae: Selective impact on myofibrillar energetic controllers. Biochimie. 2003;85:587–596. doi: 10.1016/s0300-9084(03)00090-7. [DOI] [PubMed] [Google Scholar]
  • 16.Mihm MJ, Bauer JA. Peroxynitrite-induced inhibition and nitration of cardiac myofibrillar creatine kinase. Biochimie. 2002;84:1013–1019. doi: 10.1016/s0300-9084(02)00005-6. [DOI] [PubMed] [Google Scholar]
  • 17.Tiago T, Palma PS, Gutierrez-Merino C, Aureliano M. Peroxynitrite-mediated oxidative modifications of myosin and implications on structure and function. Free radical research. 2010;44:1317–1327. doi: 10.3109/10715762.2010.502170. [DOI] [PubMed] [Google Scholar]
  • 18.Tiago T, Simao S, Aureliano M, Martin-Romero FJ, Gutierrez-Merino C. Inhibition of skeletal muscle S1-myosin atpase by peroxynitrite. Biochemistry. 2006;45:3794–3804. doi: 10.1021/bi0518500. [DOI] [PubMed] [Google Scholar]
  • 19.Passarelli C, Petrini S, Pastore A, Bonetto V, Sale P, Gaeta LM, Tozzi G, Bertini E, Canepari M, Rossi R, Piemonte F. Myosin as a potential redox-sensor: An in vitro study. Journal of muscle research and cell motility. 2008;29:119–126. doi: 10.1007/s10974-008-9145-x. [DOI] [PubMed] [Google Scholar]
  • 20.Passarelli C, Di Venere A, Piroddi N, Pastore A, Scellini B, Tesi C, Petrini S, Sale P, Bertini E, Poggesi C, Piemonte F. Susceptibility of isolated myofibrils to in vitro glutathionylation: Potential relevance to muscle functions. Cytoskeleton. 2010;67:81–89. doi: 10.1002/cm.20425. [DOI] [PubMed] [Google Scholar]
  • 21.Duke J, Takashi R, Ue K, Morales MF. Reciprocal reactivities of specific thiols when actin binds to myosin. Proc Natl Acad Sci U S A. 1976;73:302–306. doi: 10.1073/pnas.73.2.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hong SJ, Gokulrangan G, Schoneich C. Proteomic analysis of age dependent nitration of rat cardiac proteins by solution isoelectric focusing coupled to nanohplc tandem mass spectrometry. Experimental gerontology. 2007;42:639–651. doi: 10.1016/j.exger.2007.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chen FC, Ogut O. Decline of contractility during ischemia-reperfusion injury: Actin glutathionylation and its effect on allosteric interaction with tropomyosin. Am J Physiol Cell Physiol. 2006;290:C719–727. doi: 10.1152/ajpcell.00419.2005. [DOI] [PubMed] [Google Scholar]
  • 24.Pizarro GO, Ogut O. Impact of actin glutathionylation on the actomyosin-S1 ATPase. Biochemistry. 2009;48:7533–7538. doi: 10.1021/bi900669m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kanski J, Behring A, Pelling J, Schoneich C. Proteomic identification of 3-nitrotyrosine-containing rat cardiac proteins: Effects of biological aging. Am J Physiol Heart Circ Physiol. 2005;288:H371–381. doi: 10.1152/ajpheart.01030.2003. [DOI] [PubMed] [Google Scholar]
  • 26.Kanski J, Hong SJ, Schoneich C. Proteomic analysis of protein nitration in aging skeletal muscle and identification of nitrotyrosine-containing sequences in vivo by nanoelectrospray ionization tandem mass spectrometry. J Biol Chem. 2005;280:24261–24266. doi: 10.1074/jbc.M501773200. [DOI] [PubMed] [Google Scholar]
