Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Circ Heart Fail ; 12(11): e005835, 2019 11.
Article in English | MEDLINE | ID: mdl-31684756

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a genetic cardiovascular disorder, primarily involving mutations in sarcomeric proteins. HCM patients present with hypertrophy, diastolic dysfunction, and fibrosis, but there is no specific treatment. The sphingosine-1-phosphate receptor modulator, FTY720/fingolimod, is approved for treatment of multiple sclerosis. We hypothesize that modulation of the sphingosine-1-phosphate receptor by FTY720 would be of therapeutic benefit in sarcomere-linked HCM. METHODS: We treated mice with an HCM-linked mutation in tropomyosin (Tm-E180G) and nontransgenic littermates with FTY720 or vehicle for 6 weeks. Compared with vehicle-treated, FTY720-treated Tm-E180G mice had a significant reduction in left atrial size (1.99±0.19 [n=7] versus 2.70±0.44 [n=6] mm; P<0.001) and improvement in diastolic function (E/A ratio: 2.69±0.38 [n=7] versus 5.34±1.19 [n=6]; P=0.004) as assessed by echocardiography. RESULTS: Pressure-volume relations revealed significant improvements in the end-diastolic pressure volume relationship, relaxation kinetics, preload recruitable stroke work, and ejection fraction. Detergent-extracted fiber bundles revealed a significant decrease in myofilament Ca2+-responsiveness (pCa50=6.15±0.11 [n=13] versus 6.24±0.06 [n=14]; P=0.041). We attributed these improvements to a downregulation of S-glutathionylation of cardiac myosin binding protein-C in FTY720-treated Tm-E180G mice and reduction in oxidative stress by downregulation of NADPH oxidases with no changes in fibrosis. CONCLUSIONS: This is the first demonstration that modulation of S1PR results in decreased myofilament-Ca2+-responsiveness and improved diastolic function in HCM. We associated these changes with decreased oxidative modification of myofilament proteins via downregulation of NOX2. Our data support the hypothesis that modification of sphingolipid signaling may be a novel therapeutic approach in HCM.


Subject(s)
Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Cardiomyopathy, Hypertrophic/drug therapy , Fingolimod Hydrochloride/pharmacology , Myocytes, Cardiac/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine-1-Phosphate Receptors/drug effects , Animals , Calcium Signaling/drug effects , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , Diastole , Disease Models, Animal , Female , Fibrosis , Genetic Predisposition to Disease , Male , Mice, Mutant Strains , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/drug effects , Myofibrils/metabolism , Myofibrils/pathology , Oxidative Stress/drug effects , Phenotype , Sphingosine-1-Phosphate Receptors/metabolism , Tropomyosin/genetics
2.
J Biol Chem ; 294(8): 2913-2923, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30567734

ABSTRACT

Phosphorylation of cardiac sarcomeric proteins plays a major role in the regulation of the physiological performance of the heart. Phosphorylation of thin filament proteins, such as troponin I and T, dramatically affects calcium sensitivity of the myofiber and systolic and diastolic functions. Phosphorylation of the regulatory protein tropomyosin (Tpm) results in altered biochemical properties of contraction; however, little is known about the physiological effect of Tpm phosphorylation on cardiac function. To address the in vivo significance of Tpm phosphorylation, here we generated transgenic mouse lines having a phosphomimetic substitution in the phosphorylation site of α-Tpm (S283D). High expression of Tpm S283D variant in one transgenic mouse line resulted in an increased heart:body weight ratio, coupled with a severe dilated cardiomyopathic phenotype resulting in death within 1 month of birth. Moderate Tpm S283D mice expression in other lines caused mild myocyte hypertrophy and fibrosis, did not affect lifespan, and was coupled with decreased expression of extracellular signal-regulated kinase 1/2 kinase signaling. Physiological analysis revealed that the transgenic mice exhibit impaired diastolic function, without changes in systolic performance. Surprisingly, we observed no alterations in calcium sensitivity of the myofibers, cooperativity, or calcium-ATPase activity in the myofibers. Our experiments also disclosed that casein kinase 2 plays an integral role in Tpm phosphorylation. In summary, increased expression of pseudo-phosphorylated Tpm impairs diastolic function in the intact heart, without altering calcium sensitivity or cooperativity of myofibers. Our findings provide the first extensive in vivo assessment of Tpm phosphorylation in the heart and its functional role in cardiac performance.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Cardiomyopathy, Dilated/pathology , Tropomyosin/physiology , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/metabolism , Cells, Cultured , Mice , Mice, Transgenic , Mutation , Myofibrils/metabolism , Myofibrils/pathology , Phosphorylation
3.
Cardiovasc Res ; 113(8): 915-925, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28379313

