Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 17(12): 4243-4257, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30141336

ABSTRACT

Cysteine oxidative modification of cellular proteins is crucial for many aspects of cardiac hypertrophy development. However, integrated dissection of multiple types of cysteine oxidative post-translational modifications (O-PTM) of proteomes in cardiac hypertrophy is currently missing. Here we developed a novel discovery platform that encompasses a customized biotin switch-based quantitative proteomics pipeline and an advanced analytic workflow to comprehensively profile the landscape of cysteine O-PTM in an ISO-induced cardiac hypertrophy mouse model. Specifically, we identified a total of 1655 proteins containing 3324 oxidized cysteine sites by at least one of the following three modifications: reversible cysteine O-PTM, cysteine sulfinylation (CysSO2H), and cysteine sulfonylation (CysSO3H). Analyzing the hypertrophy signatures that are reproducibly discovered from this computational workflow unveiled four biological processes with increased cysteine O-PTM. Among them, protein phosphorylation, creatine metabolism, and response to elevated Ca2+ pathways exhibited an elevation of cysteine O-PTM in early stages, whereas glucose metabolism enzymes were increasingly modified in later stages, illustrating a temporal regulatory map in cardiac hypertrophy. Our cysteine O-PTM platform depicts a dynamic and integrated landscape of the cysteine oxidative proteome, through the extracted molecular signatures, and provides critical mechanistic insights in cardiac hypertrophy. Data are available via ProteomeXchange with identifier PXD010336.


Subject(s)
Cardiomegaly/metabolism , Cysteine/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Calcium/metabolism , Creatine/metabolism , Cysteine/chemistry , Glucose/metabolism , Humans , Oxidation-Reduction , Phosphorylation , Time Factors
2.
Gene ; 590(1): 90-6, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27312950

ABSTRACT

Protein kinase C-epsilon (PKCε) is an isoform of a large PKC family of enzymes that has a variety of functions in different cell types. Here we discuss two major roles of PKCε in cardiac muscle cells; specifically, its role in regulating cardiac muscle contraction via targeting the sarcomeric proteins, as well as modulating cardiac cell energy production and metabolism by targeting cardiac mitochondria. The importance of PKCε action is described within the context of intracellular localization, as substrate selectivity and specificity is achieved through spatiotemporal targeting of PKCε. Accordingly, the role of PKCε in regulating myocardial function in physiological and pathological states has been documented in both cardioprotection and cardiac hypertrophy.


Subject(s)
Cardiomegaly/enzymology , Mitochondria, Heart/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Protein Kinase C-epsilon/genetics , Sarcomeres/enzymology , Arachidonic Acid/pharmacology , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiotonic Agents/pharmacology , Enzyme Activation , Gene Expression , Humans , Isoproterenol/pharmacology , Mitochondria, Heart/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardial Contraction/drug effects , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenylephrine/pharmacology , Protein Kinase C-epsilon/metabolism , Sarcomeres/drug effects , Substrate Specificity
4.
J Mol Cell Cardiol ; 71: 32-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24140722

ABSTRACT

The ubiquitin-proteasome system (UPS) is the major intracellular degradation system, and its proper function is critical to the health and function of cardiac cells. Alterations in cardiac proteasomes have been linked to several pathological phenotypes, including cardiomyopathies, ischemia-reperfusion injury, heart failure, and hypertrophy. Defects in proteasome-dependent cellular protein homeostasis can be causal for the initiation and progression of certain cardiovascular diseases. Emerging evidence suggests that the UPS can specifically target proteins that govern pathological signaling pathways for degradation, thus altering downstream effectors and disease outcomes. Alterations in UPS-substrate interactions in disease occur, in part, due to direct modifications of 19S, 11S or 20S proteasome subunits. Post-translational modifications (PTMs) are one facet of this proteasomal regulation, with over 400 known phosphorylation sites, over 500 ubiquitination sites and 83 internal lysine acetylation sites, as well as multiple sites for caspase cleavage, glycosylation (such as O-GlcNAc modification), methylation, nitrosylation, oxidation, and SUMOylation. Changes in cardiac proteasome PTMs, which occur in ischemia and cardiomyopathies, are associated with changes in proteasome activity and proteasome assembly; however several features of this regulation remain to be explored. In this review, we focus on how some of the less common PTMs affect proteasome function and alter cellular protein homeostasis. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".


