Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Publication year range
1.
Am J Physiol Heart Circ Physiol ; 318(2): H264-H282, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31834834

ABSTRACT

The ß1-adrenergic regulation of cardiac myocyte contraction plays an important role in regulating heart function. Activation of this system leads to an increased heart rate and stronger myocyte contraction. However, chronic stimulation of the ß1-adrenergic signaling system can lead to cardiac hypertrophy and heart failure. To understand the mechanisms of action of ß1-adrenoceptors, a mathematical model of cardiac myocyte contraction that includes the ß1-adrenergic system was developed and studied. The model was able to simulate major experimental protocols for measurements of steady-state force-calcium relationships, cross-bridge release rate and force development rate, force-velocity relationship, and force redevelopment rate. It also reproduced quite well frequency and isoproterenol dependencies for intracellular Ca2+ concentration ([Ca2+]i) transients, total contraction force, and sarcomere shortening. The mathematical model suggested the mechanisms of increased contraction force and myocyte shortening on stimulation of ß1-adrenergic receptors is due to phosphorylation of troponin I and myosin-binding protein C and increased [Ca2+]i transient resulting from activation of the ß1-adrenergic signaling system. The model was used to simulate work-loop contractions and estimate the power during the cardiac cycle as well as the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The developed mathematical model can be used further for simulations of contraction of ventricular myocytes from genetically modified mice and myocytes from mice with chronic cardiac diseases.NEW & NOTEWORTHY A new mathematical model of mouse ventricular myocyte contraction that includes the ß1-adrenergic system was developed. The model simulated major experimental protocols for myocyte contraction and predicted the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The model also allowed for simulations of work-loop contractions and estimation of the power during the cardiac cycle.


Subject(s)
Heart Ventricles , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Receptors, Adrenergic, beta-1/physiology , Algorithms , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cardiotonic Agents/pharmacology , Carrier Proteins/metabolism , Computer Simulation , Heart Rate/physiology , Heart Ventricles/cytology , Heart Ventricles/drug effects , Mice , Models, Theoretical , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Phosphorylation , Potassium Channel Blockers/pharmacology , Receptors, Adrenergic, beta-1/drug effects , Sarcomeres/physiology , Troponin I/metabolism , Troponin I/physiology
2.
J Muscle Res Cell Motil ; 41(1): 71-89, 2020 03.
Article in English | MEDLINE | ID: mdl-31030382

ABSTRACT

The molecular mechanism by which Ca2+ binding and phosphorylation regulate muscle contraction through Troponin is not yet fully understood. Revealing the differences between the relaxed and active structure of cTn, as well as the conformational changes that follow phosphorylation has remained a challenge for structural biologists over the years. Here we review the current understanding of how Ca2+, phosphorylation and disease-causing mutations affect the structure and dynamics of troponin to regulate the thin filament based on electron microscopy, X-ray diffraction, NMR and molecular dynamics methodologies.


Subject(s)
Calcium/metabolism , Muscle Contraction/physiology , Troponin I/physiology , Humans
3.
J Physiol ; 594(3): 669-86, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26635197

ABSTRACT

KEY POINTS: ß-adrenergic stimulation increases cardiac myosin binding protein C (MyBP-C) and troponin I phosphorylation to accelerate pressure development and relaxation in vivo, although their relative contributions remain unknown. Using a novel mouse model lacking protein kinase A-phosphorylatable troponin I (TnI) and MyBP-C, we examined in vivo haemodynamic function before and after infusion of the ß-agonist dobutamine. Mice expressing phospho-ablated MyBP-C displayed cardiac hypertrophy and prevented full acceleration of pressure development and relaxation in response to dobutamine, whereas expression of phosphor-ablated TnI alone had little effect on the acceleration of contractile function in response to dobutamine. Our data demonstrate that MyBP-C phosphorylation is the principal mediator of the contractile response to increased ß-agonist stimulation in vivo. These results help us understand why MyBP-C dephosphorylation in the failing heart contributes to contractile dysfunction and decreased adrenergic reserve in response to acute stress. ß-adrenergic stimulation plays a critical role in accelerating ventricular contraction and speeding relaxation to match cardiac output to changing circulatory demands. Two key myofilaments proteins, troponin I (TnI) and myosin binding protein-C (MyBP-C), are phosphorylated following ß-adrenergic stimulation; however, their relative contributions to the enhancement of in vivo cardiac contractility are unknown. To examine the roles of TnI and MyBP-C phosphorylation in ß-adrenergic-mediated enhancement of cardiac function, transgenic (TG) mice expressing non-phosphorylatable TnI protein kinase A (PKA) residues (i.e. serine to alanine substitution at Ser23/24; TnI(PKA-)) were bred with mice expressing non-phosphorylatable MyBP-C PKA residues (i.e. serine to alanine substitution at Ser273, Ser282 and Ser302; MyBPC(PKA-)) to generate a novel mouse model expressing non-phosphorylatable PKA residues in TnI and MyBP-C (DBL(PKA-)). MyBP-C dephosphorylation produced cardiac hypertrophy and increased wall thickness in MyBPC(PKA-) and DBL(PKA-) mice, and in vivo echocardiography and pressure-volume catheterization studies revealed impaired systolic function and prolonged diastolic relaxation compared to wild-type and TnI(PKA-) mice. Infusion of the ß-agonist dobutamine resulted in accelerated rates of pressure development and relaxation in all mice; however, MyBPC(PKA-) and DBL(PKA-) mice displayed a blunted contractile response compared to wild-type and TnI(PKA-) mice. Furthermore, unanaesthesized MyBPC(PKA-) and DBL(PKA-) mice displayed depressed maximum systolic pressure in response to dobutamine as measured using implantable telemetry devices. Taken together, our data show that MyBP-C phosphorylation is a critical modulator of the in vivo acceleration of pressure development and relaxation as a result of enhanced ß-adrenergic stimulation, and reduced MyBP-C phosphorylation may underlie depressed adrenergic reserve in heart failure.


