Skip to main content
Top
Gepubliceerd in: Netherlands Heart Journal 10/2014

Open Access 01-10-2014 | ICIN

Troponin I phosphorylation in human myocardium in health and disease

Auteurs: P. J. M. Wijnker, A. M. Murphy, G. J. M. Stienen, J. van der Velden

Gepubliceerd in: Netherlands Heart Journal | Uitgave 10/2014

share
DELEN

Deel dit onderdeel of sectie (kopieer de link)

  • Optie A:
    Klik op de rechtermuisknop op de link en selecteer de optie “linkadres kopiëren”
  • Optie B:
    Deel de link per e-mail
insite
ZOEKEN

Abstract

Cardiac troponin I (cTnI) is well known as a biomarker for the diagnosis of myocardial damage. However, because of its central role in the regulation of contraction and relaxation in heart muscle, cTnI may also be a potential target for the treatment of heart failure. Studies in rodent models of cardiac disease and human heart samples showed altered phosphorylation at various sites on cTnI (i.e. site-specific phosphorylation). This is caused by altered expression and/or activity of kinases and phosphatases during heart failure development. It is not known whether these (transient) alterations in cTnI phosphorylation are beneficial or detrimental. Knowledge of the effects of site-specific cTnI phosphorylation on cardiomyocyte contractility is therefore of utmost importance for the development of new therapeutic strategies in patients with heart failure. In this review we focus on the role of cTnI phosphorylation in the healthy heart upon activation of the beta-adrenergic receptor pathway (as occurs during increased stress and exercise) and as a modulator of the Frank-Starling mechanism. Moreover, we provide an overview of recent studies which aimed to reveal the functional consequences of changes in cTnI phosphorylation in cardiac disease.

Introduction

Cardiac troponin I (cTnI) and its binding partner cardiac troponin T (cTnT) are the biomarkers of choice for the diagnosis of myocardial damage [1, 2]. Upon myocardial injury, cTnI is degraded by calcium-sensitive proteases such as calpain I [3] and cTnI fragments are released in the blood where they can be detected via the sensitive assays that are used in practice worldwide.
cTnI is an essential regulator of sarcomere contraction and relaxation. cTnI is the ‘inhibitor’ within the trimeric troponin complex, which together with cardiac troponin C (cTnC, where calcium binds) and cTnT, controls the position of tropomyosin on the thin actin filament in response to Ca2+ [4] (Fig. 1). In diastole (low intracellular [Ca2+]), cTnI binds actin at multiple sites maintaining tropomyosin at the outer domain of actin, and thereby blocks myosin-binding sites on the thin filament and prevents force development. In systole (high intracellular [Ca2+]), Ca2+ binds to cTnC and induces a conformational change in the troponin complex. This results in the release of cTnI from actin and a shift of tropomyosin closer into the grove of the actin filament, thereby enabling actin-myosin interactions and cardiomyocyte force development [5].
Because of its central role in the regulation of contraction and relaxation of the heart, cTnI may also be a potential target for treatment of heart failure. Alterations are known to occur in the phosphorylation status of cTnI during acute cardiac events and in patients with heart failure [68]. Studies in rodent models of cardiac disease and human heart samples showed altered phosphorylation at various sites on cTnI (i.e. site-specific phosphorylation). This is caused by altered expression and/or activity of kinases and phosphatases during the development of heart failure. It is not known whether these (transient) alterations in cTnI phosphorylation are beneficial or detrimental. Knowledge of the effects of site-specific cTnI phosphorylation on cardiomyocyte contractility is therefore of utmost importance for the development of new therapeutic strategies. In this review we focus on the role of cTnI phosphorylation in the healthy and diseased heart upon activation of the beta-adrenergic receptor pathway (as occurs during increased stress and exercise) and as a modulator of the Frank-Starling mechanism, which reflects the ability of the heart to increase stroke volume with an increase in ventricular filling (end-diastolic volume). Moreover, we provide an overview of recent studies which aimed to reveal the functional consequences of changes in cTnI phosphorylation in cardiac disease.

cTnI-Ser23/24 phosphorylation during beta-adrenergic receptor activation

Interactions within the troponin complex are regulated by kinases and phosphatases which, respectively, phosphorylate and dephosphorylate cTnI at multiple phosphorylation sites and play a central role in tuning cardiomyocyte performance. During stress and exercise, sympathetic activation of the heart increases heart rate and stroke volume to meet the increased demands of the body. This is mediated via stimulation of β1-adrenergic receptors, which leads to activation of a downstream kinase, protein kinase A (PKA)[9]. PKA enhances cardiomyocyte contractility and relaxation by phosphorylation of proteins involved in Ca2+ handling and of myofilament proteins such as cTnI, cardiac myosin-binding protein-C (cMyBP-C), and titin (for reviews, see [9, 10]). PKA-mediated phosphorylation of Ca2+-handling proteins (L-type Ca2+ channels, phospholamban and ryanodine receptor) regulates intracellular Ca2+ fluxes [10], thereby controlling the amount of Ca2+ bound to cTnC. In addition to activated (phosphorylated) Ca2+-handling proteins, PKA-mediated cTnI phosphorylation at Serines 23 and 24 (Ser23/24, human sequence) represents a central mechanism controlling cardiomyocyte force development and relaxation, independent of the intracellular Ca2+ concentration. PKA-mediated phosphorylation at cTnI-Ser23/24 reduces myofilament Ca2+ sensitivity and increases the speed of relaxation of the heart which is needed to maintain cardiac performance at increased heart rates [11, 12]. Although Ser23/24 are the so-called PKA sites, multiple other kinases, PKC [13], PKD [14] and PKG [15], are known to phosphorylate cTnI at Ser23/24. The level of phosphorylation of these two serines will be determined by the balance between the kinase activity and phosphatase activity at the myofilaments, since Ser23/24 are dephosphorylated by protein phosphatases (PP) PP1 [16] and PP2A [17, 18].