ABSTRACT

BACKGROUND: Dilated cardiomoypathies (DCM) are a heterogeneous group of inherited and acquired diseases characterized by decreased contractility and enlargement of cardiac chambers and a major cause of morbidity and mortality. Mice with Glu54Lys mutation in α-tropomyosin (Tm54) demonstrate typical DCM phenotype with reduced myofilament Ca2+ sensitivity. We tested the hypothesis that early sensitization of the myofilaments to Ca2+ in DCM can prevent the DCM phenotype. METHODS AND RESULTS: To sensitize Tm54 myofilaments, we used a genetic approach and crossbred Tm54 mice with mice expressing slow skeletal troponin I (ssTnI) that sensitizes myofilaments to Ca2+. Four groups of mice were used: non-transgenic (NTG), Tm54, ssTnI and Tm54/ssTnI (DTG). Systolic function was significantly reduced in the Tm54 mice compared to NTG, but restored in DTG mice. Tm54 mice also showed increased diastolic LV dimensions and HW/BW ratios, when compared to NTG, which were improved in the DTG group. ß-myosin heavy chain expression was increased in the Tm54 animals compared to NTG and was partially restored in DTG group. Analysis by 2D-DIGE indicated a significant decrease in two phosphorylated spots of cardiac troponin I (cTnI) in the DTG animals compared to NTG and Tm54. Analysis by 2D-DIGE also indicated no significant changes in troponin T, regulatory light chain, myosin binding protein C and tropomyosin phosphorylation. CONCLUSION: Our data indicate that decreased myofilament Ca2+ sensitivity is an essential element in the pathophysiology of thin filament linked DCM. Sensitization of myofilaments to Ca2+ in the early stage of DCM may be a useful therapeutic strategy in thin filament linked DCM.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathy, Dilated/genetics , Animals , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice, Transgenic , Myocardium/metabolism , Phosphorylation , Tropomyosin/metabolism , Ventricular Myosins/genetics
4.
Am J Physiol Heart Circ Physiol ; 309(10): H1720-30, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26432840

ABSTRACT

S-glutathionylation of cardiac myosin-binding protein C (cMyBP-C) induces Ca(2+) sensitization and a slowing of cross-bridge kinetics as a result of increased oxidative signaling. Although there is evidence for a role of oxidative stress in disorders associated with hypertrophic cardiomyopathy (HCM), this mechanism is not well understood. We investigated whether oxidative myofilament modifications may be in part responsible for diastolic dysfunction in HCM. We administered N-acetylcysteine (NAC) for 30 days to 1-mo-old wild-type mice and to transgenic mice expressing a mutant tropomyosin (Tm-E180G) and nontransgenic littermates. Tm-E180G hearts demonstrate a phenotype similar to human HCM. After NAC administration, the morphology and diastolic function of Tm-E180G mice was not significantly different from controls, indicating that NAC had reversed baseline diastolic dysfunction and hypertrophy in our model. NAC administration also increased sarco(endo)plasmic reticulum Ca(2+) ATPase protein expression, reduced extracellular signal-related kinase 1/2 phosphorylation, and normalized phosphorylation of phospholamban, as assessed by Western blot. Detergent-extracted fiber bundles from NAC-administered Tm-E180G mice showed nearly nontransgenic (NTG) myofilament Ca(2+) sensitivity. Additionally, we found that NAC increased tension cost and rate of cross-bridge reattachment. Tm-E180G myofilaments were found to have a significant increase in S-glutathionylation of cMyBP-C, which was returned to NTG levels upon NAC administration. Taken together, our results indicate that oxidative myofilament modifications are an important mediator in diastolic function, and by relieving this modification we were able to reverse established diastolic dysfunction and hypertrophy in HCM.


Subject(s)
Acetylcysteine/pharmacology , Cardiomyopathy, Hypertrophic, Familial/metabolism , Diastole/drug effects , Free Radical Scavengers/pharmacology , Heart Ventricles/drug effects , Myofibrils/drug effects , Animals , Calcium/metabolism , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Disease Models, Animal , Female , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Myofibrils/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Tropomyosin/genetics
5.
Cardiovasc Res ; 107(2): 226-34, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26045475

ABSTRACT

AIMS: Therapeutic approaches to treat familial dilated cardiomyopathy (DCM), which is characterized by depressed sarcomeric tension and susceptibility to Ca(2+)-related arrhythmias, have been generally unsuccessful. Our objective in the present work was to determine the effect of the angiotensin II type 1 receptor (AT1R) biased ligand, TRV120023, on contractility of hearts of a transgenic mouse model of familial DCM with mutation in tropomyosin at position 54 (TG-E54K). Our rationale is based on previous studies, which have supported the hypothesis that biased G-protein-coupled receptor ligands, signalling via ß-arrestin, increase cardiac contractility with no effect on Ca(2+) transients. Our previous work demonstrated that the biased ligand TRV120023 is able to block angiotensin-induced hypertrophy, while promoting an increase in sarcomere Ca(2+) response. METHODS AND RESULTS: We tested the hypothesis that the depression in cardiac function associated with DCM can be offset by infusion of the AT1R biased ligand, TRV120023. We intravenously infused saline, TRV120023, or the unbiased ligand, losartan, for 15 min in TG-E54K and non-transgenic mice to obtain left ventricular pressure-volume relations. Hearts were analysed for sarcomeric protein phosphorylation. Results showed that the AT1R biased ligand increases cardiac performance in TG-E54K mice in association with increased myosin light chain-2 phosphorylation. CONCLUSION: Treatment of mice with an AT1R biased ligand, acting via ß-arrestin signalling, is able to induce an increase in cardiac contractility associated with an increase in ventricular myosin light chain-2 phosphorylation. AT1R biased ligands may prove to be a novel inotropic approach in familial DCM.