  • 27.Grutzner A, Garcia-Manyes S, Kotter S, Badilla CL, Fernandez JM, Linke WA. Modulation of titin-based stiffness by disulfide bonding in the cardiac titin N2-Bunique sequence. Biophys J. 2009;97:825–834. doi: 10.1016/j.bpj.2009.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dai T, Tian Y, Tocchetti CG, Katori T, Murphy AM, Kass DA, Paolocci N, Gao WD. Nitroxyl increases force development in rat cardiac muscle. J Physiol. 2007;580:951–960. doi: 10.1113/jphysiol.2007.129254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gao WD, Murray CI, Tian Y, Zhong X, Dumond JF, Shen X, Stanley BA, Foster DB, Wink DA, King SB, Van Eyk JE, Paolocci N. Nitroxyl(HNO)-mediated disulfide bond formation between cardiac myofilament cysteines enhances contractile function. Circ Res. 2012 doi: 10.1161/CIRCRESAHA.112.270827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Solaro RJ, Kobayashi T. Protein phosphorylation and signal transduction in cardiac thin filaments. J Biol Chem. 2011;286:9935–9940. doi: 10.1074/jbc.R110.197731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sarma GN, Kinderman FS, Kim C, von Daake S, Chen L, Wang BC, Taylor SS. Structure of d-AKAP2:PKA RI complex: Insights into AKAP specificity and selectivity. Structure. 2010;18:155–166. doi: 10.1016/j.str.2009.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Brennan JP, Bardswell SC, Burgoyne JR, Fuller W, Schroder E, Wait R, Begum S, Kentish JC, Eaton P. Oxidant-induced activation of type I protein kinase A is mediated by RI subunit interprotein disulfide bond formation. J Biol Chem. 2006;281:21827–21836. doi: 10.1074/jbc.M603952200. [DOI] [PubMed] [Google Scholar]
  • 33.Sumandea CA, Garcia-Cazarin ML, Bozio CH, Sievert GA, Balke CW, Sumandea MP. Cardiac troponin T, a sarcomeric AKAP, tethers protein kinase A at the myofilaments. J Biol Chem. 2011;286:530–541. doi: 10.1074/jbc.M110.148684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Humphries KM, Juliano C, Taylor SS. Regulation of cAMP-dependent protein kinase activity by glutathionylation. J Biol Chem. 2002;277:43505–43511. doi: 10.1074/jbc.M207088200. [DOI] [PubMed] [Google Scholar]
  • 35.Ward NE, Stewart JR, Ioannides CG, O’Brian CA. Oxidant-induced S-glutathiolation inactivates protein kinase C-α: A potential mechanism of PKC isozyme regulation. Biochemistry. 2000;39:10319–10329. doi: 10.1021/bi000781g. [DOI] [PubMed] [Google Scholar]
  • 36.Humphries KM, Deal MS, Taylor SS. Enhanced dephosphorylation of cAMP-dependent protein kinase by oxidation and thiol modification. J Biol Chem. 2005;280:2750–2758. doi: 10.1074/jbc.M410242200. [DOI] [PubMed] [Google Scholar]
  • 37.Lee DI, Vahebi S, Tocchetti CG, Barouch LA, Solaro RJ, Takimoto E, Kass DA. PDE5a suppression of acute beta-adrenergic activation requires modulation of myocyte beta-3 signaling coupled to PKG-mediated troponin I phosphorylation. Basic Res Cardiol. 2010;105:337–347. doi: 10.1007/s00395-010-0084-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Burgoyne JR, Madhani M, Cuello F, Charles RL, Brennan JP, Schroder E, Browning DD, Eaton P. Cysteine redox sensor in PKGIα enables oxidant-induced activation. Science. 2007;317:1393–1397. doi: 10.1126/science.1144318. [DOI] [PubMed] [Google Scholar]
  • 39.Zhang DX, Borbouse L, Gebremedhin D, Mendoza SA, Zinkevich NS, Li R, Gutterman DD. H2O2-induced dilation in human coronary arterioles: Role of protein kinase G dimerization and large-conductance Ca2+-activated K+ channel activation. Circ Res. 2012;110:471–480. doi: 10.1161/CIRCRESAHA.111.258871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sumandea MP, Rybin VO, Hinken AC, Wang C, Kobayashi T, Harleton E, Sievert G, Balke CW, Feinmark SJ, Solaro RJ, Steinberg SF. Tyrosine phosphorylation modifies PKCδ-dependent phosphorylation of cardiac troponin I. J Biol Chem. 2008;283:22680–22689. doi: 10.1074/jbc.M802396200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Itoh S, Ding B, Bains CP, Wang N, Takeishi Y, Jalili T, King GL, Walsh RA, Yan C, Abe J. Role of p90 ribosomal s6 kinase (p90RSK) in reactive oxygen species and protein kinase C-β-mediated cardiac troponin I phosphorylation. J Biol Chem. 2005;280:24135–24142. doi: 10.1074/jbc.M413015200. [DOI] [PubMed] [Google Scholar]