cTnI-Ser23/24 phosphorylation during beta-adrenergic receptor activation in disease

In the end-stage failing heart, PKA activity is reduced due to alterations in the β-adrenergic signalling pathway [9]. This results in decreased cTnI-Ser23/24 phosphorylation [1821] and increased myofilament Ca2+ sensitivity [18, 20] in patients with ischaemic and dilated heart failure compared with non-failing donors. High myofilament Ca2+ sensitivity may in part underlie impaired relaxation of the diseased heart. Although cTnI-Ser23/24 bisphosphorylation has been studied extensively, effects of site-specific phosphorylation of only Ser23 or Ser24 on cardiomyocyte contractility in the human heart were unknown. Knowledge of functional consequences of monophosphorylated cTnI is important, since recent studies [21, 22] in human post-mortem control hearts and fresh donor transplant hearts with normal cardiac function revealed that approximately 40 % of cTnI is monophosphorylated. Moreover, differences in the level of monophosphorylated cTnI have been reported between donor and end-stage failing hearts [20, 21]. PKA treatment of skinned porcine cardiac muscle preparations suggested that bisphosphorylation of cTnI is required for the reduction in myofilament Ca2+ sensitivity [23]. However, PKA application is aspecific. We recently studied the consequences of site-specific phosphorylation at Ser23 or Ser24 in human cardiomyocytes by exchange of endogenous cTn with site-specific phosphorylated cTnI [24] (Fig. 1). This sophisticated protein engineering technique enabled us to study the effects of one specific cTnI phosphorylation site. For this purpose, phosphorylation sites (Ser23/24) in human recombinant cTnI were mutated to aspartic acid (D) to mimic phosphorylation or alanine (A), which resembles the non-phosphorylatable state. Various phosphorylated cTn complexes were introduced into human cardiomyocytes by protein exchange (Fig. 1). Using this method, we demonstrated that phosphorylation of both Ser23 and Ser24 of cTnI is required to reduce Ca2+ sensitivity in human cardiomyocytes, as no change in Ca2+ sensitivity was observed upon exchange with cTn complexes containing monophosphorylated cTnI [24]. Additionally, we demonstrated that the maximal reduction in myofilament Ca2+ sensitivity was reached at ~55 % bisphosphorylated cTnI [24]. Defective β-adrenergic signalling in heart failure [9] may result in a higher myofilament Ca2+ sensitivity when cTnI phosphorylation levels upon β-adrenergic stimulation remain below ~55 % cTnI bisphosphorylation. This may lead to an impaired diastolic function of the heart due to increased force development at low Ca2+ concentrations. Comparison of cardiac samples from heart failure patients with different disease severity (ranging from NYHA class I to IV) showed increased Ca2+ sensitivity only in the end-stage (NYHA class IV) of cardiac disease [25], which suggested that detrimental effects of reduced cTnI phosphorylation may only become evident at the end-stage of heart failure. However, a recent study in patients with obstructive hypertrophic cardiomyopathy (HCM) and normal systolic, but impaired diastolic function (NYHA class III) showed increased myofilament Ca2+ sensitivity [26] and lower cTnI phosphorylation [27, 28] in HCM compared with non-failing myocardium. In addition, a recent study showed that the level of cTnI bisphosphorylation in post-mortem hearts with mild hypertrophy was significantly lower (4.1 %) compared with control levels (18.4 %) [21]. Collectively, these studies indicate that cTnI bisphosphorylation and the associated impact on myofilament Ca2+ sensitivity depends on the stage of heart failure (NYHA class) as well as on aetiology.

cTnI-Ser23/24 phosphorylation as a regulator of Frank-Starling mechanism in health and disease