Subject(s)
Cardiac Myosins/metabolism , Cardiomyopathy, Dilated/metabolism , Myocardial Contraction/physiology , Myosin Light Chains/metabolism , Oligopeptides/metabolism , Animals , Arrestins/metabolism , Disease Models, Animal , Female , Heart/physiopathology , Ligands , Male , Mice, Transgenic , Phosphorylation , beta-Arrestins
6.
Basic Res Cardiol ; 109(6): 445, 2014.
Article in English | MEDLINE | ID: mdl-25280528

ABSTRACT

Although ceramide accumulation in the heart is considered a major factor in promoting apoptosis and cardiac disorders, including heart failure, lipotoxicity and ischemia-reperfusion injury, little is known about ceramide's role in mediating changes in contractility. In the present study, we measured the functional consequences of acute exposure of isolated field-stimulated adult rat cardiomyocytes to C6-ceramide. Exogenous ceramide treatment depressed the peak amplitude and the maximal velocity of shortening without altering intracellular calcium levels or kinetics. The inactive ceramide analog C6-dihydroceramide had no effect on myocyte shortening or [Ca(2+)]i transients. Experiments testing a potential role for C6-ceramide-mediated effects on activation of protein kinase C (PKC) demonstrated evidence for signaling through the calcium-independent isoform, PKCε. We employed 2-dimensional electrophoresis and anti-phospho-peptide antibodies to test whether treatment of the cardiomyocytes with C6-ceramide altered myocyte shortening via PKC-dependent phosphorylation of myofilament proteins. Compared to controls, myocytes treated with ceramide exhibited increased phosphorylation of myosin binding protein-C (cMyBP-C), specifically at Ser273 and Ser302, and troponin I (cTnI) at sites apart from Ser23/24, which could be attenuated with PKC inhibition. We conclude that the altered myofilament response to calcium resulting from multiple sites of PKC-dependent phosphorylation contributes to contractile dysfunction that is associated with cardiac diseases in which elevations in ceramides are present.


Subject(s)
Ceramides/physiology , Myocytes, Cardiac/physiology , Myofibrils/metabolism , Protein Kinase C/physiology , Animals , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction
7.
J Mol Cell Cardiol ; 77: 53-63, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25280781

ABSTRACT

The mechanisms linking the expression of sarcomeric mutant proteins to the development of pathological hypertrophy in hypertrophic cardiomyopathy (HCM) remain poorly understood. We investigated the role of the plasma membrane Ca(2+)-ATPase PMCA4 in the HCM phenotype using a transgenic model that expresses mutant (Glu180Gly) α-tropomyosin (Tm180) in heart. Immunoblot analysis revealed that cardiac PMCA4 expression was upregulated early in Tm180 disease pathogenesis. This was accompanied by an increase in levels of the L-type Ca(2+)-channel, which is implicated in pathological hypertrophy. When Tm180 mice were crossed with a PMCA4-null line, loss of PMCA4 caused the abrogation of hypertrophy in Tm180/PMCA4-null double mutant mice. RT-PCR analysis of Tm180/PMCA4-null hearts revealed blunting of the fetal program and reversion of pro-fibrotic Col1a1 and Col3a1 gene expression to wild-type levels. This was accompanied by evidence of reduced L-type Ca(2+)-channel expression, and diminished calcineurin activity. Expression of the metabolic substrate transporters glucose transporter 4 and carnitine palmitoyltransferase 1b was preserved and Tm180-related changes in mRNA levels of various contractile stress-related proteins including the cardiac ankyrin protein CARP and the N2B isoform of titin were reversed in Tm180/PMCA4-null hearts. cGMP levels were increased and phosphorylation of vasodilator-stimulated phosphoprotein was elevated in Tm180/PMCA4-null hearts. These changes were associated with a sharp reduction in left ventricular end-diastolic pressure in Tm180/PMCA4-null hearts, which occurred despite persistence of Tm180-related impairment of relaxation dynamics. These results reveal a novel and specific role for PMCA4 in the Tm180 hypertrophic phenotype, with the "protective" effects of PMCA4 deficiency encompassing multiple determinants of HCM-related hypertrophy.


