Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Am J Physiol Heart Circ Physiol ; 311(1): H107-17, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27199119

ABSTRACT

We have investigated cardiac myocytes derived from human-induced pluripotent stem cells (iPSC-CMs) from two normal control and two family members expressing a mutant cardiac troponin T (cTnT-R173W) linked to dilated cardiomyopathy (DCM). cTnT is a regulatory protein of the sarcomeric thin filament. The loss of this basic charge, which is strategically located to control tension, has consequences leading to progressive DCM. iPSC-CMs serve as a valuable platform for understanding clinically relevant mutations in sarcomeric proteins; however, there are important questions to be addressed with regard to myocyte adaptation that we model here by plating iPSC-CMs on softer substrates (100 kPa) to create a more physiologic environment during recovery and maturation of iPSC-CMs after thawing from cryopreservation. During the first week of culture of the iPSC-CMs, we have determined structural and functional characteristics as well as actin assembly dynamics. Shortening, actin content, and actin assembly dynamics were depressed in CMs from the severely affected mutant at 1 wk of culture, but by 2 wk differences were less apparent. Sarcomeric troponin and myosin isoform composition were fetal/neonatal. Furthermore, the troponin complex, reconstituted with wild-type cTnT or recombinant cTnT-R173W, depressed the entry of cross-bridges into the force-generating state, which can be reversed by the myosin activator omecamtiv mecarbil. Therapeutic doses of this drug increased both contractility and the content of F-actin in the mutant iPSC-CMs. Collectively, our data suggest the use of a myosin activation reagent to restore function within patient-specific iPSC-CMs may aid in understanding and treating this familial DCM.


Subject(s)
Actins/metabolism , Cardiac Myosins/metabolism , Cardiotonic Agents/pharmacology , Enzyme Activators/pharmacology , Induced Pluripotent Stem Cells/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Point Mutation , Sarcomeres/drug effects , Troponin T/genetics , Urea/analogs & derivatives , Animals , Animals, Newborn , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Genotype , Humans , Induced Pluripotent Stem Cells/enzymology , Myocytes, Cardiac/enzymology , Phenotype , Rats, Sprague-Dawley , Sarcomeres/enzymology , Time Factors , Troponin T/metabolism , Urea/pharmacology
2.
Transplant Proc ; 46(10): 3431-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25498067

ABSTRACT

INTRODUCTION: Physical rehabilitation programs for kidney transplant recipients are not routinely personalized to patients' physical and emotional health, which could result in a potentially limited health impact, shorter-term participation, and an overall low success rate. MATERIALS AND METHODS: We conducted an internal review board-approved randomized prospective study involving a 12-month supervised multidisciplinary rehabilitation program (GH method) initiated after kidney transplantation in obese recipients (body mass index >30). The new method incorporates 3 major components: physical exercise, behavioral interventions, and nutritional guidance. We compared 9 patients who underwent supervised rehabilitation with 8 patients who underwent standard care. Patients were followed up after the start of the intervention, and multiple assessments were performed. RESULTS: The adherence to training and follow-up was 100% in the intervention group, compared with 25% at 12 months in the control group. There was a trend for a higher glomerular filtration rate in the intervention group compared with the control group (55.5 ± 18.6 mL/min/1.73 m(2) vs 38.8 ± 18.9 mL/min/1.73 m(2), P = .06). The quality of life (SF-36) mean score improved more in the intervention group compared with the control group (583 ± 13 vs 436 ± 22, P = .008). There was a significantly higher employment rate in the intervention group, 77.7% at 12 months compared with 12.5% in the control group (P = .02). CONCLUSIONS: Our preliminary results suggest that this comprehensive approach to physical rehabilitation can improve adherence, kidney function, quality of life, and employment rate for obese patients after kidney transplantation.


Subject(s)
Exercise Therapy/methods , Exercise/physiology , Kidney Failure, Chronic/surgery , Kidney Transplantation , Obesity/rehabilitation , Quality of Life , Transplant Recipients , Body Mass Index , Female , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/physiopathology , Male , Middle Aged , Obesity/complications , Prospective Studies
3.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R837-43, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17038443