Subject(s)
Cardiomegaly/physiopathology , Carrier Proteins/physiology , Receptors, Adrenergic, beta/physiology , Troponin I/physiology , Adrenergic beta-1 Receptor Agonists/pharmacology , Animals , Blood Pressure , Cardiomegaly/pathology , Carrier Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/pharmacology , Dobutamine/pharmacology , Female , Heart/physiopathology , Male , Mice, Transgenic , Myocardium/pathology , Myofibrils/metabolism , Phosphorylation , Troponin I/genetics
4.
Arch Biochem Biophys ; 603: 20-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27184165

ABSTRACT

Cardiac tropnoin I (cTnI) plays a critical role in the regulation of diastolic function, and its low expression may result in cardiac diastolic dysfunction, which is the most common form of cardiovascular disorders in older adults. In this study, cTnI expression levels were determined in mice at various ages and cardiac function was measured and compared between young adult mice (3 and 10 months) and older mice (18 months). The data indicated that the cTnI levels reached a peak high in young adult hearts (3 months), but decreased in older hearts (18 months). Furthermore, the older hearts showed a significant diastolic dysfunction observed by P-V loop and echocardiography measurements. To further define the mechanism underlying the cTnI decrease in aging hearts, we tested DNA methylation and histone acetylation modifications of cTnI gene. We found that acetylation of histone near the promoter region of cTnI gene played an important role in regulation of cTnI expression in the heart at different ages. Our study indicates that epigenetic modification caused cTnI expression decrease is one of the possible causes that result in a reduced cTnI level and diastolic dysfunction in the older hearts.


Subject(s)
Aging , Cardiomyopathies/metabolism , Diastole , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Troponin I/physiology , Animals , Chromatin Immunoprecipitation , Echocardiography , Epigenesis, Genetic , Heart/physiology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic
5.
Circ Res ; 112(2): 355-66, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23329791

ABSTRACT

We focus here on the modulation of thin filament activity by cardiac troponin I phosphorylation as an integral and adaptive mechanism in cardiac homeostasis and as a mechanism vulnerable to maladaptive response to stress. We discuss a current concept of cardiac troponin I function in the A-band region of the sarcomere and potential signaling to cardiac troponin I in a network involving the ends of the thin filaments at the Z-disk and the M-band regions. The cardiac sarcomere represents a remarkable set of interacting proteins that functions not only as a molecular machine generating the heartbeat but also as a hub of signaling. We review how phosphorylation signaling to cardiac troponin I is integrated, with parallel signals controlling excitation-contraction coupling, hypertrophy, and metabolism.


Subject(s)
Cardiovascular Physiological Phenomena , Myocytes, Cardiac/metabolism , Troponin I/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Humans , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Phosphorylation/physiology , Troponin I/physiology
6.
Biochem Biophys Res Commun ; 431(1): 47-51, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23291173

ABSTRACT

Stretch-induced enhancement of active force (stretch activation, SA) is observed in striated muscles in general, and most conspicuously in insect flight muscle (IFM). It remains unclear whether a common mechanism underlies the SA of all muscle types, or the SA of IFM relies on its highly specialized features. Recent studies suggest that IFM-specific isoforms of thin filament regulatory proteins (troponin and tropomyosin) are implicated in SA. Among others, IFM-specific troponin-I (troponin-H or TnH), with an unusually long Pro-Ala-rich extension at the C-terminus, has been speculated to transmit the mechanical signal of stretch to the troponin complex. To verify this hypothesis, it was removed by a specific endoproteinase in bumblebee IFM, expecting that it would eliminate SA while leaving intact the capacity for Ca(2+)-activated isometric force. Electrophoretic data showed that the extension was almost completely (97%) removed from IFM fibers after treatment. Unexpectedly, SA force was still conspicuous, and its rate of rise was not affected. Therefore, the results preclude the possibility that the extension is a main part of the mechanism of SA. This leaves open the possibility that SAs of IFM and vertebrate striated muscles, which lack the extension, operate under common basic mechanisms.