Subject(s)
Cardiomyopathy, Hypertrophic/enzymology , Plasma Membrane Calcium-Transporting ATPases/genetics , Tropomyosin/genetics , Animals , Cardiomyopathy, Hypertrophic/genetics , Disease Models, Animal , Gene Expression , Gene Knockout Techniques , Heart Rate , Male , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Tropomyosin/metabolism , Ventricular Pressure
8.
Circ Cardiovasc Genet ; 7(2): 132-143, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24585742

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a common genetic disorder caused mainly by mutations in sarcomeric proteins and is characterized by maladaptive myocardial hypertrophy, diastolic heart failure, increased myofilament Ca(2+) sensitivity, and high susceptibility to sudden death. We tested the following hypothesis: correction of the increased myofilament sensitivity can delay or prevent the development of the HCM phenotype. METHODS AND RESULTS: We used an HCM mouse model with an E180G mutation in α-tropomyosin (Tm180) that demonstrates increased myofilament Ca(2+) sensitivity, severe hypertrophy, and diastolic dysfunction. To test our hypothesis, we reduced myofilament Ca(2+) sensitivity in Tm180 mice by generating a double transgenic mouse line. We crossed Tm180 mice with mice expressing a pseudophosphorylated cardiac troponin I (S23D and S24D; TnI-PP). TnI-PP mice demonstrated a reduced myofilament Ca(2+) sensitivity compared with wild-type mice. The development of pathological hypertrophy did not occur in mice expressing both Tm180 and TnI-PP. Left ventricle performance was improved in double transgenic compared with their Tm180 littermates, which express wild-type cardiac troponin I. Hearts of double transgenic mice demonstrated no changes in expression of phospholamban and sarcoplasmic reticulum Ca(2+) ATPase, increased levels of phospholamban and troponin T phosphorylation, and reduced phosphorylation of TnI compared with Tm180 mice. Moreover, expression of TnI-PP in Tm180 hearts inhibited modifications in the activity of extracellular signal-regulated kinase and zinc finger-containing transcription factor GATA in Tm180 hearts. CONCLUSIONS: Our data strongly indicate that reduction of myofilament sensitivity to Ca(2+) and associated correction of abnormal relaxation can delay or prevent development of HCM and should be considered as a therapeutic target for HCM.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Myofibrils/metabolism , Tropomyosin/genetics , Troponin I/genetics , Animals , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Hypertrophic/therapy , Humans , Mice , Mice, Transgenic , Mutation , Phosphorylation , Tropomyosin/metabolism , Troponin I/metabolism , Troponin T/metabolism
9.
J Biol Chem ; 289(13): 8818-27, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24509847

ABSTRACT

Myocardial infarction (MI) is associated with depressed cardiac contractile function and progression to heart failure. Cardiac myosin-binding protein C, a cardiac-specific myofilament protein, is proteolyzed post-MI in humans, which results in an N-terminal fragment, C0-C1f. The presence of C0-C1f in cultured cardiomyocytes results in decreased Ca(2+) transients and cell shortening, abnormalities sufficient for the induction of heart failure in a mouse model. However, the underlying mechanisms remain unclear. Here, we investigate the association between C0-C1f and altered contractility in human cardiac myofilaments in vitro. To accomplish this, we generated recombinant human C0-C1f (hC0C1f) and incorporated it into permeabilized human left ventricular myocardium. Mechanical properties were studied at short (2 µm) and long (2.3 µm) sarcomere length (SL). Our data demonstrate that the presence of hC0C1f in the sarcomere had the greatest effect at short, but not long, SL, decreasing maximal force and myofilament Ca(2+) sensitivity. Moreover, hC0C1f led to increased cooperative activation, cross-bridge cycling kinetics, and tension cost, with greater effects at short SL. We further established that the effects of hC0C1f occur through direct interaction with actin and α-tropomyosin. Our data demonstrate that the presence of hC0C1f in the sarcomere is sufficient to induce depressed myofilament function and Ca(2+) sensitivity in otherwise healthy human donor myocardium. Decreased cardiac function post-MI may result, in part, from the ability of hC0C1f to bind actin and α-tropomyosin, suggesting that cleaved C0-C1f could act as a poison polypeptide and disrupt the interaction of native cardiac myosin-binding protein C with the thin filament.


Subject(s)
Carrier Proteins/chemistry , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Peptide Fragments/metabolism , Sarcomeres/metabolism , Actins/metabolism , Actomyosin/metabolism , Adenosine Triphosphatases/metabolism , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Humans , Kinetics , Mice , Proteolysis , Tropomyosin/metabolism
10.
J Biol Chem ; 288(40): 28925-35, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23960072

ABSTRACT

Studies indicate that tropomyosin (Tm) phosphorylation status varies in different mouse models of cardiac disease. Investigation of basal and acute cardiac function utilizing a mouse model expressing an α-Tm protein that cannot be phosphorylated (S283A) shows a compensated hypertrophic phenotype with significant increases in SERCA2a expression and phosphorylation of phospholamban Ser-16 (Schulz, E. M., Correll, R. N., Sheikh, H. N., Lofrano-Alves, M. S., Engel, P. L., Newman, G., Schultz Jel, J., Molkentin, J. D., Wolska, B. M., Solaro, R. J., and Wieczorek, D. F. (2012) J. Biol. Chem. 287, 44478-44489). With these results, we hypothesized that decreasing α-Tm phosphorylation may be beneficial in the context of a chronic, intrinsic stressor. To test this hypothesis, we utilized the familial hypertrophic cardiomyopathy (FHC) α-Tm E180G model (Prabhakar, R., Boivin, G. P., Grupp, I. L., Hoit, B., Arteaga, G., Solaro, R. J., and Wieczorek, D. F. (2001) J. Mol. Cell. Cardiol. 33, 1815-1828). These FHC hearts are characterized by increased heart:body weight ratios, fibrosis, increased myofilament Ca(2+) sensitivity, and contractile defects. The FHC mice die by 6-8 months of age. We generated mice expressing both the E180G and S283A mutations and found that the hypertrophic phenotype was rescued in the α-Tm E180G/S283A double mutant transgenic animals; these mice exhibited no signs of cardiac hypertrophy and displayed improved cardiac function. These double mutant transgenic hearts showed increased phosphorylation of phospholamban Ser-16 and Thr-17 compared with the α-Tm E180G mice. This is the first study to demonstrate that decreasing phosphorylation of tropomyosin can rescue a hypertrophic cardiomyopathic phenotype.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/metabolism , Tropomyosin/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Cardiomyopathy, Hypertrophic, Familial/diagnostic imaging , Cardiomyopathy, Hypertrophic, Familial/pathology , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Gene Expression Regulation , Heart Function Tests , Immunoblotting , Mice , Mice, Transgenic , Mutant Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Phosphorylation , Ultrasonography
11.
Physiol Genomics ; 45(17): 764-73, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23800848