ABSTRACT

Compared to sham-operated controls, myofilaments from hearts of ovariectomized (OVX) rats demonstrate an increase in Ca2+ sensitivity with no change in maximum tension (Wattanapermpool J and Reiser PJ. Am J Physiol 277: H467-H473, 1999). To test the significance of this modification in intact cells, we compared intracellular Ca2+ transients and shortening of ventricular myocytes isolated from sham and 10-wk OVX rats. There was a decrease in the peak Ca2+ transient with prolonged 50% decay time in OVX cardiac myocytes without changes in the resting intracellular Ca2+ concentration. Percent cell shortening was also depressed, and relaxation was prolonged in cardiac myocytes from OVX rats compared with shams. Ovariectomy induced a sensitization of the myofilaments to Ca2+. Hypercapnic acidosis suppressed the shortening of OVX myocytes to a lesser extent than that detected in shams. Moreover, a larger compensatory increase in %cell shortening was obtained in OVX myocytes during prolonged acidosis. The elevated compensation in cell shortening was related to a higher amount of increase in the amplitude of the Ca2+ transient in OVX myocytes. However, these differences in Ca2+ transients and %cell shortening were no longer evident in the presence of 1 microM cariporide, a specific inhibitor of Na+/H+ exchanger type 1 (NHE1). Our results indicate that deprivation of female sex hormones modulates the intracellular Ca2+ concentration in cardiac myocytes, possibly via an increased NHE1 activity, which may act in concert with Ca2+ hypersensitivity of myofilament activation as a determinant of sex differences in cardiac function.


Subject(s)
Acidosis, Respiratory/physiopathology , Actin Cytoskeleton/physiology , Calcium/metabolism , Gonadal Steroid Hormones/pharmacology , Hypercapnia/physiopathology , Myocytes, Cardiac/physiology , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Calcium Signaling/physiology , Cell Separation , Estrogens/pharmacology , Female , Guanidines/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Muscle Fibers, Skeletal/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Sulfones/pharmacology
4.
Panminerva Med ; 47(3): 133-42, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16462722

ABSTRACT

Molecular signaling that induces cardiac hypertrophy and dilated cardiomyopathy and the transition to decompensation is complex and poorly understood. Extrinsic hemodynamic stresses such as hypertension as well as intrinsic stresses such as genetic defects in sarcomeric proteins and cytoskeletal proteins trigger the process. Both stresses lead to similar outcomes of altered contractility and eventually heart failure. Activation of G-protein coupled receptors initiates cascades of signaling pathways, which promote cardiac hypertrophy by phosphorylation of transcriptional factors and changes in gene expression. Stimulation of these signaling molecules also activates a variety of kinases and phosphatases that induce altered phosphorylation of myofilament proteins. In this review, we focused on these functional effects of small G-protein, Ras and Rho, signaling pathways that reside within the cytoplasm downstream of membrane receptors and upstream of the transcriptional factors. It has been demonstrated that phosphorylation of myofilament proteins alter mechano-energetics of myofilament and contractile function of the heart. Therefore, understanding the role of low molecular weight G-proteins in both cardiac and vascular biology has become particularly important in view of the development of specific inhibitors of effectors of small G-proteins such as p38 MAP kinase and Rho-dependent kinase.


Subject(s)
Heart Failure/etiology , Monomeric GTP-Binding Proteins/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Signal Transduction/physiology , Animals , Calcium/metabolism , Humans , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Phosphorylation , Protein Kinase C/physiology , Protein Serine-Threonine Kinases/physiology , p21-Activated Kinases , p38 Mitogen-Activated Protein Kinases/physiology , rho GTP-Binding Proteins/physiology
5.
J Physiol ; 536(Pt 3): 863-70, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691878

ABSTRACT

1. Acidosis in cardiac muscle is associated with a decrease in developed force. We hypothesized that slow skeletal troponin I (ssTnI), which is expressed in neonatal hearts, is responsible for the observed decreased response to acidic conditions. To test this hypothesis directly, we used adult transgenic (TG) mice that express ssTnI in the heart. Cardiac TnI (cTnI) was completely replaced by ssTnI either with a FLAG epitope introduced into the N-terminus (TG-ssTnI) or without the epitope (TG-ssTnI) in these mice. TG mice that express cTnI were also generated as a control TG line (TG-cTnI). Non-transgenic (NTG) littermates were used as controls. 2. We measured the force-calcium relationship in all four groups at pH 7.0 and pH 6.5 in detergent-extracted fibre bundles prepared from left ventricular papillary muscles. The force-calcium relationship was identical in fibre bundles from NTG and TG-cTnI mouse hearts, therefore NTG mice served as controls for TG-ssTnIand TG-ssTnI mice. Compared to NTG controls, the force generated by fibre bundles from TG mice expressing ssTnI was more sensitive to Ca(2+). The shift in EC(50) (the concentration of Ca(2+) at which half-maximal force is generated) caused by acidic pH was significantly smaller in fibre bundles isolated from TG hearts compared to those from NTG hearts. However, there was no difference in the force-calcium relationship between hearts from the TG-ssTnIand TG-ssTnI groups. 3. We also isolated papillary muscles from the right ventricle of NTG and TG mouse hearts expressing ssTnI and measured isometric force at extracellular pH 7.33 and pH 6.75. At acidic pH, after an initial decline, twitch force recovered to 60 +/- 3 % (n = 7) in NTG papillary muscles, 98 +/- 2 % (n = 5) in muscles from TG-ssTnIand 96 +/- 3 % (n = 7) in muscles from TG-ssTnI hearts. Our results indicate that TnI isoform composition plays a crucial role in the determination of myocardial force sensitivity to acidosis.