Subject(s)
Myocardium/metabolism , Proteasome Endopeptidase Complex/metabolism , Sumoylation , Ubiquitination , Animals , Homeostasis , Humans , Myocardium/enzymology , Ubiquitin/metabolism
5.
Mol Cell Proteomics ; 12(12): 3793-802, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037710

ABSTRACT

Proteasome complexes play essential roles in maintaining cellular protein homeostasis and serve fundamental roles in cardiac function under normal and pathological conditions. A functional detriment in proteasomal activities has been recognized as a major contributor to the progression of cardiovascular diseases. Therefore, approaches to restore proteolytic function within the setting of the diseased myocardium would be of great clinical significance. In this study, we discovered that the cardiac proteasomal activity could be regulated by acetylation. Histone deacetylase (HDAC) inhibitors (suberoylanilide hydroxamic acid and sodium valproate) enhanced the acetylation of 20S proteasome subunits in the myocardium and led to an elevation of proteolytic capacity. This regulatory paradigm was present in both healthy and acutely ischemia/reperfusion (I/R) injured murine hearts, and HDAC inhibition in vitro restored proteolytic capacities to baseline sham levels in injured hearts. This mechanism of regulation was also viable in failing human myocardium. With 20S proteasomal complexes purified from murine myocardium treated with HDAC inhibitors in vivo, we confirmed that acetylation of 20S subunits directly, at least in part, presents a molecular explanation for the improvement in function. Furthermore, using high-resolution LC-MS/MS, we unraveled the first cardiac 20S acetylome, which identified the acetylation of nine N-termini and seven internal lysine residues. Acetylation on four lysine residues and four N-termini on cardiac proteasomes were novel discoveries of this study. In addition, the acetylation of five lysine residues was inducible via HDAC inhibition, which correlated with the enhancement of 20S proteasomal activity. Taken as a whole, our investigation unveiled a novel mechanism of proteasomal function regulation in vivo and established a new strategy for the potential rescue of compromised proteolytic function in the failing heart using HDAC inhibitors.


Subject(s)
Heart Ventricles/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Valproic Acid/pharmacology , Acetylation/drug effects , Animals , Chromatography, Liquid , Gene Expression Regulation , Heart Failure/pathology , Heart Failure/surgery , Heart Transplantation , Heart Ventricles/enzymology , Heart Ventricles/pathology , Histone Deacetylases/metabolism , Humans , Lysine/metabolism , Male , Mice , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/isolation & purification , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Signal Transduction , Tandem Mass Spectrometry , Vorinostat
6.
Biochim Biophys Acta ; 1833(4): 823-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22940544

ABSTRACT

Previous structural studies indicated a special functional role for an acidic region composed of residues 1-10 in the unique N-terminal peptide of cardiac troponin I (cTnI). Employing LC-MS/MS, we determined the presence of phosphorylation sites at S5/S6 in cTnI from wild type mouse hearts as well as in hearts of mice chronically expressing active protein kinase C-ε (PKCε) and exhibiting severe dilated cardiomyopathy (DCM). To determine the functional significance of these phosphorylations, we cloned and expressed wild-type cTnI, (Wt), and cTnI variants expressing pseudo-phosphorylation cTnI-(S5D), cTnI(S6D), as well as cTnI(S5A) and cTnI(S6A). We exchanged native Tn of detergent-extracted (skinned) fiber bundles with Tn reconstituted with the variant cTnIs and measured tension and cross-bridge dynamics. Compared to controls, myofilaments controlled by cTnI with pseudo-phosphorylation (S6D) or Ala substitution (S6A) demonstrated a significant depression in maximum tension, ATPase rate, and ktr, but no change in half-maximally activating Ca(2+). In contrast, pseudo-phosphorylation at position 5 (S5D) had no effects, although S5A induced an increase in Ca(2+)-sensitivity with no change in maximum tension or ktr. We further tested the impact of acidic domain modifications on myofilament function in studies examining the effects of cTnI(A2V), a mutation linked to DCM. This mutation significantly altered the inhibitory activity of cTnI as well as cooperativity of activation of myofilament tension, but not when S23/S24 were pseudo-phosphorylated. Our data indicate a new functional and pathological role of amino acid modifications in the N-terminal acidic domain of cTnI that is modified by phosphorylations at cTnI(S23/S24). This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Myocardium/metabolism , Myofibrils/metabolism , Troponin I/metabolism , Adenosine Triphosphatases/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Dilated/pathology , Gene Expression , Humans , Isometric Contraction , Kinetics , Male , Mice , Mice, Transgenic , Muscle Tonus , Mutation , Myocardium/pathology , Myofibrils/pathology , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Troponin I/chemistry , Troponin I/genetics
7.
J Proteomics ; 81: 15-23, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23022582