Subject(s)
Flight, Animal/physiology , Muscle, Skeletal/physiology , Troponin I/physiology , Amino Acid Sequence , Animals , Drosophila , Molecular Sequence Data , Troponin I/chemistry , Troponin I/genetics
7.
J Physiol ; 590(23): 6047-63, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22966157

ABSTRACT

Troponin I (TnI) and myosin heavy chain (MHC) are two contractile regulatory proteins that undergo major shifts in isoform expression as cardiac myocytes mature from embryonic to adult stages. To date, many studies have investigated individual effects of embryonic vs. cardiac isoforms of either TnI or MHC on cardiac muscle function and contractile dynamics. Thus, we sought to determine whether concomitant expression of the embryonic isoforms of both TnI and MHC had functional effects that were not previously observed. Adult transgenic (TG) mice that express the embryonic isoform of TnI, slow skeletal TnI (ssTnI), were treated with propylthiouracil (PTU) to revert MHC expression from adult (α-MHC) to embryonic (ß-MHC) isoforms. Cardiac muscle fibres from these mice contained ∼80% ß-MHC and ∼34% ssTnI of total MHC or TnI, respectively, allowing us to test the functional effects of ssTnI in the presence of ß-MHC. Detergent-skinned cardiac muscle fibre bundles were used to study how the interplay between MHC and TnI modulate muscle length-mediated effect on crossbridge (XB) recruitment dynamics, Ca(2+)-activated tension, and ATPase activity. One major finding was that the model-predicted XB recruitment rate (b) was enhanced significantly by ssTnI, and this speeding effect of ssTnI on XB recruitment rate was much greater (3.8-fold) when ß-MHC was present. Another major finding was that the previously documented ssTnI-mediated increase in myofilament Ca(2+) sensitivity (pCa(50)) was blunted when ß-MHC was present. ssTnI expression increased pCa(50) by 0.33 in α-MHC fibres, whereas ssTnI increased pCa(50) by only 0.05 in ß-MHC fibres. Our study provides new evidence for significant interplay between MHC and TnI isoforms that is essential for tuning cardiac contractile function. Thus, MHC-TnI interplay may provide a developmentally dependent mechanism to enhance XB recruitment dynamics at a time when Ca(2+)-handling mechanisms are underdeveloped, and to prevent excessive ssTnI-dependent inotropy (increased Ca(2+) sensitivity) in the embryonic myocardium.


Subject(s)
Heart/physiology , Myosin Heavy Chains/physiology , Troponin I/physiology , Animals , Calcium/physiology , Isometric Contraction , Mice , Mice, Transgenic , Myofibrils/physiology , Papillary Muscles/physiology , Propylthiouracil/pharmacology , Protein Isoforms/physiology
8.
J Physiol ; 590(6): 1443-63, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22250211

ABSTRACT

Oxidation can decrease or increase the Ca2+ sensitivity of the contractile apparatus in rodent fast-twitch (type II) skeletal muscle fibres, but the reactions and molecular targets involved are unknown. This study examined whether increased Ca2+ sensitivity is due to S-glutathionylation of particular cysteine residues. Skinned muscle fibres were directly activated in heavily buffered Ca2+ solutions to assess contractile apparatus Ca2+ sensitivity. Rat type II fibres were subjected to S-glutathionylation by successive treatments with 2,2'-dithiodipyridine (DTDP) and glutathione (GSH), and displayed a maximal increase in pCa50 (−log10 [Ca2+] at half-maximal force) of ∼0.24 pCa units, with little or no effect on maximum force or Hill coefficient. Partial similar effect was produced by exposure to oxidized gluthathione (GSSG, 10 mM) for 10 min at pH 7.1, and near-maximal effect by GSSG treatment at pH 8.5. None of these treatments significantly altered Ca2+ sensitivity in rat type I fibres. Western blotting showed that both the DTDP­GSH and GSSG­pH 8.5 treatments caused marked S-glutathionylation of the fast troponin I isoform (TnI(f)) present in type II fibres, but not of troponin C (TnC) or myosin light chain 2. Both the increased Ca2+ sensitivity and glutathionylation of TnI(f) were blocked by N-ethylmaleimide (NEM). S-nitrosoglutathione (GSNO) also increased Ca2+ sensitivity, but only in conditions where it caused S-glutathionylation of TnI(f). In human type II fibres from vastus lateralis muscle, DTDP­GSH treatment also caused similar increased Ca2+ sensitivity and S-glutathionylation of TnI(f). When the slow isoform of TnI in type I fibres of rat was partially substituted (∼30%) with TnI(f), DTDP­GSH treatment caused a significant increase in Ca2+ sensitivity (∼0.08 pCa units). TnIf in type II fibres from toad and chicken muscle lack Cys133 present in mammalian TnIf, and such fibres showed no change in Ca2+ sensitivity with DTDP­GSH nor any S-glutathionylation of TnI(f) (latter examined only in toad). Following 40 min of cycling exercise in human subjects (at ∼60% peak oxygen consumption), TnI(f) in vastus lateralis muscle displayed a marked increase in S-glutathionylation (∼4-fold). These findings show that S-glutathionylation of TnI(f), most probably at Cys133, increases the Ca2+ sensitivity of the contractile apparatus, and that this occurs in exercising humans, with likely beneficial effects on performance.