Frank-Starling’s law reflects the ability of the heart to increase stroke volume with an increase in ventricular filling (end-diastolic volume), a property based on length-dependent activation (LDA) of the myofilaments [29, 30]. An increase in sarcomere length within the working range of the heart enhances the maximal force generating capacity and the sensitivity of the myofilaments to Ca2+ (Fig. 2a); i.e. for the same calcium concentration more force is generated by the sarcomeres. The contraction becomes more powerful and the ejection fraction increases. Length-dependent myofilament activation thus represents an important cellular mechanism to adjust cardiac performance in response to increased preload of the heart. The role of cTnI as a regulator of LDA of the myofilaments was demonstrated more than a decade ago via replacement of endogenous cTnI by slow skeletal troponin I (ssTnI) in cardiac muscle. This significantly blunted the increase in myofilament Ca2+ sensitivity upon an increase in sarcomere length [31, 32]. Furthermore, cTnI phosphorylation by PKA was demonstrated to enhance length-dependent changes in the force-Ca2+ relation, since PKA only enhanced LDA in non-transgenic mice, and not in a transgenic murine model where cTnI was replaced by ssTnI which misses the N-terminal region where the PKA sites Ser23/24 are located [32]. Recently, we demonstrated that PKA treatment enhanced the length-dependent increase in myofilament Ca2+ sensitivity in human cardiomyocytes with low levels of phosphorylated cTnI [33] (Fig. 2b: Wt vs Wt + PKA). In addition, we showed that phosphorylation at both Ser23 and Ser24 is needed to enhance the length-dependent increase in myofilament Ca2+ sensitivity [33]. Our data indicate that PKA-mediated cTnI-Ser23/24 phosphorylation is an important mediator of length-dependent activation in human cardiac muscle tissue.
As mentioned above, reduced cTnI-Ser23/24 phosphorylation levels have been reported in end-stage heart failure relative to explanted donor tissue [1821]. Since cTnI-Ser23/24 phosphorylation enhances the length-dependent increase in myofilament Ca2+ sensitivity, reduced phosphorylation levels may reduce LDA in heart failure. This may explain why a reduced length-dependent increase in Ca2+ sensitivity was found in skinned fibres from terminally failing human myocardium [34]. In our study [33] replacement of endogenous cTn with exogenous bisphosphorylated cTnI-Ser23/24 enhanced the length-dependent increase in myofilament Ca2+ sensitivity in human failing cardiomyocytes. This suggests that the blunted length-dependent myofilament activation in end-stage heart failure [34] may be at least partly caused by low bisphosphorylation at cTnI-Ser23/24.

cTnI phosphorylation by protein kinase C in the failing heart

While PKA activity and phosphorylation of cTnI-Ser23/24 are reduced in heart failure, it has been demonstrated that protein kinase C (PKC) isoform expression (α, β1, β2) [35] and activity [35] are increased in heart failure. Higher cTnI phosphorylation has been reported at several PKC sites [36] in failing human myocardium [6]. An increase has been found in cTnI phosphorylation at the well-known PKC sites Ser42/44 and Thr143 in end-stage failing compared with donor myocardium [6]. Although PKC-mediated phosphorylation at Ser42/44 and Thr143 and its effects on muscle contractility have been studied in vitro and in rodent models, [37] effects in humans were unknown. Therefore, we recently studied the effects of site-specific phosphorylation at Ser42/44 and Thr143 in human cardiomyocytes.
Studies in rodents demonstrated that PKC-mediated phosphorylation at Ser42/44 decreases maximal force [38]. The reduction in maximal force generating capacity mediated by PKC might underlie the reduced cardiac performance observed in heart failure. However, treatment of human cardiomyocytes with the catalytic domain of PKC and different PKC isoforms (PKCα, PKCε) did not affect maximal force in non-failing donor and failing hearts [39, 40]. Our recent study in human cardiomyocytes demonstrated that replacement of endogenous cTn with exogenous bisphosphorylated cTnI-Ser42/44 induces a relatively large decrease in myofilament Ca2+ sensitivity (Fig. 3) without affecting maximal force development [41]. Interestingly, Ser42/44 pseudo-phosphorylation largely blunted the PKA-mediated increase in length dependence of myofilament Ca2+ sensitivity (Fig. 2b: 42D/44D + PKA vs WT + PKA) [41]. Since the drop in Ca2+ sensitivity at physiological Ca2+ levels was relatively large compared with phosphorylation of Ser23/24, PKC-mediated cTnI-Ser42/44 phosphorylation may result in a reduced force development in vivo, since the maximal intracellular Ca2+ concentration is ~3.98 μM [42].
Phosphorylation of cTnI-Thr143 has been reported to increase myofilament Ca2+ sensitivity of contraction in rodents [43]. In line with this finding, we found an increase in myofilament Ca2+ sensitivity in human cardiomyocytes exchanged with pseudo-phosphorylated cTnI-Thr143 [33] (Fig. 3). In addition, we studied whether phosphorylation of Thr143 affects LDA, since it has been demonstrated that the threonine residue 143 of the inhibitory region of cTnI is essential for length-dependent alterations in myofilament Ca2+ sensitivity [44]. Although Thr143 is essential for LDA in rodents [44], exchange with phosphorylated cTnI-Thr143 did not alter LDA (in the absence or presence of PKA) in human cardiomyocytes [33] (Fig. 2b).

cTnI as a therapeutic target?