  • 42.Bardswell SC, Cuello F, Rowland AJ, Sadayappan S, Robbins J, Gautel M, Walker JW, Kentish JC, Avkiran M. Distinct sarcomeric substrates are responsible for protein kinase D-mediated regulation of cardiac myofilament Ca2+ sensitivity and cross-bridge cycling. J Biol Chem. 2010;285:5674–5682. doi: 10.1074/jbc.M109.066456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cuello F, Bardswell SC, Haworth RS, Yin X, Lutz S, Wieland T, Mayr M, Kentish JC, Avkiran M. Protein kinase D selectively targets cardiac troponin I and regulates myofilament Ca2+ sensitivity in ventricular myocytes. Circulation Research. 2007;100:864–873. doi: 10.1161/01.RES.0000260809.15393.fa. [DOI] [PubMed] [Google Scholar]
  • 44.Haworth RS, Cuello F, Herron TJ, Franzen G, Kentish JC, Gautel M, Avkiran M. Protein kinase D is a novel mediator of cardiac troponin I phosphorylation and regulates myofilament function. Circulation Research. 2004;95:1091–1099. doi: 10.1161/01.RES.0000149299.34793.3c. [DOI] [PubMed] [Google Scholar]
  • 45.Avkiran M, Rowland AJ, Cuello F, Haworth RS. Protein kinase D in the cardiovascular system: Emerging roles in health and disease. Circulation Research. 2008;102:157–163. doi: 10.1161/CIRCRESAHA.107.168211. [DOI] [PubMed] [Google Scholar]
  • 46.Nishikawa K, Toker A, Johannes FJ, Songyang Z, Cantley LC. Determination of the specific substrate sequence motifs of protein kinase C isozymes. J Biol Chem. 1997;272:952–960. doi: 10.1074/jbc.272.2.952. [DOI] [PubMed] [Google Scholar]
  • 47.Waldron RT, Whitelegge JP, Faull KF, Rozengurt E. Identification of a novel phosphorylation site in c-jun directly targeted in vitro by protein kinase D. Biochem Biophys Res Commun. 2007;356:361–367. doi: 10.1016/j.bbrc.2007.02.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hinchliffe KA, Irvine RF. Regulation of type II PIP kinase by PKD phosphorylation. Cell Signal. 2006;18:1906–1913. doi: 10.1016/j.cellsig.2006.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Du C, Jaggi M, Zhang C, Balaji KC. Protein kinase D1-mediated phosphorylation and subcellular localization of β-catenin. Cancer Res. 2009;69:1117–1124. doi: 10.1158/0008-5472.CAN-07-6270. [DOI] [PubMed] [Google Scholar]
  • 50.Storz P, Doppler H, Johannes FJ, Toker A. Tyrosine phosphorylation of protein kinase D in the pleckstrin homology domain leads to activation. J Biol Chem. 2003;278:17969–17976. doi: 10.1074/jbc.M213224200. [DOI] [PubMed] [Google Scholar]
  • 51.Doppler H, Storz P. A novel tyrosine phosphorylation site in protein kinase D contributes to oxidative stress-mediated activation. J Biol Chem. 2007;282:31873–31881. doi: 10.1074/jbc.M703584200. [DOI] [PubMed] [Google Scholar]
  • 52.Guo J, Gertsberg Z, Ozgen N, Sabri A, Steinberg SF. Protein kinase D isoforms are activated in an agonist-specific manner in cardiomyocytes. J Biol Chem. 2010 doi: 10.1074/jbc.M110.208058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Takeishi Y, Chu G, Kirkpatrick DM, Li Z, Wakasaki H, Kranias EG, King GL, Walsh RA. In vivo phosphorylation of cardiac troponin I by protein kinase Cβ decreases cardiomyocyte calcium responsiveness and contractility in transgenic mouse hearts. J Clin Invest. 1998;102:72–78. doi: 10.1172/JCI2709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jideama NM, Noland TA, Raynor RL, Blobe GC, Fabbro D, Kazanietz MG, Blumberg PM, Hannun YA, Kuo JF. Phosphorylation specificities of protein kinase C isozymes for bovine cardiac troponin I and troponin T and sites within these proteins and regulation of myofilament properties. J Biol Chem. 1996;271:23277–23283. doi: 10.1074/jbc.271.38.23277. [DOI] [PubMed] [Google Scholar]