Subject(s)
Calcium/physiology , Muscle Fibers, Fast-Twitch/physiology , Troponin I/physiology , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/pharmacology , Adult , Animals , Bufo marinus , Chickens , Cysteine/physiology , Disulfides/pharmacology , Exercise/physiology , Female , Glutathione/pharmacology , Glutathione Disulfide/pharmacology , Humans , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/physiology , Rabbits , Rats , Rats, Long-Evans , Swine , Young Adult
9.
Am J Physiol Heart Circ Physiol ; 302(4): H923-33, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22140044

ABSTRACT

In addition to the core structure conserved in all troponin I isoforms, cardiac troponin I (cTnI) has an ∼30 amino acids NH(2)-terminal extension. This peptide segment is a heart-specific regulatory structure containing two Ser residues that are substrates of PKA. Under ß-adrenergic regulation, phosphorylation of cTnI in the NH(2)-terminal extension increases the rate of myocardial relaxation. The NH(2)-terminal extension of cTnI is also removable by restrictive proteolysis to produce functional adaptation to hemodynamic stresses. The molecular mechanism for the NH(2)-terminal modifications to regulate the function of cTnI is not fully understood. In the present study, we tested a hypothesis that the NH(2)-terminal extension functions by modulating the conformation of other regions of cTnI. Monoclonal antibody epitope analysis and protein binding experiments demonstrated that deletion of the NH(2)-terminal segment altered epitopic conformation in the middle, but not COOH-terminal, region of cTnI. PKA phosphorylation produced similar effects. This targeted long-range conformational modulation corresponded to changes in the binding affinities of cTnI for troponin T and for troponin C in a Ca(2+)-dependent manner. The data suggest that the NH(2)-terminal extension of cTnI regulates cardiac muscle function through modulating molecular conformation and function of the core structure of cTnI.


Subject(s)
Heart/physiology , Molecular Conformation , Myocardial Contraction/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Troponin I/chemistry , Troponin I/physiology , Animals , Cattle , Cyclic AMP-Dependent Protein Kinases/metabolism , Epitopes/genetics , Gene Deletion , Humans , Mice , Models, Animal , Muscle Relaxation , Myocardium/metabolism , Peptide Fragments/genetics , Phosphorylation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Troponin I/genetics , Troponin T/metabolism
10.
Exp Cell Res ; 317(2): 188-94, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20828558

ABSTRACT

Assays to quantify myocardial renewal rely on the accurate identification of cardiomyocyte nuclei. We previously ¹4C birth dated human cardiomyocytes based on the nuclear localization of cTroponins T and I. A recent report by Kajstura et al. suggested that cTroponin I is only localized to the nucleus in a senescent subpopulation of cardiomyocytes, implying that ¹4C birth dating of cTroponin T and I positive cell populations underestimates cardiomyocyte renewal in humans. We show here that the isolation of cell nuclei from the heart by flow cytometry with antibodies against cardiac Troponins T and I, as well as pericentriolar material 1 (PCM-1), allows for isolation of close to all cardiomyocyte nuclei, based on ploidy and marker expression. We also present a reassessment of cardiomyocyte ploidy, which has important implications for the analysis of cell turnover, and iododeoxyuridine (IdU) incorporation data. These data provide the foundation for reliable analysis of cardiomyocyte turnover in humans.