ABSTRACT

We report the development of a multiple-reaction monitoring (MRM) strategy specifically tailored to the detection and quantification of mitochondrial protein phosphorylation. We recently derived 68 MRM transitions specific to protein modifications in the respiratory chain, voltage-dependent anion channel, and adenine nucleotide translocase. Here, we have now expanded the total number of MRM transitions to 176 to cover proteins from the tricarboxylic acid cycle, pyruvate dehydrogenase complex, and branched-chain alpha-keto acid dehydrogenase complex. We utilized the transition set to analyze endogenous protein phosphorylation in human heart, mouse heart, and mouse liver. The data demonstrate the potential utility of the MRM workflow for studying the functional details of mitochondrial phosphorylation signaling. This article is part of a Special Issue entitled: From protein structures to clinical applications.


Subject(s)
Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Signal Transduction/physiology , Animals , Mice , Phosphorylation/physiology
8.
J Muscle Res Cell Motil ; 33(6): 439-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22527640

ABSTRACT

Following activation by G-protein-coupled receptor agonists, protein kinase C (PKC) modulates cardiac myocyte function by phosphorylation of intracellular targets including myofilament proteins cardiac troponin I (cTnI) and cardiac myosin binding protein C (cMyBP-C). Since PKC phosphorylation has been shown to decrease myofibril ATPase activity, we hypothesized that PKC phosphorylation of cTnI and cMyBP-C will lower myocyte power output and, in addition, attenuate the elevation in power in response to protein kinase A (PKA)-mediated phosphorylation. We compared isometric force and power generating capacity of rat skinned cardiac myocytes before and after treatment with the catalytic subunit of PKC. PKC increased phosphorylation levels of cMyBP-C and cTnI and decreased both maximal Ca(2+) activated force and Ca(2+) sensitivity of force. Moreover, during submaximal Ca(2+) activations PKC decreased power output by 62 %, which arose from both the fall in force and slower loaded shortening velocities since depressed power persisted even when force levels were matched before and after PKC. In addition, PKC blunted the phosphorylation of cTnI by PKA, reduced PKA-induced spontaneous oscillatory contractions, and diminished PKA-mediated elevations in myocyte power. To test whether altered thin filament function plays an essential role in these contractile changes we investigated the effects of chronic cTnI pseudo-phosphorylation on myofilament function using myocyte preparations from transgenic animals in which either only PKA phosphorylation sites (Ser-23/Ser-24) (PP) or both PKA and PKC phosphorylation sites (Ser-23/Ser-24/Ser-43/Ser-45/T-144) (All-P) were replaced with aspartic acid. Cardiac myocytes from All-P transgenic mice exhibited reductions in maximal force, Ca(2+) sensitivity of force, and power. Similarly diminished power generating capacity was observed in hearts from All-P mice as determined by in situ pressure-volume measurements. These results imply that PKC-mediated phosphorylation of cTnI plays a dominant role in depressing contractility, and, thus, increased PKC isozyme activity may contribute to maladaptive behavior exhibited during the progression to heart failure.