cTnI phosphorylation thus affects cardiac performance and its impact differs between the failing and healthy heart. Therefore, cTnI phosphorylation could be a potentially effective target for therapy. Potentially cTnI-Ser23/24 phosphorylation may be a target for correcting high myofilament Ca2+ sensitivity and blunted length-dependent activation in heart failure. Increased myofilament Ca2+ sensitivity may lead to an impaired diastolic function of the heart due to increased force development at low Ca2+ concentrations (Fig. 3). Also, a blunted length-dependent increase in Ca2+ sensitivity in end-stage heart failure has been found [33, 34], which was restored to control levels after exchange with exogenous phosphorylated Ser23/24 [33]. Length-dependent activation is the cellular basis of the Frank-Starling mechanism, which may be depressed in heart failure [45, 46]. Therefore, increasing cTnI-Ser23/24 phosphorylation in failing cardiomyocytes may be beneficial for cardiac function. This is supported by our work in human HCM samples where we showed that restoring phosphorylation at the PKA targets in vitro normalised myofilament function in sarcomere mutation-negative HCM and in HCM with truncating MYBPC3 mutations [47].
The effect of PKC-mediated phosphorylation of cTnI in human depends on which sites are phosphorylated. Phosphorylation of the PKC site Thr143 did not affect LDA properties of human heart muscle (Fig. 2b), but did increase Ca2+ sensitivity as illustrated in Figs. 2 and 3 (Wt versus 143D, Wt + PKA versus 143D + PKA). The high myofilament Ca2+ sensitivity may aid to maintain cardiac output of the failing heart, however, it may also contribute to diastolic dysfunction (Fig. 3).
cTnI phosphorylation at Ser42/44 reduces Ca2+ sensitivity of the myofilaments and blunts the enhanced length-dependent increase in Ca2+ sensitivity mediated by PKA (Fig. 2b). While the low force development at physiological Ca2+ concentrations may reduce cardiac pump function (Fig. 3), PKC-mediated Ser42/44 phosphorylation may exert beneficial effects. The decrease in force development coincides with a decrease in ATP utilisation by the myofilaments [41], which may benefit the energy status of the failing heart. Moreover, the decrease in myofilament Ca2+ sensitivity may also improve diastolic function, as was suggested previously (Fig. 3) [48].
Studying cTnI phosphorylation in different disease phenotypes and at different disease stages will help us to gain a better understanding of the role of PKC-mediated cTnI phosphorylation in cardiac disease. Additionally, with this increasing knowledge about cTnI phosphorylation in cardiac disease, cTnI phosphorylation patterns at the different PKA- and PKC-phosphorylation sites may become promising biomarkers since cTnI phosphorylation status can be determined from blood samples after acute cardiac insult.

Funding

This work was supported by National Institute of Health (NIH) [grant numbers R01 HL063038, NIH R01 HL76038]; the Netherlands Organisation for Scientific Research (NWO; VIDI grant) and a European Society of Cardiology Research Grant.

Conflict of Interest

None declared.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Onze productaanbevelingen

Netherlands Heart Journal

Het Netherlands Heart Journal wordt uitgegeven in samenwerking met de Nederlandse Vereniging voor Cardiologie en de Nederlandse Hartstichting. Het tijdschrift is Engelstalig en wordt gratis beschikbaa ...