Subject(s)
Cell Nucleus/metabolism , Myocytes, Cardiac/diagnostic imaging , Ploidies , Cell Proliferation , Cell Separation , Flow Cytometry , Humans , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Troponin I/physiology , Troponin T/physiology , Ultrasonography
11.
J Card Fail ; 17(5): 359-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21549291

ABSTRACT

BACKGROUND: Autoimmune mechanisms, particularly through generation of autoantibodies, may contribute to the pathophysiology of idiopathic dilated cardiomyopathy (iDCM). The precise role of cellular autoimmune responses to cardiac-specific antigens has not been well described in humans. The purpose of this study was to characterize the cellular autoimmune response to cardiac troponin I (cTnI), specifically, the release of cytokines by peripheral blood mononuclear cells (PBMCs), in subjects with iDCM and healthy control subjects. METHODS AND RESULTS: We performed enzyme-linked immunospot assays on PBMCs isolated from subjects with iDCM and healthy control subjects to examine the ex vivo interferon-gamma (IFN-γ) and interleukin-10 (IL-10) production in response to cTnI exposure. Thirty-five consecutive subjects with iDCM (mean age 53 ± 11 years, 60% male, left ventricular ejection fraction 23 ± 7%) and 26 control subjects (mean age 46 ± 13 years, 46% male) were prospectively enrolled. IFN-γ production in response to cTnI did not differ between the groups (number of secreting cells 26 ± 49 vs 38 ± 53, respectively; P = .1). In contrast, subjects with iDCM showed significantly higher IL-10 responses to cTnI compared with control subjects (number of secreting cells 386 ± 428 vs 152 ± 162, respectively; P < .05). Among iDCM subjects, heightened IL-10 response to cTnI was associated with reduced systemic inflammation and lower prevalence of advanced diastolic dysfunction compared with those with normal IL-10 response to cTnI. CONCLUSIONS: Our preliminary findings suggest that a heightened cellular autoimmune IL-10 response to cTnI is detectable in a subset of patients with iDCM, which may be associated with reduced systemic levels of high-sensitivity C-reactive protein and lower prevalence of advanced diastolic dysfunction.


Subject(s)
Cardiomyopathy, Dilated/immunology , Interferon-gamma/physiology , Interleukin-10/physiology , Leukocytes, Mononuclear/immunology , Troponin I/pharmacology , Adult , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/prevention & control , Case-Control Studies , Cohort Studies , Cross-Sectional Studies , Female , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Pilot Projects , Prospective Studies , Troponin I/physiology
12.
J Mol Cell Cardiol ; 49(3): 402-11, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20580639

ABSTRACT

Several cardiac troponin I (cTnI) mutations are associated with restrictive cardiomyopathy (RCM) in humans. We have created transgenic mice (cTnI(193His) mice) that express the corresponding human RCM R192H mutation. Phenotype of this RCM animal model includes restrictive ventricles, biatrial enlargement and sudden cardiac death, which are similar to those observed in RCM patients carrying the same cTnI mutation. In the present study, we modified the overall cTnI in cardiac muscle by crossing cTnI(193His) mice with transgenic mice expressing an N-terminal truncated cTnI (cTnI-ND) that enhances relaxation. Protein analyses determined that wild type cTnI was replaced by cTnI-ND in the heart of double transgenic mice (Double TG), which express only cTnI-ND and cTnI R193H in cardiac myocytes. The presence of cTnI-ND effectively rescued the lethal phenotype of RCM mice by reducing the mortality rate. Cardiac function was significantly improved in Double TG mice when measured by echocardiography. The hypersensitivity to Ca(2+) and the prolonged relaxation of RCM cTnI(193His) cardiac myocytes were completely reversed by the presence of cTnI-ND in RCM hearts. The results demonstrate that myofibril hypersensitivity to Ca(2+) is a key mechanism that causes impaired relaxation in RCM cTnI mutant hearts and Ca(2+) desensitization by cTnI-ND can correct diastolic dysfunction and rescue the RCM phenotypes, suggesting that Ca(2+) desensitization in myofibrils is a therapeutic option for treatment of diastolic dysfunction without interventions directed at the systemic beta-adrenergic-PKA pathways.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Restrictive/physiopathology , Mutation/genetics , Myocytes, Cardiac/pathology , Troponin I/physiology , Animals , Blotting, Western , Diastole , Echocardiography , Female , Humans , Male , Mice , Mice, Transgenic , Myocardial Contraction , Myocytes, Cardiac/metabolism , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Physiol Genomics ; 42(3): 406-19, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484158

ABSTRACT

Troponin I (TnI) and myosin binding protein-C (MyBP-C) are key regulatory proteins of contractile function in vertebrate muscle. TnI modulates the Ca(2+) activation signal, while MyBP-C regulates cross-bridge cycling kinetics. In vertebrates, each protein is distributed as tissue-specific paralogs in fast skeletal (fs), slow skeletal (ss), and cardiac (c) muscles. The purpose of this study is to characterize how TnI and MyBP-C have changed during the evolution of vertebrate striated muscle and how tissue-specific paralogs have adapted to different physiological conditions. To accomplish this we have completed phylogenetic analyses using the amino acid sequences of all known TnI and MyBP-C isoforms. This includes 99 TnI sequences (fs, ss, and c) from 51 different species and 62 MyBP-C sequences from 26 species, with representatives from each vertebrate group. Results indicate that the role of protein kinase A (PKA) and protein kinase C (PKC) in regulating contractile function has changed during the evolution of vertebrate striated muscle. This is reflected in an increased number of phosphorylatable sites in cTnI and cMyBP-C in endothermic vertebrates and the loss of two PKC sites in fsTnI in a common ancestor of mammals, birds, and reptiles. In addition, we find that His(132), Val(134), and Asn(141) in human ssTnI, previously identified as enabling contractile function during cellular acidosis, are present in all vertebrate cTnI isoforms except those from monotremes, marsupials, and eutherian mammals. This suggests that the replacement of these residues with alternative residues coincides with the evolution of endothermy in the mammalian lineage.