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) is a disease characterized by ventricular hypertrophy, fibrosis, and aberrant systolic and/or diastolic function. Our laboratories have previously developed two mouse models that affect cardiac performance. One mouse model encodes an FHC-associated mutation in α-tropomyosin: Glu → Gly at amino acid 180, designated as Tm180. These mice display a phenotype that is characteristic of FHC, including severe cardiac hypertrophy with fibrosis and impaired physiological performance. The other model was a gene knockout of phospholamban (PLN KO), a regulator of calcium uptake in the sarcoplasmic reticulum of cardiomyocytes; these hearts exhibit hypercontractility with no pathological abnormalities. Previous work in our laboratories shows that when mice were genetically crossed between the PLN KO and Tm180, the progeny (PLN KO/Tm180) display a rescued hypertrophic phenotype with improved morphology and cardiac function. To understand the changes in gene expression that occur in these models undergoing cardiac remodeling (Tm180, PLN KO, PLN KO/Tm180, and nontransgenic control mice), we conducted microarray analyses of left ventricular tissue at 4 and 12 mo of age. Expression profiling reveals that 1,187 genes changed expression in direct response to the three genetic models. With these 1,187 genes, 11 clusters emerged showing normalization of transcript expression in the PLN KO/Tm180 hearts. In addition, 62 transcripts are highly involved in suppression of the hypertrophic phenotype. Confirmation of the microarray analysis was conducted by quantitative RT-PCR. These results provide insight into genes that alter expression during cardiac remodeling and are active during modulation of the cardiomyopathic phenotype.


Subject(s)
Calcium-Binding Proteins/genetics , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Gene Expression , Animals , Calcium-Binding Proteins/metabolism , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Heart Ventricles/physiopathology , Mice, Knockout , Mice, Transgenic , Microarray Analysis
12.
J Muscle Res Cell Motil ; 34(3-4): 239-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23793376

ABSTRACT

The focus of this review is on the very recent work we have conducted that addresses the molecular, morphological, and physiological significance of cardiac tropomyosin phosphorylation in the heart. We employ transgenic mice to address questions of how cardiomyocytes and the whole heart respond when the tropomyosin phosphorylation site (Ser283) is converted to a non-phosphorylatable amino acid (Ala). We address the phenotype of these mice during normal development and in response to acute cardiac stress (transaortic coarctation). In addition, we also examined how transgenic mice encoding the altered tropomyosin phosphorylation site (Ser283Ala) would respond to chronic cardiac stress through an encoded hypertrophic cardiomyopathy mutation (Glu180Gly). These studies are the first to address the in vivo significance of tropomyosin phosphorylation in the heart. In this review manuscript, we report the recent findings of these investigations.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Myocardium/metabolism , Tropomyosin/metabolism , Animals , Humans , Mice , Mice, Transgenic , Phosphorylation
13.
J Biol Chem ; 288(23): 16235-16246, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23609439

ABSTRACT

α-Tropomyosin (α-TM) has a conserved, charged Asp-137 residue located in the hydrophobic core of its coiled-coil structure, which is unusual in that the residue is found at a position typically occupied by a hydrophobic residue. Asp-137 is thought to destabilize the coiled-coil and so impart structural flexibility to the molecule, which is believed to be crucial for its function in the heart. A previous in vitro study indicated that the conversion of Asp-137 to a more typical canonical Leu alters flexibility of TM and affects its in vitro regulatory functions. However, the physiological importance of the residue Asp-137 and altered TM flexibility is unknown. In this study, we further analyzed structural properties of the α-TM-D137L variant and addressed the physiological importance of TM flexibility in cardiac function in studies with a novel transgenic mouse model expressing α-TM-D137L in the heart. Our NMR spectroscopy data indicated that the presence of D137L introduced long range rearrangements in TM structure. Differential scanning calorimetry measurements demonstrated that α-TM-D137L has higher thermal stability compared with α-TM, which correlated with decreased flexibility. Hearts of transgenic mice expressing α-TM-D137L showed systolic and diastolic dysfunction with decreased myofilament Ca(2+) sensitivity and cardiomyocyte contractility without changes in intracellular Ca(2+) transients or post-translational modifications of major myofilament proteins. We conclude that conversion of the highly conserved Asp-137 to Leu results in loss of flexibility of TM that is important for its regulatory functions in mouse hearts. Thus, our results provide insight into the link between flexibility of TM and its function in ejecting hearts.