Literatuur
1.
go back to reference Thygesen K, Mair J, Katus H, et al. Study group on biomarkers in cardiology of the ESC working group on acute cardiac care. Recommendations for the use of cardiac troponin measurement in acute cardiac care. Eur Heart J. 2010;31:2197–204.PubMedCrossRef Thygesen K, Mair J, Katus H, et al. Study group on biomarkers in cardiology of the ESC working group on acute cardiac care. Recommendations for the use of cardiac troponin measurement in acute cardiac care. Eur Heart J. 2010;31:2197–204.PubMedCrossRef
2.
go back to reference Newby LK, Jesse RL, Babb JD, et al. ACCF 2012 expert consensus document on practical clinical considerations in the interpretation of troponin elevations: a report of the american college of cardiology foundation task force on clinical expert consensus documents. J Am Coll Cardiol. 2012;60:2427–63.PubMedCrossRef Newby LK, Jesse RL, Babb JD, et al. ACCF 2012 expert consensus document on practical clinical considerations in the interpretation of troponin elevations: a report of the american college of cardiology foundation task force on clinical expert consensus documents. J Am Coll Cardiol. 2012;60:2427–63.PubMedCrossRef
3.
go back to reference Barta J, Tóth A, Edes I, et al. Calpain-1-sensitive myofibrillar proteins of the human myocardium. Mol Cell Biochem. 2005;278:1–8.PubMedCrossRef Barta J, Tóth A, Edes I, et al. Calpain-1-sensitive myofibrillar proteins of the human myocardium. Mol Cell Biochem. 2005;278:1–8.PubMedCrossRef
4.
go back to reference Kobayashi T, Solaro RJ. Calcium, thin filaments, and the integrative biology of cardiac contractility. Annu Rev Physiol. 2005;67:39–67.PubMedCrossRef Kobayashi T, Solaro RJ. Calcium, thin filaments, and the integrative biology of cardiac contractility. Annu Rev Physiol. 2005;67:39–67.PubMedCrossRef
5.
go back to reference Lehman W, Craig R. Tropomyosin and the steric mechanism of muscle regulation. Adv Exp Med Biol. 2008;644:95–109.PubMedCrossRef Lehman W, Craig R. Tropomyosin and the steric mechanism of muscle regulation. Adv Exp Med Biol. 2008;644:95–109.PubMedCrossRef
6.
go back to reference Zhang P, Kirk JA, Ji W, et al. Multiple reaction monitoring to identify site-specific troponin I phosphorylated residues in the failing human heart. Circulation. 2012;126:1828–37.PubMedCrossRefPubMedCentral Zhang P, Kirk JA, Ji W, et al. Multiple reaction monitoring to identify site-specific troponin I phosphorylated residues in the failing human heart. Circulation. 2012;126:1828–37.PubMedCrossRefPubMedCentral
11.
go back to reference Solaro RJ, Moir AJ, Perry SV. Phosphorylation of troponin I and the inotropic effect of adrenaline in the perfused rabbit heart. Nature. 1976;262:615–7.PubMedCrossRef Solaro RJ, Moir AJ, Perry SV. Phosphorylation of troponin I and the inotropic effect of adrenaline in the perfused rabbit heart. Nature. 1976;262:615–7.PubMedCrossRef
12.
go back to reference Takimoto E, Soergel DG, Janssen PM, Stull LB, Kass DA, Murphy AM. Frequency- and afterload-dependent cardiac modulation in vivo by troponin I with constitutively active protein kinase a phosphorylation sites. Circ Res. 2004;94:496–504.PubMedCrossRef Takimoto E, Soergel DG, Janssen PM, Stull LB, Kass DA, Murphy AM. Frequency- and afterload-dependent cardiac modulation in vivo by troponin I with constitutively active protein kinase a phosphorylation sites. Circ Res. 2004;94:496–504.PubMedCrossRef
13.
go back to reference Swiderek K, Jaquet K, Meyer HE, Schachtele C, Hofmann F, Heilmeyer Jr LM. Sites phosphorylated in bovine cardiac troponin T and I. Characterization by 31P-NMR spectroscopy and phosphorylation by protein kinases. Eur J Biochem. 1990;190:575–82.PubMedCrossRef Swiderek K, Jaquet K, Meyer HE, Schachtele C, Hofmann F, Heilmeyer Jr LM. Sites phosphorylated in bovine cardiac troponin T and I. Characterization by 31P-NMR spectroscopy and phosphorylation by protein kinases. Eur J Biochem. 1990;190:575–82.PubMedCrossRef
14.
go back to reference Haworth RS, Cuello F, Herron TJ, et al. Protein kinase D is a novel mediator of cardiac troponin I phosphorylation and regulates myofilament function. Circ Res. 2004;95:1091–9.PubMedCrossRef Haworth RS, Cuello F, Herron TJ, et al. Protein kinase D is a novel mediator of cardiac troponin I phosphorylation and regulates myofilament function. Circ Res. 2004;95:1091–9.PubMedCrossRef
15.
go back to reference Blumenthal DK, Stull JT, Gill GN. Phosphorylation of cardiac troponin by guanosine 3′:5′-monophosphate-dependent protein kinase. J Biol Chem. 1978;253:324–6.PubMed Blumenthal DK, Stull JT, Gill GN. Phosphorylation of cardiac troponin by guanosine 3′:5′-monophosphate-dependent protein kinase. J Biol Chem. 1978;253:324–6.PubMed
16.
go back to reference Jideama NM, Crawford BH, Hussain AK, Raynor RL. Dephosphorylation specificities of protein phosphatase for cardiac troponin I, troponin T, and sites within troponin T. Int J Biol Sci. 2006;2:1–9.PubMedCrossRefPubMedCentral Jideama NM, Crawford BH, Hussain AK, Raynor RL. Dephosphorylation specificities of protein phosphatase for cardiac troponin I, troponin T, and sites within troponin T. Int J Biol Sci. 2006;2:1–9.PubMedCrossRefPubMedCentral