Subject(s)
Cardiac Output/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Animals , Male , Myocytes, Cardiac/enzymology , Rats , Rats, Sprague-Dawley
9.
Am J Physiol Heart Circ Physiol ; 303(1): H9-18, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22523251

ABSTRACT

Proteasomes are ubiquitously expressed multicatalytic complexes that serve as key regulators of protein homeostasis. There are several lines of evidence indicating that proteasomes exist in heterogeneous subpopulations in cardiac muscle, differentiated, in part, by post-translational modifications (PTMs). PTMs regulate numerous facets of proteasome function, including catalytic activities, complex assembly, interactions with associating partners, subcellular localization, substrate preference, and complex turnover. Classical technologies used to identify PTMs on proteasomes have lacked the ability to determine site specificity, quantify stoichiometry, and perform large-scale, multi-PTM analysis. Recent advancements in proteomic technologies have largely overcome these limitations. We present here a discussion on the importance of PTMs in modulating proteasome function in cardiac physiology and pathophysiology, followed by the presentation of a state-of-the-art proteomic workflow for identifying and quantifying PTMs of cardiac proteasomes.


Subject(s)
Heart/physiology , Myocardium/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Proteomics , Animals , Computational Biology , Humans , Molecular Sequence Data , Myocytes, Cardiac/physiology , Signal Transduction/physiology , Subcellular Fractions/metabolism
10.
J Proteomics ; 75(15): 4602-9, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22387130

ABSTRACT

The regulation of mitochondrial function is essential for cardiomyocyte adaptation to cellular stress. While it has long been understood that phosphorylation regulates flux through metabolic pathways, novel phosphorylation sites are continually being discovered in all functionally distinct areas of the mitochondrial proteome. Extracting biologically meaningful information from these phosphorylation sites requires an adaptable, sensitive, specific and robust method for their quantification. Here we report a multiple reaction monitoring-based mass spectrometric workflow for quantifying site-specific phosphorylation of mitochondrial proteins. Specifically, chromatographic and mass spectrometric conditions for 68 transitions derived from 23 murine and human phosphopeptides, and their corresponding unmodified peptides, were optimized. These methods enabled the quantification of endogenous phosphopeptides from the outer mitochondrial membrane protein VDAC, and the inner membrane proteins ANT and ETC complexes I, III and V. The development of this quantitative workflow is a pivotal step for advancing our knowledge and understanding of the regulatory effects of mitochondrial protein phosphorylation in cardiac physiology and pathophysiology. This article is part of a Special Issue entitled: Translational Proteomics.


Subject(s)
Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Proteomics/methods , Animals , Humans , Mice , Phosphorylation
11.
Mol Cell Biochem ; 363(1-2): 203-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22160857

ABSTRACT

Myocardial physiology in the aftermath of myocardial infarction (MI) before remodeling is an under-explored area of investigation. Here, we describe the effects of MI on the cardiac sarcomere with focus on the possible contributions of reactive oxygen species. We surgically induced MI in 6-7-month-old female CD1 mice by ligation of the left anterior descending coronary artery. Data were collected 3-4 days after MI or sham (SH) surgery. MI hearts demonstrated ventricular dilatation and systolic dysfunction upon echo cardiographic analysis. Sub-maximum Ca-activated tension in detergent-extracted fiber bundles from papillary muscles increased significantly in the preparations from MI hearts. Ca(2+) sensitivity increased after MI, whereas cooperativity of activation decreased. To assess myosin enzymatic integrity we measured splitting of Ca-ATP in myofibrillar preparations, which demonstrated a decline in Ca-ATPase activity of myofilament myosin. Biochemical analysis demonstrated post-translational modification of sarcomeric proteins. Phosphorylation of cardiac troponin I and myosin light chain 2 was reduced after MI in papillary samples, as measured using a phospho-specific stain. Tropomyosin was oxidized after MI, forming disulfide products detectable by diagonal non-reducing-reducing SDS-PAGE. Our analysis of myocardial protein oxidation post-MI also demonstrated increased S-glutathionylation. We functionally linked protein oxidation with sarcomere function by treating skinned fibers with the sulfhydryl reducing agent dithiothreitol, which reduced Ca(2+) sensitivity in MI, but not SH, samples. Our data indicate important structural and functional alterations to the cardiac sarcomere after MI, and the contribution of protein oxidation to this process.