Subject(s)
Mutation, Missense , Myocardial Contraction , Myocardium/metabolism , Stroke Volume , Tropomyosin/biosynthesis , Amino Acid Substitution , Animals , Mice , Mice, Transgenic , Myocardium/pathology , Nuclear Magnetic Resonance, Biomolecular , Protein Stability , Rats , Structure-Activity Relationship , Tropomyosin/chemistry , Tropomyosin/genetics
14.
J Physiol ; 591(5): 1217-34, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23207592

ABSTRACT

Abstract Cardiac troponin T (cTnT) has a highly acidic extended N-terminus, the physiological role of which remains poorly understood. To decipher the physiological role of this unique region, we deleted specific regions within the N-terminus of mouse cTnT (McTnT) to create McTnT1-44 and McTnT45-74 proteins. Contractile function and dynamic force-length measurements were made after reconstituting the McTnT deletion proteins into detergent-skinned cardiac papillary fibres harvested from non-transgenic mice that expressed α-tropomyosin (Tm). To further understand how the functional effects of the N-terminus of cTnT are modulated by Tm isoforms, McTnT deletion proteins were reconstituted into detergent-skinned cardiac papillary fibres harvested from transgenic mice that expressed both α- and ß-Tm. McTnT1-44, but not McTnT45-74, attenuated maximal activation of the thin filament. Myofilament Ca(2+) sensitivity, as measured by pCa50 (-log of [Ca(2+)]free required for half-maximal activation), decreased in McTnT1-44 (α-Tm) fibres. The desensitizing effect of McTnT1-44 on pCa50 was ablated in ß-Tm fibres. McTnT45-74 enhanced pCa50 in both α- and ß-Tm fibres, with ß-Tm having a bigger effect. The Hill coefficient of tension development was significantly attenuated by McTnT45-74, suggesting an effect on thin-filament cooperativity. The rate of cross-bridge (XB) detachment and the strained XB-mediated impact on other XBs were augmented by McTnT1-44 in ß-Tm fibres. The magnitude of the length-mediated recruitment of XBs was attenuated by McTnT1-44 in ß-Tm fibres. Our data demonstrate that the 1-44 region of McTnT is essential for maximal activation, whereas the cardiac-specific 45-74 region of McTnT is essential for augmenting cooperativity. Moreover, our data show that α- and ß-Tm isoforms have divergent effects on McTnT deletion mutant's ability to modulate cardiac thin-filament activation and Ca(2+) sensitivity. Our results not only provide the first explicit evidence for the existence of two distinct functional regions within the N-terminus of cTnT, but also offer mechanistic insights into the divergent physiological roles of these regions in mediating cardiac contractile activation.


Subject(s)
Myocardial Contraction , Papillary Muscles/metabolism , Troponin T/metabolism , Animals , Blotting, Western , Calcium/metabolism , Electrophoresis, Polyacrylamide Gel , Kinetics , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Mutation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Isoforms , Recombinant Proteins/metabolism , Tropomyosin/genetics , Tropomyosin/metabolism , Troponin T/chemistry , Troponin T/genetics
15.
J Biol Chem ; 287(53): 44478-89, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23148217

ABSTRACT

Phosphorylation of tropomyosin (Tm) has been shown to vary in mouse models of cardiac hypertrophy. Little is known about the in vivo role of Tm phosphorylation. This study examines the consequences of Tm dephosphorylation in the murine heart. Transgenic (TG) mice were generated with cardiac specific expression of α-Tm with serine 283, the phosphorylation site of Tm, mutated to alanine. Echocardiographic analysis and cardiomyocyte cross-sectional area measurements show that α-Tm S283A TG mice exhibit a hypertrophic phenotype at basal levels. Interestingly, there are no alterations in cardiac function, myofilament calcium (Ca(2+)) sensitivity, cooperativity, or response to ß-adrenergic stimulus. Studies of Ca(2+) handling proteins show significant increases in sarcoplasmic reticulum ATPase (SERCA2a) protein expression and an increase in phospholamban phosphorylation at serine 16, similar to hearts under exercise training. Compared with controls, the decrease in phosphorylation of α-Tm results in greater functional defects in TG animals stressed by transaortic constriction to induce pressure overload-hypertrophy. This is the first study to investigate the in vivo role of Tm dephosphorylation under both normal and cardiac stress conditions, documenting a role for Tm dephosphorylation in the maintenance of a compensated or physiological phenotype. Collectively, these results suggest that modification of the Tm phosphorylation status in the heart, depending upon the cardiac state/condition, may modulate the development of cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Tropomyosin/metabolism , Animals , Calcium/metabolism , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Female , Heart/physiopathology , Humans , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Phosphorylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Tropomyosin/genetics
16.
J Mol Cell Cardiol ; 51(5): 812-20, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21840315