17.
go back to reference Deshmukh PA, Blunt BC, Hofmann PA. Acute modulation of PP2a and troponin I phosphorylation in ventricular myocytes: studies with a novel PP2a peptide inhibitor. Am J Physiol Heart Circ Physiol. 2007;292:H792–799.PubMedCrossRef Deshmukh PA, Blunt BC, Hofmann PA. Acute modulation of PP2a and troponin I phosphorylation in ventricular myocytes: studies with a novel PP2a peptide inhibitor. Am J Physiol Heart Circ Physiol. 2007;292:H792–799.PubMedCrossRef
18.
go back to reference Wijnker PJM, Boknik P, Gergs U, et al. Protein phosphatase 2A affects myofilament contractility in non-failing but not in failing human myocardium. J Muscle Res Cell Motil. 2011;32:221–33.PubMedCrossRefPubMedCentral Wijnker PJM, Boknik P, Gergs U, et al. Protein phosphatase 2A affects myofilament contractility in non-failing but not in failing human myocardium. J Muscle Res Cell Motil. 2011;32:221–33.PubMedCrossRefPubMedCentral
19.
go back to reference Hamdani N, Borbely A, Veenstra SP, et al. More severe cellular phenotype in human idiopathic dilated cardiomyopathy compared to ischemic heart disease. J Muscle Res Cell Motil. 2010;31:289–301.PubMedCrossRefPubMedCentral Hamdani N, Borbely A, Veenstra SP, et al. More severe cellular phenotype in human idiopathic dilated cardiomyopathy compared to ischemic heart disease. J Muscle Res Cell Motil. 2010;31:289–301.PubMedCrossRefPubMedCentral
20.
go back to reference van der Velden J, Papp Z, Zaremba R, et al. Increased Ca2+-sensitivity of the contractile apparatus in end-stage human heart failure results from altered phosphorylation of contractile proteins. Cardiovasc Res. 2003;57:37–47.PubMedCrossRef van der Velden J, Papp Z, Zaremba R, et al. Increased Ca2+-sensitivity of the contractile apparatus in end-stage human heart failure results from altered phosphorylation of contractile proteins. Cardiovasc Res. 2003;57:37–47.PubMedCrossRef
21.
go back to reference Zhang J, Guy MJ, Norman HS, et al. Top-down quantitative proteomics identified phosphorylation of cardiac troponin I as a candidate biomarker for chronic heart failure. J Proteome Res. 2011;10:4054–65.PubMedCrossRefPubMedCentral Zhang J, Guy MJ, Norman HS, et al. Top-down quantitative proteomics identified phosphorylation of cardiac troponin I as a candidate biomarker for chronic heart failure. J Proteome Res. 2011;10:4054–65.PubMedCrossRefPubMedCentral
22.
go back to reference Zabrouskov V, Ge Y, Schwartz J, Walker JW. Unraveling molecular complexity of phosphorylated human cardiac troponin I by top down electron capture dissociation/electron transfer dissociation mass spectrometry. Mol Cell Proteomics. 2008;7:1838–49.PubMedCrossRef Zabrouskov V, Ge Y, Schwartz J, Walker JW. Unraveling molecular complexity of phosphorylated human cardiac troponin I by top down electron capture dissociation/electron transfer dissociation mass spectrometry. Mol Cell Proteomics. 2008;7:1838–49.PubMedCrossRef
23.
go back to reference Zhang R, Zhao J, Potter JD. Phosphorylation of both serine residues in cardiac troponin I is required to decrease the Ca2+ affinity of cardiac troponin C. J Biol Chem. 1995;270:30773–80.PubMedCrossRef Zhang R, Zhao J, Potter JD. Phosphorylation of both serine residues in cardiac troponin I is required to decrease the Ca2+ affinity of cardiac troponin C. J Biol Chem. 1995;270:30773–80.PubMedCrossRef
24.
go back to reference Wijnker PJM, Foster DB, Tsao AL, et al. Impact of site-specific phosphorylation of protein kinase A sites Ser23 and Ser24 of cardiac troponin I in human cardiomyocytes. Am J Physiol Heart Circ Physiol. 2013;304:H260–8.PubMedCrossRefPubMedCentral Wijnker PJM, Foster DB, Tsao AL, et al. Impact of site-specific phosphorylation of protein kinase A sites Ser23 and Ser24 of cardiac troponin I in human cardiomyocytes. Am J Physiol Heart Circ Physiol. 2013;304:H260–8.PubMedCrossRefPubMedCentral
25.
go back to reference van der Velden J, Klein LJ, Zaremba R, et al. Effects of calcium, inorganic phosphate, and pH on isometric force in single skinned cardiomyocytes from donor and failing human hearts. Circulation. 2001;104:1140–6.CrossRef van der Velden J, Klein LJ, Zaremba R, et al. Effects of calcium, inorganic phosphate, and pH on isometric force in single skinned cardiomyocytes from donor and failing human hearts. Circulation. 2001;104:1140–6.CrossRef
26.
go back to reference van Dijk SJ, Paalberends ER, Najafi A, et al. Contractile dysfunction irrespective of the mutant protein in human hypertrophic cardiomyopathy with normal systolic function. Circ Heart Fail. 2012;5:36–46.PubMedCrossRef van Dijk SJ, Paalberends ER, Najafi A, et al. Contractile dysfunction irrespective of the mutant protein in human hypertrophic cardiomyopathy with normal systolic function. Circ Heart Fail. 2012;5:36–46.PubMedCrossRef
27.