Subject(s)
Muscle Proteins/metabolism , Myocardial Contraction , Myocardial Infarction/metabolism , Papillary Muscles/metabolism , Protein Processing, Post-Translational , Sarcomeres/metabolism , Ventricular Function, Left , Amino Acid Sequence , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Cardiac Myosins/metabolism , Disease Models, Animal , Dithiothreitol/pharmacology , Electrophoresis, Polyacrylamide Gel , Female , Glutathione/metabolism , Mice , Molecular Sequence Data , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Myosin Light Chains/metabolism , Oxidation-Reduction , Papillary Muscles/drug effects , Papillary Muscles/physiopathology , Phosphorylation , Reactive Oxygen Species/metabolism , Reducing Agents/pharmacology , Sarcomeres/drug effects , Stroke Volume , Tropomyosin/metabolism , Troponin I/metabolism , Ultrasonography
12.
Physiology (Bethesda) ; 26(2): 106-14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21487029

ABSTRACT

Proteasome biology has taken central stage in cardiac physiology and pathophysiology. The molecular heterogeneity of proteasome subpopulations supports the specificity of proteasome function to degrade diverse substrate repertoires. Unveiling the dynamics of proteasome function should inspire new therapeutic strategies for combating cardiac disease.


Subject(s)
Heart/physiology , Myocardium/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Humans , Myocardium/chemistry , Proteasome Endopeptidase Complex/chemistry , Signal Transduction , Substrate Specificity
13.
Mol Cell Proteomics ; 10(5): M110.006122, 2011 May.
Article in English | MEDLINE | ID: mdl-21357515

ABSTRACT

Myocardial proteasomes are comprised of 20S core particles and 19S regulatory particles, which together carry out targeted degradation of cardiac proteins. The 19S complex is unique among the regulators of proteasomes in that it affects both the capacity and specificity of protein degradation. However, a comprehensive molecular characterization of cardiac 19S complexes is lacking. In this investigation, we tailored a multidimensional chromatography-based purification strategy to isolate structurally intact and functionally viable 19S complexes from murine hearts. Two distinct subpopulations of 19S complexes were isolated based upon (1) potency of activating 20S proteolytic activity, and (2) molecular composition using a combination of immuno-detection, two-dimensional-differential gel electrophoresis, and MS-based approaches. Heat shock protein 90 (Hsp90) was identified to be characteristic to 19S subpopulation I. The physical interaction of Hsp90 with 19S complexes was demonstrated via multiple approaches. Inhibition of Hsp90 activity using geldanamycin or BIIB021 potentiated the ability of subpopulation I to activate 20S proteasomes in the murine heart, thus demonstrating functional specificity of Hsp90 in subpopulation I. This investigation has advanced our understanding of the molecular heterogeneity of cardiac proteasomes by identifying molecularly and functionally distinct cardiac 19S complexes. The preferential association of Hsp90 with 19S subpopulation I unveils novel targets for designing proteasome-based therapeutic interventions for combating cardiac disease.


Subject(s)
Myocardium/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Amino Acid Sequence , Animals , Chromatography, Liquid , Enzyme Activation , HSP90 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred ICR , Molecular Sequence Data , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/isolation & purification , Tandem Mass Spectrometry
14.
Arch Biochem Biophys ; 510(2): 129-34, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21345328