ABSTRACT

We have recently shown that a temporary increase in sarcoplasmic reticulum (SR) cycling via adenovirus-mediated overexpression of sarcoplasmic reticulum ATPase (SERCA2) transiently improves relaxation and delays hypertrophic remodeling in a familial hypertrophic cardiomyopathy (FHC) caused by a mutation in the thin filament protein, tropomyosin (i.e., α-TmE180G or Tm180). In this study, we sought to permanently alter calcium fluxes via phospholamban (PLN) gene deletion in Tm180 mice in order to sustain long-term improvements in cardiac function and adverse cardiac remodeling/hypertrophy. While similar work has been done in FHCs resulting from mutations in thick myofilament proteins, no one has studied these effects in an FHC resulting from a thin filament protein mutation. Tm180 transgenic (TG) mice were crossbred with PLN knockout (KO) mice and four groups were studied in parallel: 1) non-TG (NTG), 2) Tm180, 3) PLNKO/NTG and 4) PLNKO/Tm180. Tm180 mice exhibit increased heart weight/body weight and hypertrophic gene markers compared to NTG mice, but levels in PLNKO/Tm180 mice were similar to NTG. Tm180 mice also displayed altered function as assessed via in situ pressure-volume analysis and echocardiography at 3-6 months and one year; however, altered function in Tm180 mice was rescued back to NTG levels in PLNKO/Tm180 mice. Collagen deposition, as assessed by Picrosirius Red staining, was increased in Tm180 mice but was similar in NTG and in PLNKO/Tm180 mice. Extracellular signal-regulated kinase (ERK1/2) phosphorylation increased in Tm180 mice while levels in PLNKO/Tm180 mice were similar to NTGs. The present study shows that by modulating SR calcium cycling, we were able to rescue many of the deleterious aspects of FHC caused by a mutation in the thin filament protein, Tm.


Subject(s)
Biomarkers/metabolism , Calcium-Binding Proteins/deficiency , Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial , Extracellular Signal-Regulated MAP Kinases/metabolism , Tropomyosin/genetics , Animals , Body Weight , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/therapeutic use , Cardiomyopathy, Hypertrophic, Familial/diagnostic imaging , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cardiomyopathy, Hypertrophic, Familial/therapy , Disease Models, Animal , Echocardiography , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression , Humans , Mice , Mice, Transgenic , Mutation , Myocardial Contraction/genetics , Myocardium/cytology , Myocardium/metabolism , Organ Size , Phosphorylation , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Tropomyosin/metabolism
17.
Am J Physiol Heart Circ Physiol ; 301(4): H1646-55, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21743000

ABSTRACT

The effects of nicotine (NIC) on normal hearts are fairly well established, yet its effects on hearts displaying familial hypertrophic cardiomyopathy have not been tested. We studied both the acute and chronic effects of NIC on a transgenic (TG) mouse model of FHC caused by a mutation in α-tropomyosin (Tm; i.e., α-Tm D175N TG, or Tm175). For acute effects, intravenously injected NIC increased heart rate, left ventricular (LV) pressure, and the maximal rate of LV pressure increase (+dP/dt) in non-TG (NTG) and Tm175 mice; however, Tm175 showed a significantly smaller increase in the maximal rate of LV pressure decrease (-dP/dt) compared with NTGs. Western blots revealed phosphorylation of phospholamban Ser16 and Thr17 residue increased in NTG mice following NIC injection but not in Tm175 mice. In contrast, phosphorylation of troponin I at serine residues 23 and 24 increased equally in both NTG and Tm175. Thus the attenuated increase in relaxation in Tm175 mice following acute NIC appears to result primarily from attenuated phospholamban phosphorylation. Chronic NIC administration (equivalent to smoking 2 packs of cigarettes/day for 4 mo) also increased +dP/dt in NTG and Tm175 mice compared with chronic saline. However, chronic NIC had little effect on heart rate, LV pressure, -dP/dt, LV wall and chamber dimensions, or collagen content for either group of mice.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/drug therapy , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Tropomyosin/genetics , Animals , Blood Pressure/drug effects , Calcium Signaling/drug effects , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cell Separation , Collagen/metabolism , Echocardiography , Female , Fluorescent Dyes , Fura-2 , Heart Rate/drug effects , Hemodynamics/drug effects , Male , Mice , Myocardial Contraction/drug effects , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Ventricular Function, Left/physiology , Ventricular Remodeling/drug effects
18.
J Muscle Res Cell Motil ; 31(5-6): 315-22, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21221740