Subject(s)
Carrier Proteins/physiology , Evolution, Molecular , Muscle, Striated/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Troponin I/physiology , Vertebrates/genetics , Amino Acid Sequence , Animals , Birds , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Mammals , Marsupialia , Molecular Sequence Data , Monotremata , Muscle Contraction/genetics , Muscle Contraction/physiology , Phylogeny , Reptiles , Sequence Homology, Amino Acid , Troponin I/genetics , Troponin I/metabolism
14.
Circ Res ; 101(11): 1081-3, 2007 Nov 26.
Article in English | MEDLINE | ID: mdl-17975107

ABSTRACT

Myofilament length-dependent activation is the main cellular mechanism responsible for the Frank-Starling law of the heart. All striated muscle display length-dependent activation properties, but it is most pronounced in cardiac muscle and least in slow skeletal muscle. Cardiac muscle expressing slow skeletal troponin (ssTn)I instead of cardiac troponin (cTn)I displays reduced myofilament length-dependent activation. The inhibitory region of troponin (Tn)I differs by a single residue, proline at position 112 in ssTnI versus threonine at position 144 in cTnI. Here we tested whether this substitution was important for myofilament length-dependent activation; using recombinant techniques, we prepared wild-type cTnI, ssTnI, and 2 mutants: cTnI(Thr>Pro) and ssTnI(Pro>Thr). Purified proteins were complexed with recombinant cardiac TnT/TnC and exchanged into skinned rat cardiac trabeculae. Force-Ca2+ relationships were determined to derive myofilament Ca2+ sensitivity (EC50) at 2 sarcomere lengths: 2.0 and 2.2 microm (n=7). Myofilament length-dependent activation was indexed as deltaEC50, the difference in EC50 between sarcomere lengths of 2.0 and 2.2 microm. Incorporation of ssTnI compared with cTnI into the cardiac sarcomere reduced deltaEC50 from 1.26+/-0.30 to 0.19+/-0.04 micromol/L. A similar reduction also could be observed when Tn contained cTnI(Thr>Pro) (deltaEC50=0.24+/-0.04 micromol/L), whereas the presence of ssTnI(Pro>Thr) increased deltaEC50 to 0.94+/-0.12 micromol/L. These results suggest that Thr144 in cardiac TnI modulates cardiac myofilament length-dependent activation.


Subject(s)
Actin Cytoskeleton/physiology , Myocardium/metabolism , Threonine , Troponin I/physiology , Amino Acid Substitution , Animals , Calcium , Heart/physiology , In Vitro Techniques , Rats , Recombinant Proteins , Sarcomeres , Troponin I/genetics
15.
J Coll Physicians Surg Pak ; 19(9): 544-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19728937

ABSTRACT

OBJECTIVE: To determine the serum levels of troponin-I in identifying left ventricular ejection fraction (LVEF) of 63.5 ng/ml predicted LVEF of 87.5 ng/ml predicted LVEF < 40% with a sensitivity of 86% and specificity of 100%. CONCLUSION: Troponin-I concentration of > 63.5 ng/ml and > 87.5 ng/ml can predict LVEF

Subject(s)
Myocardial Infarction/blood , Stroke Volume , Troponin I/physiology , Ventricular Function, Left , Angioplasty, Balloon, Coronary , Female , Fibrinolytic Agents/therapeutic use , Humans , Immunoenzyme Techniques , Male , Middle Aged , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , ROC Curve , Sensitivity and Specificity , Streptokinase/therapeutic use , Troponin I/blood
16.
Nat Commun ; 10(1): 3295, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337768

ABSTRACT

HCN channels underlie the depolarizing funny current (If) that contributes importantly to cardiac pacemaking. If is upregulated in failing and infarcted hearts, but its implication in disease mechanisms remained unresolved. We generated transgenic mice (HCN4tg/wt) to assess functional consequences of HCN4 overexpression-mediated If increase in cardiomyocytes to levels observed in human heart failure. HCN4tg/wt animals exhibit a dilated cardiomyopathy phenotype with increased cellular arrhythmogenicity but unchanged heart rate and conduction parameters. If augmentation induces a diastolic Na+ influx shifting the Na+/Ca2+ exchanger equilibrium towards 'reverse mode' leading to increased [Ca2+]i. Changed Ca2+ homeostasis results in significantly higher systolic [Ca2+]i transients and stimulates apoptosis. Pharmacological inhibition of If prevents the rise of [Ca2+]i and protects from ventricular remodeling. Here we report that augmented myocardial If alters intracellular Ca2+ homeostasis leading to structural cardiac changes and increased arrhythmogenicity. Inhibition of myocardial If per se may constitute a therapeutic mechanism to prevent cardiomyopathy.