ABSTRACT

It has been over 35 years since the first identification of phosphorylation of myosin light chains in skeletal and cardiac muscle. Yet only in the past few years has the role of these phosphorylations in cardiac dynamics been more fully understood. Advances in this understanding have come about with further evidence on the control mechanisms regulating the level of phosphorylation by kinases and phosphatases. Moreover, studies clarifiying the role of light chain phosphorylation in short and long term control of cardiac contractility and as a factor in cardiac remodeling have improved our knowledge. Especially important in these advances has been the use of gain and loss of function approaches, which have not only testedthe role of kinases and phosphatases, but also the effects of loss of RLC phosphorylation sites. Major conclusions from these studies indicate that (i) two negatively-charged post-translational modifications occupy the ventricular RLC N-terminus, with mouse RLC being doubly phosphorylated (Ser 14/15), and human RLC being singly phosphorylated (Ser 15) and singly deamidated(Asn14/16 to Asp); (ii)a distinct cardiac myosin light kinase (cMLCK) and a unique myosin phosphatase targeting peptide (MYPT2) control phosphoryl group transfer;and (iii) ablation of RLC phosphorylationdecreases ventricular power, lengthens the duration of ventricular ejection, and may also modify other sarcomeric proteins (e.g., troponin I) as substrates for kinases and/or phosphatases. A long term effect of low levels of RLC phosphorylation in mouse models also involves remodeling of the heart with hypertrophy, depressed contractility, and sarcomeric disarray. Data demonstrating altered levels of RLC phosphorylation in comparisons of samples from normal and stressed human hearts indicate the significance of these findings in translational medicine.


Subject(s)
Heart/physiology , Myosin Light Chains/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Myosin Light Chains/chemistry , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase/chemistry , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation
16.
Mol Cell Proteomics ; 9(9): 1804-18, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20445002

ABSTRACT

The molecular conformation of the cardiac myosin motor is modulated by intermolecular interactions among the heavy chain, the light chains, myosin binding protein-C, and titin and is governed by post-translational modifications (PTMs). In-gel digestion followed by LC/MS/MS has classically been applied to identify cardiac sarcomeric PTMs; however, this approach is limited by protein size, pI, and difficulties in peptide extraction. We report a solution-based work flow for global separation of endogenous cardiac sarcomeric proteins with a focus on the regulatory light chain (RLC) in which specific sites of phosphorylation have been unclear. Subcellular fractionation followed by OFFGEL electrophoresis resulted in isolation of endogenous charge variants of sarcomeric proteins, including regulatory and essential light chains, myosin heavy chain, and myosin-binding protein-C of the thick filament. Further purification of RLC using reverse-phase HPLC separation and UV detection enriched for RLC PTMs at the intact protein level and provided a stoichiometric and quantitative assessment of endogenous RLC charge variants. Digestion and subsequent LC/MS/MS unequivocally identified that the endogenous charge variants of cardiac RLC focused in unique OFFGEL electrophoresis fractions were unphosphorylated (78.8%), singly phosphorylated (18.1%), and doubly phosphorylated (3.1%) RLC. The novel aspects of this study are that 1) milligram amounts of endogenous cardiac sarcomeric subproteome were focused with resolution comparable with two-dimensional electrophoresis, 2) separation and quantification of post-translationally modified variants were achieved at the intact protein level, 3) separation of intact high molecular weight thick filament proteins was achieved in solution, and 4) endogenous charge variants of RLC were separated; a novel doubly phosphorylated form was identified in mouse, and singly phosphorylated, singly deamidated, and deamidated/phosphorylated forms were identified and quantified in human non-failing and failing heart samples, thus demonstrating the clinical utility of the method.


Subject(s)
Muscle Proteins/isolation & purification , Myocardium/chemistry , Sarcomeres/chemistry , Adult , Chromatography, High Pressure Liquid , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Male , Middle Aged , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Phosphorylation , Protein Conformation , Spectrophotometry, Ultraviolet , Tandem Mass Spectrometry
17.
J Biol Chem ; 284(8): 5097-106, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19106098