ABSTRACT

Tropomyosin-kappa (TPM1-κ) is a newly discovered tropomyosin (TM) isoform that is exclusively expressed in the human heart and generated by an alternative splicing of the α-TM gene. We reported that TPM1-κ expression was increased in the hearts of patients with chronic dilated cardiomyopathy (DCM). To increase our understanding of the significance of this shift in isoform population, we generated transgenic (TG) mice expressing TPM1-κ in the cardiac compartment where TPM1-κ replaces 90% of the native TM. We previously showed that there was a significant inhibition of the ability of strongly bound cross-bridges to induce activation of TG myofilaments (Rajan et al., Circulation 121:410-418, 2010). Here, we compared the force-Ca(2+) relations in detergent extracted (skinned) fiber bundles isolated from non-transgenic (NTG) and TG-TPM1-κ hearts at two sarcomere lengths (SLs). Our data demonstrated a significant decrease in the Ca(2+) sensitivity of the myofilaments from TG-TPM1-κ hearts with no change in the maximum developed tension, length-dependent activation, and the ratio of ATPase rate to tension. There was also no difference in the affinity and cooperativity of Ca(2+)-binding to troponin in thin filaments reconstituted with either TPM1-κ or α-TM. We also compared protein phosphorylation in NTG and TG-TPM1-κ myofilaments. There was a decrease in the total phosphorylation of TPM1-κ compared to α-TM, but no significant change in other major sarcomeric proteins. Our results identify a novel mode of myofilament desensitization to Ca(2+) associated with a DCM linked switch in TM isoform population.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathy, Dilated/physiopathology , Myocardial Contraction , Tropomyosin/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Dilated/genetics , Mice , Mice, Transgenic , Phosphorylation , Tropomyosin/genetics
19.
J Mol Cell Cardiol ; 50(1): 137-46, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056571

ABSTRACT

The AE3 Cl(-)/HCO(3)(-) exchanger is abundantly expressed in the sarcolemma of cardiomyocytes, where it mediates Cl(-)-uptake and HCO(3)(-)-extrusion. Inhibition of AE3-mediated Cl(-)/HCO(3)(-) exchange has been suggested to protect against cardiac hypertrophy; however, other studies indicate that AE3 might be necessary for optimal cardiac function. To test these hypotheses we crossed AE3-null mice, which appear phenotypically normal, with a hypertrophic cardiomyopathy mouse model carrying a Glu180Gly mutation in α-tropomyosin (TM180). Loss of AE3 had no effect on hypertrophy; however, survival of TM180/AE3 double mutants was sharply reduced compared with TM180 single mutants. Analysis of cardiac performance revealed impaired cardiac function in TM180 and TM180/AE3 mutants. TM180/AE3 double mutants were more severely affected and exhibited little response to ß-adrenergic stimulation, a likely consequence of their more rapid progression to heart failure. Increased expression of calmodulin-dependent kinase II and protein phosphatase 1 and differences in methylation and localization of protein phosphatase 2A were observed, but were similar in single and double mutants. Phosphorylation of phospholamban on Ser16 was sharply increased in both single and double mutants relative to wild-type hearts under basal conditions, leading to reduced reserve capacity for ß-adrenergic stimulation of phospholamban phosphorylation. Imaging analysis of isolated myocytes revealed reductions in amplitude and decay of Ca(2+) transients in both mutants, with greater reductions in TM180/AE3 mutants, consistent with the greater severity of their heart failure phenotype. Thus, in the TM180 cardiomyopathy model, loss of AE3 had no apparent anti-hypertrophic effect and led to more rapid decompensation and heart failure.


Subject(s)
Antiporters/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cation Transport Proteins , Female , Heart Failure/genetics , Immunoblotting , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Myocytes, Cardiac/pathology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers
20.
J Mol Cell Cardiol ; 50(3): 442-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21047515

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) is a leading cause of sudden cardiac death among young athletes but the functional effects of the myofilament mutations during FHC-associated ischemia and acidosis, due in part to increased extravascular compressive forces and microvascular dysfunction, are not well characterized. We tested the hypothesis that the FHC-linked tropomyosin (Tm) mutation Tm-E180G alters the contractile response to acidosis via increased myofilament Ca(2+) sensitivity. Intact papillary muscles from transgenic (TG) mice expressing Tm-E180G and exposed to acidic conditions (pH 6.9) exhibited a significantly smaller decrease in normalized isometric tension compared to non-transgenic (NTG) preparations. Times to peak tension and to 90% of twitch force relaxation in TG papillary muscles were significantly prolonged. Intact single ventricular TG myocytes demonstrated significantly less inhibition of unloaded shortening during moderate acidosis (pH 7.1) than NTG myocytes. The peak Ca(2+) transients were not different for TG or NTG at any pH tested. The time constant of re-lengthening was slower in TG myocytes, but not the rate of Ca(2+) decline. TG detergent-extracted fibers demonstrated increased Ca(2+) sensitivity of force and maximal tension compared to NTG at both normal and acidic pH (pH 6.5). Tm phosphorylation was not different between TG and NTG muscles at either pH. Our data indicate that acidic pH diminished developed force in hearts of TG mice less than in NTG due to their inherently increased myofilament Ca(2+) sensitivity, thus potentially contributing to altered energy demands and increased propensity for contractile dysfunction.


Subject(s)
Acidosis/genetics , Acidosis/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Cardiomyopathy, Hypertrophic, Familial/metabolism , Mutation , Tropomyosin/genetics , Acidosis/physiopathology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cells, Cultured , Death, Sudden, Cardiac , Isometric Contraction/physiology , Mice , Mice, Transgenic , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Papillary Muscles/metabolism , Papillary Muscles/physiopathology , Phosphorylation , Tropomyosin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...