Subject(s)
Muscle, Skeletal/metabolism , Myocardium/metabolism , Troponin I/biosynthesis , Acidosis/physiopathology , Animals , Animals, Newborn , Calcium/metabolism , Epitopes/physiology , Hydrogen-Ion Concentration , In Vitro Techniques , Isometric Contraction/physiology , Mice , Mice, Transgenic , Muscle Contraction/physiology , Muscle Fibers, Skeletal/physiology , Muscle Relaxation/physiology , Myocardial Contraction/physiology , Papillary Muscles/physiology , Sarcoplasmic Reticulum/metabolism
6.
J Mol Cell Cardiol ; 33(10): 1815-28, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11603924

ABSTRACT

Tropomyosin, an essential component of the sarcomere, regulates muscle contraction through Ca(2+)-mediated activation. Familial hypertrophic cardiomyopathy (FHC) is caused by mutations in numerous cardiac sarcomeric proteins, including myosin heavy and light chains, actin, troponin T and I, myosin binding protein C, and alpha-tropomyosin. This study developed transgenic mouse lines that encode an FHC mutation in alpha-tropomyosin; this mutation is an amino acid substitution at codon 180 (Glu180Gly) which occurs in a troponin T binding region. Non-transgenic and control mice expressing wild-type alpha-tropomyosin demonstrate no morphological or physiological changes. Expression of exogenous mutant tropomyosin leads to a concomitant decrease in endogenous alpha-tropomyosin without altering the expression of other contractile proteins. Histological analysis shows that initial pathological changes, which include ventricular concentric hypertrophy, fibrosis and atrial enlargement, are detected within 1 month. The disease-associated changes progressively increase and result in death between 4 and 5 months. Physiological analyses of the FHC mice using echocardiography, work-performing heart analyses, and force measurements of cardiac myofibers, demonstrate dramatic functional differences in diastolic performance and increased sensitivity to calcium. This report demonstrates that mutations in alpha-tropomyosin can be severely disruptive of sarcomeric function, which consequently triggers a dramatic hypertrophic response that culminates in lethality.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Mutation , Tropomyosin/genetics , Age Factors , Amino Acids/chemistry , Animals , Calcium/pharmacology , Dose-Response Relationship, Drug , Echocardiography , Mice , Mice, Transgenic , RNA/metabolism , Time Factors
7.
J Mol Cell Cardiol ; 33(7): 1309-20, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11437537

ABSTRACT

Ca2+-activation of cardiac muscle myofilaments is more sensitive to depression by acidic pH than is the case with skeletal myofilaments. We tested the hypothesis that this difference is related to specific regions of the TnI (troponin I) isoforms in these muscles. We exchanged native Tn complex in detergent-extracted fiber bundles from mouse ventricles with Tn containing various combinations of fast (fsTnI) or slow skeletal (ssTnI) complexed with either cardiac TnC (cTnC) or fsTnC, and with cTnC complexed with the following chimeras: (1) fsTnI N-terminal region (fN) plus cTnI inhibitory peptide (cIp) and cTnI C-terminal region (cC); and (2) cTnI N-terminal region (cN)-cIp-fsTnI C-terminal region (fC). We determined the change in half maximal Ca2+(DeltaEC50) for tension activation at pH 7.0 and pH 6.5. Similar DeltaEC50 values were obtained for unextracted controls (5.53+/-0.30 microm), for preparations containing cTnI-cTnC (5.74+/-0.40 microm), and preparations exchanged with cTnI-fsTnC (5.63+/-0.40 microm). However, replacement of cTnI with fsTnI significantly decreased DeltaEC50 to 3.95+/-0.17 microm. Replacement of cTnI with ssTnI also significantly depressed DeltaEC50 to 2.07+/-0.15 microm. Results of studies using the chimeras demonstrated that the C-terminal domains of cTnI and fsTnI are responsible for these differences. This conclusion also fits with data from experiments in which we measured Ca2+-binding to the regulatory site of cTnC in binary complexes containing cTnC with cTnI, fsTnI, or the chimeras. Our results localize a region of TnI important in effects of acidosis on cardiac myofilaments and extend our earlier data indicating that C-terminal regions of cTnI outside the Ip are critical for activation by Ca2+.