ABSTRACT

There is little direct evidence on the role of myosin regulatory light chain phosphorylation in ejecting hearts. In studies reported here we determined the effects of regulatory light chain (RLC) phosphorylation on in situ cardiac systolic mechanics and in vitro myofibrillar mechanics. We compared data obtained from control nontransgenic mice (NTG) with a transgenic mouse model expressing a cardiac specific nonphosphorylatable RLC (TG-RLC(P-). We also determined whether the depression in RLC phosphorylation affected phosphorylation of other sarcomeric proteins. TG-RLC(P-) demonstrated decreases in base-line load-independent measures of contractility and power and an increase in ejection duration together with a depression in phosphorylation of myosin-binding protein-C (MyBP-C) and troponin I (TnI). Although TG-RLC(P-) displayed a significantly reduced response to beta(1)-adrenergic stimulation, MyBP-C and TnI were phosphorylated to a similar level in TG-RLC(P-) and NTG, suggesting cAMP-dependent protein kinase signaling to these proteins was not disrupted. A major finding was that NTG controls were significantly phosphorylated at RLC serine 15 following beta(1)-adrenergic stimulation, a mechanism prevented in TG-RLC(P-), thus providing a biochemical difference in beta(1)-adrenergic responsiveness at the level of the sarcomere. Our measurements of Ca(2+) tension and Ca(2+)-ATPase rate relations in detergent-extracted fiber bundles from LV trabeculae demonstrated a relative decrease in maximum Ca(2+)-activated tension and tension cost in TG-RLC(P-) fibers, with no change in Ca(2+) sensitivity. Our data indicate that RLC phosphorylation is critical for normal ejection and response to beta(1)-adrenergic stimulation. Our data also indicate that the lack of RLC phosphorylation promotes compensatory changes in MyBP-C and TnI phosphorylation, which when normalized do not restore function.


Subject(s)
Actin Cytoskeleton/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Diseases/metabolism , Myocardial Contraction , Myocardium/metabolism , Myosin Light Chains/metabolism , Actin Cytoskeleton/genetics , Animals , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Heart Diseases/genetics , Heart Ventricles/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/genetics , Myosin Light Chains/genetics , Phosphorylation/genetics , Sarcomeres/genetics , Sarcomeres/metabolism , Stroke Volume/genetics , Troponin I/genetics , Troponin I/metabolism
18.
J Mol Cell Cardiol ; 40(4): 465-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16445938

ABSTRACT

We have previously reported a transgenic mouse that over-expresses constitutively active PKCepsilon in the myocardium and exhibits a steady progression to heart failure. Associated with the decline in function was an increased phosphorylation of sarcomeric proteins including cardiac troponin I (cTnI). To determine whether PKCepsilon phosphorylation of cTnI is sufficient to induce cardiac maladaptation, we have generated a double transgenic mouse (DbTG) that expresses constitutively active PKCepsilon and cTnI harboring non-phosphorylatable mutations in the putative PKC phosphorylation sites (S43A, S45A). We compared the hemodynamic and biochemical properties of the hearts from the DbTG mice to the non-transgenic and single transgenic lines at both 3 and 12 months of age. While no significant differences in LV function were noted in 3-month groups, the depression of function in the PKCepsilon mice was attenuated in the double transgenic mice at 12 months. The improvement in cardiac function was correlated with decreased beta-myosin heavy chain and ANF mRNA expression in the 12m DbTG mice. The extent of cTnI phosphorylation was determined using a novel one-dimensional, non-equilibrium isoelectric focusing technique. At 3 months the migration of cTnI phospho-species was different in the PKCepsilon mice and to a lesser degree in the DbTG compared to all other groups. At 12 months additional phospho-species were observed in both the PKCepsilon and DbTG samples, along with an overall shift in the distribution of phospho-species in all groups due to age. These results suggest that phosphorylation of cTnI by PKCepsilon is associated with contractile dysfunction and partial replacement of serines 43/45 improves cardiac performance. Therefore, we conclude that phosphorylation of cTnI at Ser 43 and 45 may contribute to the progression of failure.


Subject(s)
Amino Acid Substitution , Myocardial Contraction , Myocardium/metabolism , Protein Kinase C-epsilon/metabolism , Protein Processing, Post-Translational , Troponin I/metabolism , Aging/genetics , Aging/metabolism , Animals , Cell Line , Heart Failure/genetics , Heart Failure/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/genetics , Myocardium/pathology , Phosphorylation , Protein Processing, Post-Translational/genetics , Troponin I/genetics , Ventricular Function, Left/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...