  • 55.Noland TA, Jr, Raynor RL, Jideama NM, Guo X, Kazanietz MG, Blumberg PM, Solaro RJ, Kuo JF. Differential regulation of cardiac actomyosin S-1 Mg ATPase by protein kinase C isozyme-specific phosphorylation of specific sites in cardiac troponin I and its phosphorylation site mutants. Biochemistry. 1996;35:14923–14931. doi: 10.1021/bi9616357. [DOI] [PubMed] [Google Scholar]
  • 56.Noland TA, Raynor RL, Kuo JF. Identification of sites phosphorylated in bovine cardiac troponin I and troponin T by protein kinase C and comparative substrate activity of synthetic peptides containing phosphorylation sites. J Biol Chem. 1989;264:20778–20785. [PubMed] [Google Scholar]
  • 57.Lu QW, Hinken AC, Patrick SE, Solaro RJ, Kobayashi T. Phosphorylation of cardiac troponin I at protein kinase C site threonine 144 depresses cooperative activation of thin filaments. J Biol Chem. 2010;285:11810–11817. doi: 10.1074/jbc.M109.055657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang H, Grant JE, Doede CM, Sadayappan S, Robbins J, Walker JW. PKC-βII sensitizes cardiac myofilaments to Ca2+ by phosphorylating troponin Ion threonine144. J Mol Cell Cardiol. 2006;41:823–833. doi: 10.1016/j.yjmcc.2006.08.016. [DOI] [PubMed] [Google Scholar]
  • 59.Gopalakrishna R, Jaken S. Protein kinase C signaling and oxidative stress. Free Radic Biol Med. 2000;28:1349–1361. doi: 10.1016/s0891-5849(00)00221-5. [DOI] [PubMed] [Google Scholar]
  • 60.Yamamoto S, Yang G, Zablocki D, Liu J, Hong C, Kim SJ, Soler S, Odashima M, Thaisz J, Yehia G, Molina CA, Yatani A, Vatner DE, Vatner SF, Sadoshima J. Activation of MST1 causes dilated cardiomyopathy by stimulating apoptosis without compensatory ventricular myocyte hypertrophy. J Clin Invest. 2003;111:1463–1474. doi: 10.1172/JCI17459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.You B, Yan G, Zhang Z, Yan L, Li J, Ge Q, Jin JP, Sun J. Phosphorylation of cardiac troponin I by mammalian sterile 20-like kinase 1. Biochem J. 2009;418:93–101. doi: 10.1042/BJ20081340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pfleiderer P, Sumandea MP, Rybin VO, Wang C, Steinberg SF. Raf-1: A novel cardiac troponin T kinase. J Muscle Res Cell Motil. 2009;30:67–72. doi: 10.1007/s10974-009-9176-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sumandea MP, Pyle WG, Kobayashi T, de Tombe PP, Solaro RJ. Identification of a functionally critical protein kinase C phosphorylation residue of cardiac troponin T. J Biol Chem. 2003;278:35135–35144. doi: 10.1074/jbc.M306325200. [DOI] [PubMed] [Google Scholar]
  • 64.Hirotani S, Otsu K, Nishida K, Higuchi Y, Morita T, Nakayama H, Yamaguchi O, Mano T, Matsumura Y, Ueno H, Tada M, Hori M. Involvement of nuclear factor-κB and apoptosis signal-regulating kinase 1 in G-protein-coupled receptor agonist-induced cardiomyocyte hypertrophy. Circulation. 2002;105:509–515. doi: 10.1161/hc0402.102863. [DOI] [PubMed] [Google Scholar]
  • 65.Yamaguchi O, Higuchi Y, Hirotani S, Kashiwase K, Nakayama H, Hikoso S, Takeda T, Watanabe T, Asahi M, Taniike M, Matsumura Y, Tsujimoto I, Hongo K, Kusakari Y, Kurihara S, Nishida K, Ichijo H, Hori M, Otsu K. Targeted deletion of apoptosis signal-regulating kinase 1 attenuates left ventricular remodeling. Proc Natl Acad Sci US A. 2003;100:15883–15888. doi: 10.1073/pnas.2136717100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhang W, Zheng S, Storz P, Min W. Protein kinase D specifically mediates apoptosis signal-regulating kinase 1-JNK signaling induced by H2O ζ but not tumor necrosis factor. J Biol Chem. 2005;280:19036–19044. doi: 10.1074/jbc.M414674200. [DOI] [PubMed] [Google Scholar]