Subject(s)
Acid-Base Equilibrium/physiology , Actin Cytoskeleton/metabolism , Myocardium/metabolism , Recombinant Fusion Proteins/metabolism , Troponin C/metabolism , Troponin I/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/drug effects , Amino Acid Motifs/physiology , Animals , Calcium/metabolism , Calcium/pharmacology , Humans , In Vitro Techniques , Mice , Myocardium/chemistry , Myofibrils/chemistry , Myofibrils/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Troponin C/chemistry , Troponin I/chemistry
8.
Circ Res ; 88(10): 1059-65, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11375276

ABSTRACT

Phosphorylation of cardiac myofibrils by cAMP-dependent protein kinase (PKA) can increase the intrinsic rate of myofibrillar relaxation, which may contribute to the shortening of the cardiac twitch during beta-adrenoceptor stimulation. However, it is not known whether the acceleration of myofibrillar relaxation is due to phosphorylation of troponin I (TnI) or of myosin binding protein-C (MyBP-C). To distinguish between these possibilities, we used transgenic mice that overexpress the nonphosphorylatable, slow skeletal isoform of TnI in the myocardium and do not express the normal, phosphorylatable cardiac TNI: The intrinsic rate of relaxation of myofibrils from wild-type and transgenic mice was measured using flash photolysis of diazo-2 to rapidly decrease the [Ca(2+)] within skinned muscles from the mouse ventricles. Incubation with PKA nearly doubled the intrinsic rate of myofibrillar relaxation in muscles from wild-type mice (relaxation half-time fell from approximately 150 to approximately 90 ms at 22 degrees C) but had no effect on the relaxation rate of muscles from the transgenic mice. In parallel studies with intact muscles, we assessed crossbridge kinetics indirectly by determining f(min) (the frequency for minimum dynamic stiffness) during tetanic contractions. Stimulation of beta-adrenoceptors with isoproterenol increased f(min) from 1.9 to 3.1 Hz in muscles from wild-type mice but had no effect on f(min) in muscles from transgenic mice. We conclude that the acceleration of myofibrillar relaxation rate by PKA is due to phosphorylation of TnI, rather than MyBP-C, and that this may be due, at least in part, to faster crossbridge cycle kinetics.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/metabolism , Myocardial Contraction/physiology , Troponin I/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Chelating Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/pharmacology , Diazonium Compounds , Heart Ventricles/drug effects , In Vitro Techniques , Isometric Contraction/drug effects , Isometric Contraction/physiology , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardium/metabolism , Myofibrils/drug effects , Myofibrils/metabolism , Phenoxyacetates , Phosphorylation , Photolysis , Receptors, Adrenergic, beta/metabolism , Stress, Mechanical , Troponin I/genetics
9.
Am J Physiol Cell Physiol ; 280(5): C1114-20, 2001 May.
Article in English | MEDLINE | ID: mdl-11287324

ABSTRACT

Activation of protein kinase C (PKC) in heart muscle signals hypertrophy and may also directly affect contractile function. We tested this idea using a transgenic (TG) mouse model in which conditionally expressed PKCbeta was turned on at 10 wk of age and remained on for either 6 or 10 mo. Compared with controls, TG cardiac myocytes demonstrated an increase in the peak amplitude of the Ca(2+) transient, an increase in the extent and rate of shortening, and an increase in the rate of relengthening at both 6 and 10 mo of age. Phospholamban phosphorylation and Ca(2+)-uptake rates of sarcoplasmic reticulum vesicles were the same in TG and control heart preparations. At 10 mo, TG skinned fiber bundles demonstrated the same sensitivity to Ca(2+) as controls, but maximum tension was depressed and there was increased myofilament protein phosphorylation. Our results differ from studies in which PKCbeta was constitutively overexpressed in the heart and in studies that reported a depression of myocyte contraction with no change in the Ca(2+) transient.