go back to reference Messer AE, Gallon CE, McKenna WJ, Dos Remedios CG, Marston SB. The use of phosphate-affinity SDS-PAGE to measure the cardiac troponin I phosphorylation site distribution in human heart muscle. Proteomics Clin Appl. 2009;3:1371–82.PubMed Messer AE, Gallon CE, McKenna WJ, Dos Remedios CG, Marston SB. The use of phosphate-affinity SDS-PAGE to measure the cardiac troponin I phosphorylation site distribution in human heart muscle. Proteomics Clin Appl. 2009;3:1371–82.PubMed
28.
go back to reference van Dijk SJ, Dooijes D, Dos Remedios CG, et al. Cardiac myosin-binding protein C mutations and hypertrophic cardiomyopathy: haploinsufficiency, deranged phosphorylation, and cardiomyocyte dysfunction. Circulation. 2009;119:1473–83.PubMedCrossRef van Dijk SJ, Dooijes D, Dos Remedios CG, et al. Cardiac myosin-binding protein C mutations and hypertrophic cardiomyopathy: haploinsufficiency, deranged phosphorylation, and cardiomyocyte dysfunction. Circulation. 2009;119:1473–83.PubMedCrossRef
29.
30.
go back to reference Fuchs F, Martyn DA. Length-dependent Ca2+ activation in cardiac muscle: some remaining questions. J Muscle Res Cell Motil. 2005;26:199–212.PubMedCrossRef Fuchs F, Martyn DA. Length-dependent Ca2+ activation in cardiac muscle: some remaining questions. J Muscle Res Cell Motil. 2005;26:199–212.PubMedCrossRef
31.
go back to reference Arteaga GM, Palmiter KA, Leiden JM, Solaro RJ. Attenuation of length dependence of calcium activation in myofilaments of transgenic mouse hearts expressing slow skeletal troponin I. J Physiol. 2000;526:541–9.PubMedCrossRefPubMedCentral Arteaga GM, Palmiter KA, Leiden JM, Solaro RJ. Attenuation of length dependence of calcium activation in myofilaments of transgenic mouse hearts expressing slow skeletal troponin I. J Physiol. 2000;526:541–9.PubMedCrossRefPubMedCentral
32.
go back to reference Konhilas JP, Irving TC, Wolska BM, et al. Troponin I in the murine myocardium: influence on length-dependent activation and interfilament spacing. J Physiol. 2003;547:951–61.PubMedCrossRefPubMedCentral Konhilas JP, Irving TC, Wolska BM, et al. Troponin I in the murine myocardium: influence on length-dependent activation and interfilament spacing. J Physiol. 2003;547:951–61.PubMedCrossRefPubMedCentral
33.
go back to reference Wijnker PJM, Sequeira V, Foster DB, et al. Length-dependent activation is modulated by cardiac troponin I bisphosphorylation at Ser23 and Ser24 but not by Thr143 phosphorylation. Am J Physiol Heart Circ Physiol. 2014;306:H1171–1181.PubMedCrossRef Wijnker PJM, Sequeira V, Foster DB, et al. Length-dependent activation is modulated by cardiac troponin I bisphosphorylation at Ser23 and Ser24 but not by Thr143 phosphorylation. Am J Physiol Heart Circ Physiol. 2014;306:H1171–1181.PubMedCrossRef
34.
go back to reference Schwinger RH, Bohm M, Koch A, et al. The failing human heart is unable to use the Frank-Starling mechanism. Circ Res. 1994;74:959–69.PubMedCrossRef Schwinger RH, Bohm M, Koch A, et al. The failing human heart is unable to use the Frank-Starling mechanism. Circ Res. 1994;74:959–69.PubMedCrossRef
35.
go back to reference Bowling N, Walsh RA, Song G, et al. Increased protein kinase C activity and expression of Ca2+-sensitive isoforms in the failing human heart. Circulation. 1999;99:384–91.PubMedCrossRef Bowling N, Walsh RA, Song G, et al. Increased protein kinase C activity and expression of Ca2+-sensitive isoforms in the failing human heart. Circulation. 1999;99:384–91.PubMedCrossRef
36.
go back to reference Noland Jr TA, Raynor RL, Kuo JF. Identification of sites phosphorylated in bovine cardiac troponin I and troponin T by protein kinase C and comparative substrate activity of synthetic peptides containing the phosphorylation sites. J Biol Chem. 1989;264:20778–85.PubMed Noland Jr TA, Raynor RL, Kuo JF. Identification of sites phosphorylated in bovine cardiac troponin I and troponin T by protein kinase C and comparative substrate activity of synthetic peptides containing the phosphorylation sites. J Biol Chem. 1989;264:20778–85.PubMed
37.
go back to reference Sumandea MP, Burkart EM, Kobayashi T, De Tombe PP, Solaro RJ. Molecular and integrated biology of thin filament protein phosphorylation in heart muscle. Ann N Y Acad Sci. 2004;1015:39–52.PubMedCrossRef Sumandea MP, Burkart EM, Kobayashi T, De Tombe PP, Solaro RJ. Molecular and integrated biology of thin filament protein phosphorylation in heart muscle. Ann N Y Acad Sci. 2004;1015:39–52.PubMedCrossRef
38.
go back to reference Burkart EM, Sumandea MP, Kobayashi T, et al. Phosphorylation or glutamic acid substitution at protein kinase C sites on cardiac troponin I differentially depress myofilament tension and shortening velocity. J Biol Chem. 2003;278:11265–72.PubMedCrossRef Burkart EM, Sumandea MP, Kobayashi T, et al. Phosphorylation or glutamic acid substitution at protein kinase C sites on cardiac troponin I differentially depress myofilament tension and shortening velocity. J Biol Chem. 2003;278:11265–72.PubMedCrossRef
39.
go back to reference Kooij V, Boontje N, Zaremba R, et al. Protein kinase C alpha and epsilon phosphorylation of troponin and myosin binding protein C reduce Ca2+ sensitivity in human myocardium. Basic Res Cardiol. 2010;105:289–300.PubMedCrossRefPubMedCentral Kooij V, Boontje N, Zaremba R, et al. Protein kinase C alpha and epsilon phosphorylation of troponin and myosin binding protein C reduce Ca2+ sensitivity in human myocardium. Basic Res Cardiol. 2010;105:289–300.PubMedCrossRefPubMedCentral