  • 67.Zhou J, Shao Z, Kerkela R, Ichijo H, Muslin AJ, Pombo C, Force T. Serine 58 of 14-3-3 ζ is a molecular switch regulating ASK1 and oxidant stress-induced cell death. Mol Cell Biol. 2009;29:4167–4176. doi: 10.1128/MCB.01067-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hamaguchi A, Suzuki E, Murayama K, Fujimura T, Hikita T, Iwabuchi K, Handa K, Withers DA, Masters SC, Fu H, Hakomori S. Sphingosine-dependent protein kinase-1 (SDK1), directed to 14-3-3, is identified as the kinase domain of PKCδ. J Biol Chem. 2003;278:41557–41565. doi: 10.1074/jbc.M305294200. [DOI] [PubMed] [Google Scholar]
  • 69.He X, Liu Y, Sharma V, Dirksen RT, Waugh R, Sheu SS, Min W. ASK1 associates with troponin T and induces troponin T phosphorylation and contractile dysfunction in cardiomyocytes. Am J Pathol. 2003;163:243–251. doi: 10.1016/S0002-9440(10)63647-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Copeland O, Sadayappan S, Messer AE, Steinen GJ, van der Velden J, Marston SB. Analysis of cardiac myosin binding protein-Cphosphorylation in human heart muscle. Journal of molecular and cellular cardiology. 2010;49:1003–1011. doi: 10.1016/j.yjmcc.2010.09.007. [DOI] [PubMed] [Google Scholar]
  • 71.Gautel M, Zuffardi O, Freiburg A, Labeit S. Phosphorylation switches specific for the cardiac isoform of myosin binding protein-C: A modulator of cardiac contraction? EMBO J. 1995;14:1952–1960. doi: 10.1002/j.1460-2075.1995.tb07187.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jia W, Shaffer JF, Harris SP, Leary JA. Identification of novel protein kinase A phosphorylation sites in the m-domain of human and murine cardiac myosin binding protein-C using mass spectrometry analysis. Journal of proteome research. 2010;9:1843–1853. doi: 10.1021/pr901006h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sadayappan S, Gulick J, Osinska H, Barefield D, Cuello F, Avkiran M, Lasko VM, Lorenz JN, Maillet M, Martin JL, Brown JH, Bers DM, Molkentin JD, James J, Robbins J. A critical function for Ser-282 in cardiac myosin binding protein-Cphosphorylation and cardiac function. Circ Res. 2011;109:141–150. doi: 10.1161/CIRCRESAHA.111.242560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cuello F, Bardswell SC, Haworth RS, Ehler E, Sadayappan S, Kentish JC, Avkiran M. Novel role for p90 ribosomal S6 kinase in the regulation of cardiac myofilament phosphorylation. J Biol Chem. 2011;286:5300–5310. doi: 10.1074/jbc.M110.202713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Dirkx E, Cazorla O, Schwenk RW, Lorenzen-Schmidt I, Sadayappan S, Van Lint J, Carrier L, van Eys GJ, Glatz JF, Luiken JJ. Protein kinase D increases maximal Ca2+-activated tension of cardiomyocyte contraction by phosphorylation of cMyBP-C-Ser315. Am J Physiol Heart Circ Physiol. 2012;303:H323–331. doi: 10.1152/ajpheart.00749.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Erickson JR, Joiner ML, Guan X, Kutschke W, Yang J, Oddis CV, Bartlett RK, Lowe JS, O’Donnell SE, Aykin-Burns N, Zimmerman MC, Zimmerman K, Ham AJ, Weiss RM, Spitz DR, Shea MA, Colbran RJ, Mohler PJ, Anderson ME. A dynamic pathway for calcium-independent activation of CaMKII by methionine oxidation. Cell. 2008;133:462–474. doi: 10.1016/j.cell.2008.02.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yamasaki R, Wu Y, McNabb M, Greaser M, Labeit S, Granzier H. Protein kinase A phosphorylates titin’s cardiac-specific N2B domain and reduces passive tension in rat cardiac myocytes. Circulation Research. 2002;90:1181–1188. doi: 10.1161/01.res.0000021115.24712.99. [DOI] [PubMed] [Google Scholar]