Subject(s)
Calcium Signaling/physiology , Heart/physiology , Isoenzymes/metabolism , Myocardial Contraction/physiology , Protein Kinase C/metabolism , Aging , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Gene Expression Regulation, Enzymologic , Heart/growth & development , Isoenzymes/genetics , Mice , Mice, Transgenic , Myocardium/cytology , Phosphates/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C beta
10.
J Biol Chem ; 276(27): 25456-66, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11320096

ABSTRACT

Ca(2+) binding to cardiac troponin C (cTnC) triggers contraction in heart muscle. In heart failure, myofilaments response to Ca(2+) are often altered and compounds that sensitize the myofilaments to Ca(2+) possess therapeutic value in this syndrome. One of the most potent and selective Ca(2+) sensitizers is the thiadiazinone derivative EMD 57033, which increases myocardial contractile function both in vivo and in vitro and interacts with cTnC in vitro. We have determined the NMR structure of the 1:1 complex between Ca(2+)-saturated C-domain of human cTnC (cCTnC) and EMD 57033. Favorable hydrophobic interactions between the drug and the protein position EMD 57033 in the hydrophobic cleft of the protein. The drug molecule is orientated such that the chiral group of EMD 57033 fits deep in the hydrophobic pocket and makes several key contacts with the protein. This stereospecific interaction explains why the (-)-enantiomer of EMD 57033 is inactive. Titrations of the cCTnC.EMD 57033 complex with two regions of cardiac troponin I (cTnI(34-71) and cTnI(128-147)) reveal that the drug does not share a common binding epitope with cTnI(128-147) but is completely displaced by cTnI(34-71). These results have important implications for elucidating the mechanism of the Ca(2+) sensitizing effect of EMD 57033 in cardiac muscle contraction.


Subject(s)
Calcium/metabolism , Cardiotonic Agents/chemistry , Cardiotonic Agents/metabolism , Quinolines/chemistry , Quinolines/metabolism , Thiadiazines/chemistry , Thiadiazines/metabolism , Troponin C/chemistry , Troponin C/metabolism , Epitope Mapping , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Myocardial Contraction , Protein Binding , Protein Conformation , Stereoisomerism
11.
Am J Pathol ; 158(3): 1079-90, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238056

ABSTRACT

The molecular mechanisms that regulate the cardiac hypertrophic response and the progression from compensated hypertrophy to decompensated heart failure have not been thoroughly defined. Alteration in cardiac extracellular matrix is a distinguishing characteristic of these pathological processes. Integrins, cell surface receptors that mediate cellular adhesion to the extracellular matrix, are signaling molecules that possess mechanotransduction properties. Therefore, we hypothesized that integrins are likely candidates to play an important role in cardiac function. To test this hypothesis, transgenic mice were constructed in which normal integrin function was disrupted by expression of a chimeric molecule encoding the transmembrane and extracellular domains of the Tac subunit of the IL-2 receptor, fused to the cytoplasmic domain of beta(1A) integrin (Tacbeta(1A)). Using the alpha myosin heavy chain promoter to target expression of this chimera to the cardiac myocyte, transgenic mice were generated that had varied levels of transgene expression. Multiple transgenic founders that expressed the transgene at high levels, died perinatally and exhibited replacement fibrosis. Lines that survived showed 1) hypertrophic changes concordant with reduction in endogenous beta(1) integrin levels, or 2) reduced basal contractility and relaxation as well as alterations in components of integrin signaling pathways. These data support an important role for beta(1) integrin in normal cardiac function.


Subject(s)
Cardiomegaly/etiology , Fibrosis/etiology , Heart/physiopathology , Integrin beta1/physiology , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Adhesion , Cells, Cultured , Extracellular Matrix/physiology , Female , Fetal Death , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/physiopathology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Gene Expression , Hemodynamics , Integrin beta1/genetics , Integrin beta1/metabolism , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Protein-Tyrosine Kinases/metabolism , Receptors, Interleukin-2/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
12.
Am J Physiol Heart Circ Physiol ; 280(2): H705-13, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158969

ABSTRACT

The functional consequences of the R92Q mutation in cardiac troponin T (cTnT), linked to familial hypertrophic cardiomyopathy in humans, are not well understood. We have studied steady- and pre-steady-state mechanical activity of detergent-skinned fiber bundles from a transgenic (TG) mouse model in which 67% of the total cTnT in the heart was replaced by the R92Q mutant cTnT. TG fibers were more sensitive to Ca(2+) than nontransgenic (NTG) fibers [negative logarithm of half maximally activating molar Ca(2+) (pCa(50)) = 5.84 +/- 0.01 and 6.12 +/- 0.01 for NTG and TG fibers, respectively]. The shift in pCa(50) caused by increasing the sarcomere length from 1.9 to 2.3 microm was significantly higher for TG than for NTG fibers (DeltapCa(50) = 0.13 +/- 0.01 and 0.29 +/- 0.02 for NTG and TG fibers, respectively). The relationships between rate of ATP consumption and steady-state isometric tension were linear, and the slopes were the same in NTG and TG fibers. Rate of tension redevelopment was more sensitive to Ca(2+) in TG than in NTG fibers (pCa(50) = 5.71 +/- 0.02 and 6.07 +/- 0.02 for NTG and TG fibers, respectively). We concluded that overall cross-bridge cycling kinetics are not altered by the R92Q mutation but that altered troponin-tropomyosin interactions could be responsible for the increase in myofilament Ca(2+) sensitivity in TG myofilaments.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Muscle Fibers, Skeletal/physiology , Troponin T/genetics , Troponin T/metabolism , Acidosis/physiopathology , Adenosine Triphosphate/metabolism , Animals , Calcium/pharmacology , Cross-Linking Reagents/metabolism , Detergents , Genes, myc/genetics , Humans , Mice , Mice, Transgenic , Muscle Contraction/drug effects , Muscle Contraction/physiology , Mutation/physiology , Promoter Regions, Genetic/physiology
13.
Am J Physiol Heart Circ Physiol ; 280(2): H835-43, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158984

ABSTRACT

To determine the in vivo functional significance of troponin I (TnI) protein kinase C (PKC) phosphorylation sites, we created a transgenic mouse expressing mutant TnI, in which PKC phosphorylation sites at serines-43 and -45 were replaced by alanine. When we used high-perfusate calcium as a PKC activator, developed pressures in transgenic (TG) perfused hearts were similar to wild-type (WT) hearts (P = not significant, NS), though there was a 35% and 32% decrease in peak-systolic intracellular calcium (P < 0.01) and diastolic calcium (P < 0.005), respectively. The calcium transient duration was prolonged in the TG mice also (12-27%, ANOVA, P < 0.01). During global ischemia, TG hearts developed ischemic contracture to a greater extent than WT hearts (41 +/- 18 vs. 69 +/- 10 mmHg, perfusate calcium 3.5 mM, P < 0.01). In conclusion, expression of mutant TnI lacking PKC phosphorylation sites results in a marked alteration in the calcium-pressure relationship, and thus susceptibility to ischemic contracture. The reduced intracellular calcium and prolonged calcium transients suggests that a potent feedback mechanism exists between the myofilament and the processes controlling calcium homeostasis.


Subject(s)
Myocardial Ischemia/metabolism , Protein Kinase C/metabolism , Troponin I/genetics , Troponin I/metabolism , Alanine/genetics , Animals , Binding Sites/genetics , Calcium/metabolism , Feedback/physiology , Female , Homeostasis/physiology , In Vitro Techniques , Mice , Mice, Transgenic , Mutagenesis/physiology , Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Myocardium/pathology , Organ Size , Oxygen Consumption/physiology , Phosphorylation , Protein Structure, Tertiary , Serine/genetics , Troponin I/chemistry , Ventricular Pressure/physiology
14.
Am J Physiol Heart Circ Physiol ; 280(3): H1011-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179042

ABSTRACT

Protein kinase C (PKC)-mediated phosphorylation of cardiac troponin I (cTnI) and troponin T (cTnT) has been shown to diminish maximum activation of myofilaments. The functional role of cTnI phosphorylation has been investigated. However, the impact of cTnT phosphorylation on myofilament force is not well studied. We tested the effect of endogenous PKC activation on steady-state tension development and Ca(2+) sensitivity in skinned fiber bundles from transgenic (TG) mouse hearts expressing fast skeletal TnT (fsTnT), which naturally lacks the PKC sites present in cTnT. The 12-O-tetradecanoylphorbol 13-acetate (TPA) treatment induced a 29% (46.1 +/- 2.5 vs. 33.4 +/- 2.6 mN/mm(2)) reduction in maximum tension in the nontransgenic (NTG) preparations (n = 7) and was inhibited with chelerythrine. However, TPA did not induce a change in the maximum tension in the TG preparations (n = 11). TPA induced a small but significant (P < 0.02) increase in Ca(2+) sensitivity (untreated pCa(50) = 5.63 +/- 0.01 vs. treated pCa(50) = 5.72 +/- 0.01) only in TG preparations. In TG preparations, (32)P incorporation was not evident in TnT and was also significantly diminished in cTnI, compared with NTG. Our data indicate that incorporation of fsTnT into the cardiac myofilament lattice blunts PKC-mediated depression of maximum tension. These data also suggest that cTnT may play an important role in amplifying the myofilament depression induced by PKC-mediated phosphorylation of cTnI.


Subject(s)
Actin Cytoskeleton/enzymology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Protein Kinase C/metabolism , Troponin T/genetics , Troponin T/metabolism , Alkaloids , Animals , Benzophenanthridines , Carcinogens/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , Mice , Mice, Transgenic , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal/enzymology , Phenanthridines/pharmacology , Phosphorylation , Tetradecanoylphorbol Acetate/pharmacology
15.
Am J Physiol Heart Circ Physiol ; 279(5): H2414-23, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11045979

ABSTRACT

We used transgenic (TG) mice overexpressing mutant alpha-tropomyosin [alpha-Tm(Asp175Asn)], linked to familial hypertrophic cardiomyopathy (FHC), to test the hypothesis that this mutation impairs cardiac function by altering the sensitivity of myofilaments to Ca(2+). Left ventricular (LV) pressure was measured in anesthetized nontransgenic (NTG) and TG mice. In control conditions, LV relaxation was 6,970 +/- 297 mmHg/s in NTG and 5,624 +/- 392 mmHg/s in TG mice (P < 0.05). During beta-adrenergic stimulation, the rate of relaxation increased to 8,411 +/- 323 mmHg/s in NTG and to 6,080 +/- 413 mmHg/s in TG mice (P < 0.05). We measured the pCa-force relationship (pCa = -log [Ca(2+)]) in skinned fiber bundles from LV papillary muscles of NTG and TG hearts. In control conditions, the Ca(2+) concentration producing 50% maximal force (pCa(50)) was 5.77 +/- 0.02 in NTG and 5.84 +/- 0.01 in TG myofilament bundles (P < 0.05). After protein kinase A-dependent phosphorylation, the pCa(50) was 5.71 +/- 0.01 in NTG and 5.77 +/- 0. 02 in TG myofilament bundles (P < 0.05). Our results indicate that mutant alpha-Tm(Asp175Asn) increases myofilament Ca(2+)-sensitivity, which results in decreased relaxation rate and blunted response to beta-adrenergic stimulation.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Hemodynamics/genetics , Point Mutation/genetics , Tropomyosin/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Ca(2+) Mg(2+)-ATPase/metabolism , Calcium/metabolism , Calcium/pharmacokinetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/pharmacology , Dose-Response Relationship, Drug , Hemodynamics/drug effects , In Vitro Techniques , Mice , Mice, Transgenic , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Papillary Muscles/cytology , Papillary Muscles/metabolism , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Tropomyosin/metabolism , Ventricular Function, Left/genetics
17.
J Physiol ; 526 Pt 3: 541-9, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10922006

ABSTRACT

We compared sarcomere length (SL) dependence of the Ca2+-force relation of detergent-extracted bundles of fibres dissected from the left ventricle of wild-type (WT) and transgenic mouse hearts expressing slow skeletal troponin I (ssTnI-TG). Fibre bundles from the hearts of the ssTnI-TG demonstrated a complete replacement of the cardiac troponin I (cTnI) by ssTnI. Compared to WT controls, ssTnI-TG fibre bundles were more sensitive to Ca2+ at both short SL (1.9 +/- 0.1 micrometer) and long SL (2.3 +/- 0.1 micrometer). However, compared to WT controls, the increase in Ca2+ sensitivity (change in half-maximally activating free Ca2+; DeltaEC50) associated with the increase in SL was significantly blunted in the ssTnI-TG myofilaments. Agents that sensitize the myofilaments to Ca2+ by promoting the actin-myosin reaction (EMD 57033 and CGP-48506) significantly reduced the length-dependent DeltaEC50 for Ca2+ activation, when SL in WT myofilaments was increased from 1.9 to 2.3 micrometer. Exposure of myofilaments to calmidazolium (CDZ), which binds to cTnC and increases its affinity for Ca2+, sensitized force developed by WT myofilaments to Ca2+ at SL 1.9 micrometer and desensitized the WT myofilaments at SL 2.3 micrometer. There were no significant effects of CDZ on ssTnI-TG myofilaments at either SL. Our results indicate that length-dependent Ca2+ activation is modified by specific changes in thin filament proteins and by agents that promote the actin-myosin interaction. Thus, these in vitro results provide a basis for using these models to test the relative significance of the length dependence of activation in situ.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Myocardium/metabolism , Troponin I/biosynthesis , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actins/drug effects , Actins/metabolism , Animals , Azocines/pharmacology , Calcium/pharmacology , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, Transgenic , Muscle Contraction/drug effects , Phosphodiesterase Inhibitors/pharmacology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Quinolines/pharmacology , Sarcomeres/drug effects , Sarcomeres/metabolism , Thiadiazines/pharmacology , Troponin I/genetics
18.
J Biol Chem ; 275(27): 20610-7, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10801883

ABSTRACT

Previously, we utilized (15)N transverse relaxation rates to demonstrate significant mobility in the linker region and conformational exchange in the regulatory domain of Ca(2+)-saturated cardiac troponin C bound to the isolated N-domain of cardiac troponin I (Gaponenko, V., Abusamhadneh, E., Abbott, M. B., Finley, N., Gasmi-Seabrook, G., Solaro, R.J., Rance, M., and Rosevear, P.R. (1999) J. Biol. Chem. 274, 16681-16684). Here we show a large decrease in cardiac troponin C linker flexibility, corresponding to residues 85-93, when bound to intact cardiac troponin I. The addition of 2 m urea to the intact cardiac troponin I-troponin C complex significantly increased linker flexibility. Conformational changes in the regulatory domain of cardiac troponin C were monitored in complexes with troponin I-(1-211), troponin I-(33-211), troponin I-(1-80) and bisphosphorylated troponin I-(1-80). The cardiac specific N terminus, residues 1-32, and the C-domain, residues 81-211, of troponin I are both capable of inducing conformational changes in the troponin C regulatory domain. Phosphorylation of the cardiac specific N terminus reversed its effects on the regulatory domain. These studies provide the first evidence that the cardiac specific N terminus can modulate the function of troponin C by altering the conformational equilibrium of the regulatory domain.


Subject(s)
Myocardium/metabolism , Troponin C/chemistry , Troponin I/metabolism , Animals , Calcium/metabolism , Fluorescence , Macromolecular Substances , Magnetic Resonance Spectroscopy , Naphthalenesulfonates , Phosphorylation , Protein Binding , Protein Conformation , Urea/pharmacology
19.
Ann Biomed Eng ; 28(8): 991-1001, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11144684

ABSTRACT

The syndrome of congestive heart failure (CHF) is an entity of ever increasing clinical significance. CHF is characterized by a steady decrease in cardiac pump function, which is eventually lethal. The mechanisms that underlie the decline in cardiac function are incompletely understood. A central theme in solving the mystery of heart failure is the identification of mechanisms by which the myofilament contractile machine of the myocardium is altered in CHF and how these alterations act in concert with pathways that signal cell growth and death. The cardiac myofilaments are a point of confluence of signals that promote the hypertrophic/failure process. Our hypothesis is that a prevailing hemodynamic stress leads to an increased strain on the myocardium. The increased strain in turn leads to miscues of the normal physiological pathway by which heart cells are signaled to match and adapt the intensity and dynamics of their mechanical activity to prevailing hemodynamic demands. These miscues result in a maladaptation to the stressor and failure of the heart to respond to hemodynamic loads at optimal end diastolic volumes. The result is a vicious cycle exacerbating the failure. Cardiac myofilament activity, the ultimate determinant of cellular dynamics and force, is a central player in the integration and regulation of pathways that signal hypertrophy and failure.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomegaly/physiopathology , Heart Failure/physiopathology , Actin Cytoskeleton/drug effects , Animals , Calcium/metabolism , Calcium/pharmacology , Cardiomegaly/etiology , Disease Progression , Dogs , Heart Failure/complications , Humans , Myocardium/metabolism , Protein Processing, Post-Translational , Signal Transduction , Stress, Mechanical , Tropomyosin/metabolism , Troponin/metabolism , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...