Subject(s)
Calcium/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Muscle Proteins/physiology , Potassium Channels/physiology , Animals , Apoptosis , Cardiac Electrophysiology , Gene Expression Profiling , Heart/physiology , Homeostasis , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Troponin I/genetics , Troponin I/metabolism , Troponin I/physiology
17.
J Mol Biol ; 361(3): 420-35, 2006 Aug 18.
Article in English | MEDLINE | ID: mdl-16857209

ABSTRACT

Activation of striated muscle contraction is a highly cooperative signal transduction process converting calcium binding by troponin C (TnC) into interactions between thin and thick filaments. Once calcium is bound, transduction involves changes in protein interactions along the thin filament. The process is thought to involve three different states of actin-tropomyosin (Tm) resulting from changes in troponin's (Tn) interaction with actin-Tm: a blocked (B) state preventing myosin interaction, a closed (C) state allowing weak myosin interactions and favored by calcium binding to Tn, and an open or M state allowing strong myosin interactions. This was tested by measuring the apparent rate of Tn dissociation from rigor skeletal myofibrils using labeled Tn exchange. The location and rate of exchange of Tn or its subunits were measured by high-resolution fluorescence microscopy and image analysis. Three different rates of Tn exchange were observed that were dependent on calcium concentration and strong cross-bridge binding that strongly support the three-state model. The rate of Tn dissociation in the non-overlap region was 200-fold faster at pCa 4 (C-state region) than at pCa 9 (B-state region). When Tn contained engineered TnC mutants with weakened regulatory TnI interactions, the apparent exchange rate at pCa 4 in the non-overlap region increased proportionately with TnI-TnC regulatory affinity. This suggests that the mechanism of calcium enhancement of the rate of Tn dissociation is by favoring a TnI-TnC interaction over a TnI-actin-Tm interaction. At pCa 9, the rate of Tn dissociation in the overlap region (M-state region) was 100-fold faster than the non-overlap region (B-state region) suggesting that strong cross-bridges increase the rate of Tn dissociation. At pCa 4, the rate of Tn dissociation was twofold faster in the non-overlap region (C-state region) than the overlap region (M-state region) that likely involved a strong cross-bridge influence on TnT's interaction with actin-Tm. At sub-maximal calcium (pCa 6.2-5.8), there was a long-range influence of the strong cross-bridge on Tn to enhance its dissociation rate, tens of nanometers from the strong cross-bridge. These observations suggest that the three different states of actin-Tm are associated with three different states of Tn. They also support a model in which strong cross-bridges shift the regulatory equilibrium from a TnI-actin-Tm interaction to a TnC-TnI interaction that likely enhances calcium binding by TnC.


Subject(s)
Muscle, Skeletal/physiology , Myofibrils/physiology , Troponin C/physiology , Troponin I/physiology , Troponin T/physiology , Actins/physiology , Animals , Calcium/physiology , Chickens , In Vitro Techniques , Models, Biological , Muscle Contraction/physiology , Protein Binding , Rabbits , Signal Transduction , Tropomyosin/physiology
18.
Circ Res ; 97(1): 70-7, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15961720

ABSTRACT

Compared with the adult, neonatal heart muscle is less sensitive to deactivation by acidic pH. We hypothesized that expression of slow skeletal troponin I (ssTnI), the embryonic isoform, in adult heart would help maintain left ventricular (LV) systolic function during respiratory hypercapnia. We assessed LV function by transthoracic 2D-targeted M-mode and pulsed Doppler echocardiography in transgenic (TG) mice in which cardiac TnI was replaced with ssTnI and in nontransgenic (NTG) littermates. Anesthetized mice were ventilated with either 100% oxygen or 35% CO2 balanced with oxygen. Arterial blood pH with 35% CO2 decreased to the same levels in both groups of animals. In the absence of propranolol, the LV fractional shortening was higher in TG compared with NTG mice throughout most of the experimental protocol. LV diastolic function was impaired in TG compared with NTG mice both at 100% oxygen and 35% CO2 because E-to-A wave ratio of mitral flow was significantly lower, and E-wave deceleration time and LV isovolumic relaxation time were longer in TG compared with NTG mice. When compensatory mechanisms that occur through stimulation of beta-adrenergic receptors during hypercapnia were blocked by continuous perfusion with propranolol, we found that NTG mice died within 3 to 4 minutes after switching to 35% CO2, whereas TG mice survived. Our experiments demonstrate the first evidence that specific replacement of cardiac TnI with ssTnI has a protective effect on the LV systolic function during hypercapnic acidosis in situ.