  • 78.Kruger M, Kotter S, Grutzner A, Lang P, Andresen C, Redfield MM, Butt E, dos Remedios CG, Linke WA. Protein kinase G modulates human myocardial passive stiffness by phosphorylation of the titin springs. Circulation Research. 2009;104:87–94. doi: 10.1161/CIRCRESAHA.108.184408. [DOI] [PubMed] [Google Scholar]
  • 79.Hidalgo C, Hudson B, Bogomolovas J, Zhu Y, Anderson B, Greaser M, Labeit S, Granzier H. PKC phosphorylation of titin’s PEVK element: A novel and conserved pathway for modulating myocardial stiffness. Circulation Research. 2009;105:631–638. 617. doi: 10.1161/CIRCRESAHA.109.198465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Westfall MV, Solaro RJ. Alterations in myofibrillar function and protein profiles after complete global ischemia in rat hearts. Circ Res. 1992;70:302–313. doi: 10.1161/01.res.70.2.302. [DOI] [PubMed] [Google Scholar]
  • 81.Van Eyk JE, Powers F, Law W, Larue C, Hodges RS, Solaro RJ. Breakdown and release of myofilament proteins during ischemia and ischemia/reperfusion in rat hearts: Identification of degradation products and effects on the pCa-force relation. Circ Res. 1998;82:261–271. doi: 10.1161/01.res.82.2.261. [DOI] [PubMed] [Google Scholar]
  • 82.Di Lisa F, De Tullio R, Salamino F, Barbato R, Melloni E, Siliprandi N, Schiaffino S, Pontremoli S. Specific degradation of troponin T and I by μ-calpain and its modulation by substrate phosphorylation. Biochem J. 1995;308:57–61. doi: 10.1042/bj3080057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.McDonough JL, Arrell DK, Van Eyk JE. Troponin I degradation and covalent complex formation accompanies myocardial ischemia/reperfusion injury. Circ Res. 1999;84:9–20. doi: 10.1161/01.res.84.1.9. [DOI] [PubMed] [Google Scholar]
  • 84.Zhang Z, Biesiadecki BJ, Jin JP. Selective deletion of the NH2-terminal variable region of cardiac troponin T in ischemia reperfusion by myofibril-associated μ-calpain cleavage. Biochemistry. 2006;45:11681–11694. doi: 10.1021/bi060273s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kang MY, Zhang Y, Matkovich SJ, Diwan A, Chishti AH, Dorn GW. Receptor-independent cardiac protein kinase C-α activation by calpain-mediated truncation of regulatory domains. Circulation Research. 2010;107:903–912. doi: 10.1161/CIRCRESAHA.110.220772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kishimoto A, Mikawa K, Hashimoto K, Yasuda I, Tanaka S, Tominaga M, Kuroda T, Nishizuka Y. Limited proteolysis of protein kinase C subspecies by calcium-dependent neutral protease (calpain) J Biol Chem. 1989;264:4088–4092. [PubMed] [Google Scholar]
  • 87.Yamakawa H, Banno Y, Nakashima S, Yoshimura S, Sawada M, Nishimura Y, Nozawa Y, Sakai N. Crucial role of calpain in hypoxic PC12 cell death: Calpain, but not caspases, mediates degradation of cytoskeletal proteins and protein kinase C-α and -δ. Neurological research. 2001;23:522–530. doi: 10.1179/016164101101198776. [DOI] [PubMed] [Google Scholar]
  • 88.Kennett SB, Roberts JD, Olden K. Requirement of protein kinase C-μ activation and calpain-mediated proteolysis for arachidonic acid-stimulated adhesion of MDA-MB-435 human mammary carcinoma cells to collagen type IV. J Biol Chem. 2004;279:3300–3307. doi: 10.1074/jbc.M305734200. [DOI] [PubMed] [Google Scholar]