40.
go back to reference van der Velden J, Narolska NA, Lamberts RR, et al. Functional effects of protein kinase C-mediated myofilament phosphorylation in human myocardium. Cardiovasc Res. 2006;69:876–87.PubMedCrossRef van der Velden J, Narolska NA, Lamberts RR, et al. Functional effects of protein kinase C-mediated myofilament phosphorylation in human myocardium. Cardiovasc Res. 2006;69:876–87.PubMedCrossRef
41.
go back to reference Wijnker PJM, Sequeira V, Witjas-Paalberends ER, et al. Phosphorylation of protein kinase C sites Ser42/44 decreases Ca2+-sensitivity and blunts enhanced length-dependent activation in response to protein kinase A in human cardiomyocytes. Arch Biochem Biophys. 2014;554:11–21.PubMedCrossRef Wijnker PJM, Sequeira V, Witjas-Paalberends ER, et al. Phosphorylation of protein kinase C sites Ser42/44 decreases Ca2+-sensitivity and blunts enhanced length-dependent activation in response to protein kinase A in human cardiomyocytes. Arch Biochem Biophys. 2014;554:11–21.PubMedCrossRef
42.
go back to reference Fabiato A. Myoplasmic free calcium concentration reached during the twitch of an intact isolated cardiac cell and during calcium-induced release of calcium from the sarcoplasmic reticulum of a skinned cardiac cell from the adult rat or rabbit ventricle. J Gen Physiol. 1981;78:457–97.PubMedCrossRef Fabiato A. Myoplasmic free calcium concentration reached during the twitch of an intact isolated cardiac cell and during calcium-induced release of calcium from the sarcoplasmic reticulum of a skinned cardiac cell from the adult rat or rabbit ventricle. J Gen Physiol. 1981;78:457–97.PubMedCrossRef
43.
go back to reference Wang H, Grant JE, Doede CM, Sadayappan S, Robbins J, Walker JW. PKC-betaII sensitizes cardiac myofilaments to Ca2+ by phosphorylating troponin I on threonine-144. J Mol Cell Cardiol. 2006;41:823–33.PubMedCrossRef Wang H, Grant JE, Doede CM, Sadayappan S, Robbins J, Walker JW. PKC-betaII sensitizes cardiac myofilaments to Ca2+ by phosphorylating troponin I on threonine-144. J Mol Cell Cardiol. 2006;41:823–33.PubMedCrossRef
44.
go back to reference Tachampa K, Wang H, Farman GP, de Tombe PP. Cardiac troponin I threonine 144: role in myofilament length dependent activation. Circ Res. 2007;101:1081–3.PubMedCrossRef Tachampa K, Wang H, Farman GP, de Tombe PP. Cardiac troponin I threonine 144: role in myofilament length dependent activation. Circ Res. 2007;101:1081–3.PubMedCrossRef
45.
go back to reference Kitzman DW, Higginbotham MB, Cobb FR, Sheikh KH, Sullivan MJ. Exercise intolerance in patients with heart failure and preserved left ventricular systolic function: failure of the Frank-Starling mechanism. J Am Coll Cardiol. 1991;17:1065–72.PubMedCrossRef Kitzman DW, Higginbotham MB, Cobb FR, Sheikh KH, Sullivan MJ. Exercise intolerance in patients with heart failure and preserved left ventricular systolic function: failure of the Frank-Starling mechanism. J Am Coll Cardiol. 1991;17:1065–72.PubMedCrossRef
46.
go back to reference Ross Jr J, Braunwald E. Studies on Starling’s law of the heart. IX. The effects of impeding venous return on performance of the normal and failing human left ventricle. Circulation. 1964;30:719–27.PubMedCrossRef Ross Jr J, Braunwald E. Studies on Starling’s law of the heart. IX. The effects of impeding venous return on performance of the normal and failing human left ventricle. Circulation. 1964;30:719–27.PubMedCrossRef
47.
go back to reference Sequeira V, Wijnker PJM, Nijenkamp LL, et al. Perturbed length-dependent activation in human hypertrophic cardiomyopathy with missense sarcomeric gene mutations. Circ Res. 2013;112:1491–505.PubMedCrossRefPubMedCentral Sequeira V, Wijnker PJM, Nijenkamp LL, et al. Perturbed length-dependent activation in human hypertrophic cardiomyopathy with missense sarcomeric gene mutations. Circ Res. 2013;112:1491–505.PubMedCrossRefPubMedCentral
48.
go back to reference Lang SE, Robinson DA, Wu HC, Herron TJ, Wahr PA, Westfall MV. Myofilament incorporation and contractile function after gene transfer of cardiac troponin I Ser43/45Ala. Arch Biochem Biophys. 2013;535:49–55.PubMedCrossRefPubMedCentral Lang SE, Robinson DA, Wu HC, Herron TJ, Wahr PA, Westfall MV. Myofilament incorporation and contractile function after gene transfer of cardiac troponin I Ser43/45Ala. Arch Biochem Biophys. 2013;535:49–55.PubMedCrossRefPubMedCentral
Metagegevens
Titel
Troponin I phosphorylation in human myocardium in health and disease
Auteurs
P. J. M. Wijnker
A. M. Murphy
G. J. M. Stienen
J. van der Velden
Publicatiedatum
01-10-2014
Uitgeverij
Bohn Stafleu van Loghum
Gepubliceerd in
Netherlands Heart Journal / Uitgave 10/2014
Print ISSN: 1568-5888
Elektronisch ISSN: 1876-6250
DOI
https://doi.org/10.1007/s12471-014-0590-4

Andere artikelen Uitgave 10/2014

Netherlands Heart Journal 10/2014 Naar de uitgave