Subject(s)
Hypercapnia/physiopathology , Myocardium/metabolism , Systole , Troponin I/physiology , Ventricular Function, Left , Acidosis/physiopathology , Animals , Blood Gas Analysis , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Diastole , Female , Male , Mice , Mice, Transgenic , Phosphorylation , Propranolol/pharmacology , Troponin I/genetics
19.
J Biomech ; 40(9): 2044-52, 2007.
Article in English | MEDLINE | ID: mdl-17140583

ABSTRACT

Familial hypertrophic cardiomyopathy (FHC) is an inherited disease that is characterized by ventricular hypertrophy, cardiac arrhythmias and increased risk of premature sudden death. FHC is caused by autosomal-dominant mutations in genes for a number of sarcomeric proteins; many mutations in Ca(2+)-regulatory proteins of the cardiac thin filament are associated with increased Ca(2+) sensitivity of myofilament function. Computational simulations were used to investigate the possibility that these mutations could affect the Ca(2+) transient and mechanical response of a myocyte during a single cardiac cycle. We used existing experimental data for specific mutations of cardiac troponin I that exhibit increased Ca(2+) sensitivity in physiological and biophysical assays. The simulated Ca(2+) transients were used as input for a three-dimensional half-sarcomere biomechanical model with filament compliance to predict the resulting force. Mutations with the highest Ca(2+) affinity (lowest K(m)) values, exhibit the largest decrease in peak Ca(2+) assuming a constant influx of Ca(2+) into the cytoplasm; they also prolong Ca(2+) removal but have little effect on diastolic Ca(2+). Biomechanical model results suggest that these cTnI mutants would increase peak force despite the decrease in peak [Ca(2+)](i). There is a corresponding increase in net ATP hydrolysis, with no change in tension cost (ATP hydrolyzed per unit of time-integrated tension). These simulations suggest that myofilament-initiated hypertrophic signaling could be associated with decreased [Ca(2+)](i), increased stress/strain, and/or increased ATP flux.


Subject(s)
Actin Cytoskeleton/metabolism , Adenosine Triphosphatases/metabolism , Calcium/metabolism , Cardiomyopathy, Hypertrophic, Familial/genetics , Computer Simulation , Troponin I/genetics , Actin Cytoskeleton/enzymology , Biomechanical Phenomena , Calcium/physiology , Cardiomyopathy, Hypertrophic, Familial/enzymology , Cardiomyopathy, Hypertrophic, Familial/metabolism , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Humans , Models, Biological , Myocardial Contraction/genetics , Signal Transduction/genetics , Troponin I/physiology
20.
Acta Cardiol ; 62(5): 467-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17982967

ABSTRACT

OBJECTIVE: To identify cTnI rise after percutaneous ISD closure and to determine its prognostic significance. BACKGROUND: Cardiac troponin-I (cTnI) is a very specific and sensitive marker of myocardial injury. A significant increase of cTnI levels after percutaneous interatrial septal defect (ISD) closure has been reported. METHODS: Patients admitted for transcatheter atrial septal defect (ASD) and patent foramen ovale (PFO) closure, were enrolled in this prospective study. Standard protocol was unchanged, except for blood sampling, to which serial cTnI before, 6, and 24 hours after the procedure was added. RESULTS: Seventy-eight patients (female/male 37/41, mean age 55 +/- 13 y, 14 ASD and 64 PFO) were included. In none of the patients cTnI was elevated before the procedure and in no patients a cTnI rise above the WHO-criteria for myocardial infarction (> = 0.4 ng/ml) was seen. However, in 32 patients (41%) minimal cTnI rise (range from 0.04 ng/ml to 0.37 ng/ml) was present within a time frame of 24 hours. The increase in cTnI was not related to peri-procedural complications and to short-term outcome (mean follow-up time of 7 +/- 5 months). In addition, cTnI rise was significantly related with the presence of hyperlipideamia (P = 0.004), device size (P = 0.008), and the absence of balloon sizing (P = 0.017). CONCLUSIONS: Transcatheter closure of ISD seems to be a safe procedure. A minimal but significant increase of cTnI is noted in some patients after the procedure, but it seems not to be related to short-term prognosis. However, cTnI rise is clearly associated with patients' characteristics and peri-procedural variables.


Subject(s)
Cardiac Catheterization , Foramen Ovale, Patent/therapy , Heart Septal Defects, Atrial/therapy , Heart Septum/pathology , Myocardium , Troponin I/physiology , Adult , Aged , Female , Heart Septal Defects, Atrial/physiopathology , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Risk Factors , Troponin I/analysis
SELECTION OF CITATIONS
SEARCH DETAIL