  • 89.Communal C, Sumandea M, De Tombe P, Narula J, Solaro RJ, Hajjar RJ. Functional consequences of caspase activation in cardiac myocytes. Proc Natl Acad Sci US A. 2002;99:6252–6256. doi: 10.1073/pnas.092022999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Huang Z, Pinto JT, Deng H, Richie JP., Jr Inhibition of caspase-3 activity and activation by protein glutathionylation. Biochem Pharmacol. 2008;75:2234–2244. doi: 10.1016/j.bcp.2008.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mitchell DA, Marletta MA. Thioredoxin catalyzes the s-nitrosation of the caspase-3 active site cysteine. Nature chemical biology. 2005;1:154–158. doi: 10.1038/nchembio720. [DOI] [PubMed] [Google Scholar]
  • 92.Pan S, Berk BC. Glutathiolation regulates tumor necrosis factor-alpha-induced caspase-3 cleavage and apoptosis: Key role for glutaredoxin in the death pathway. Circ Res. 2007;100:213–219. doi: 10.1161/01.RES.0000256089.30318.20. [DOI] [PubMed] [Google Scholar]
  • 93.Ali MA, Stepanko A, Fan X, Holt A, Schulz R. Calpain inhibitors exhibit matrix metalloproteinase-2 inhibitory activity. Biochem Biophys Res Commun. 2012;423:1–5. doi: 10.1016/j.bbrc.2012.05.005. [DOI] [PubMed] [Google Scholar]
  • 94.Galvez AS, Diwan A, Odley AM, Hahn HS, Osinska H, Melendez JG, Robbins J, Lynch RA, Marreez Y, Dorn GW., 2nd Cardiomyocyte degeneration with calpain deficiency reveals a critical role in protein homeostasis. Circ Res. 2007;100:1071–1078. doi: 10.1161/01.RES.0000261938.28365.11. [DOI] [PubMed] [Google Scholar]
  • 95.Viappiani S, Nicolescu AC, Holt A, Sawicki G, Crawford BD, Leon H, van MT, Schulz R. Activation and modulation of 72 kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochemical Pharmacology. 2009;77:826–834. doi: 10.1016/j.bcp.2008.11.004. [DOI] [PubMed] [Google Scholar]
  • 96.Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation. 2002;106:1543–1549. doi: 10.1161/01.cir.0000028818.33488.7b. [DOI] [PubMed] [Google Scholar]
  • 97.Sawicki G, Leon H, Sawicka J, Sariahmetoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: A new intracellular target for matrix metalloproteinase-2. Circulation. 2005;112:544–552. doi: 10.1161/CIRCULATIONAHA.104.531616. [DOI] [PubMed] [Google Scholar]
  • 98.Doroszko A, Polewicz D, Cadete VJ, Sawicka J, Jones M, Szczesna-Cordary D, Cheung PY, Sawicki G. Neonatal asphyxia induces the nitration of cardiac myosin light chain 2 that is associated with cardiac systolic dysfunction. Shock. 2010;34:592–600. doi: 10.1097/SHK.0b013e3181e14f1d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Polewicz D, Cadete VJ, Doroszko A, Hunter BE, Sawicka J, Szczesna-Cordary D, Light PE, Sawicki G. Ischemia induced peroxynitrite dependent modifications of cardiomyocyte MLC1 increases its degradation by MMP-2 leading to contractile dysfunction. Journal of cellular and molecular medicine. 2011;15:1136–1147. doi: 10.1111/j.1582-4934.2010.01094.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sung MM, Schulz CG, Wang W, Sawicki G, Bautista-Lopez NL, Schulz R. Matrix metalloproteinase-2 degrades the cytoskeletal protein α-actinin in peroxynitrite mediated myocardial injury. Journal of molecular and cellular cardiology. 2007;43:429–436. doi: 10.1016/j.yjmcc.2007.07.055. [DOI] [PubMed] [Google Scholar]
  • 101.Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R. Titin is a target of matrix metalloproteinase-2: Implications in myocardial ischemia/reperfusion injury. Circulation. 2010;122:2039–2047. doi: 10.1161/CIRCULATIONAHA.109.930222. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES