Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Mol Cell Cardiol ; 52(5): 1145-54, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22281395

RESUMO

Cardiac myosin binding protein C (cMyBP-C) is a myofibrillar protein important for normal myocardial contractility and stability. In mutated form it can cause cardiomyopathy and heart failure. cMyBP-C appears to have separate regions for different functions. Three phosphorylation sites near the N terminus modulate contractility by their effect on both the kinetics of contraction and the binding site of the N-terminus. The C terminal region binds to myosin rods and stabilizes thick filament structure. The aim of the study reported here was to test whether cMyBPC is important in producing the structural and functional changes that result from ischemia/reperfusion. In this study the sequential changes in cMyBP-C, contractility, and thick filament structure following dephosphorylation of cMyBP-C associated with ischemia and reperfusion have been studied in biopsied specimens from chronically instrumented dogs. One and two dimensional electrophoresis, electron microscopy and immunocytochemistry with multiple antibodies generated against different domains in cMyBP-C have been used to follow structural changes in cMyBP-C. Ischemia produced dephosphorylation of cMyBP-C. Subsequent reperfusion released the dephosphorylated cMyBP-C from myofibrils and activated proteolysis of the cytoplasmic cMyBP-C. This in turn leads to increased vulnerability of cMyBP-C to proteolysis and increased degradation of thick filaments. The state of cMyBP-C appears to be closely related to phosphorylation and dephosphorylation of serine 282. In the absence of the stabilizing action of cMyBP-C either as a consequence of genetic mutation or dephosphorylation, premature degradation of thick filaments occurs and is accompanied by persistent contractile dysfunction.


Assuntos
Proteínas de Transporte/fisiologia , Contração Miocárdica , Isquemia Miocárdica/metabolismo , Animais , Proteínas de Transporte/metabolismo , Cães , Feminino , Coração/fisiopatologia , Hemodinâmica , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Miofibrilas/metabolismo , Fosforilação , Transporte Proteico , Proteólise , Reperfusão
2.
J Mol Cell Cardiol ; 52(1): 154-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21971072

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) is a thick filament assembly protein that stabilizes sarcomeric structure and regulates cardiac function; however, the profile of cMyBP-C degradation after myocardial infarction (MI) is unknown. We hypothesized that cMyBP-C is sensitive to proteolysis and is specifically increased in the bloodstream post-MI in rats and humans. Under these circumstances, elevated levels of degraded cMyBP-C could be used as a diagnostic tool to confirm MI. To test this hypothesis, we first established that cMyBP-C dephosphorylation is directly associated with increased degradation of this myofilament protein, leading to its release in vitro. Using neonatal rat ventricular cardiomyocytes in vitro, we were able to correlate the induction of hypoxic stress with increased cMyBP-C dephosphorylation, degradation, and the specific release of N'-fragments. Next, to define the proteolytic pattern of cMyBP-C post-MI, the left anterior descending coronary artery was ligated in adult male rats. Degradation of cMyBP-C was confirmed by a reduction in total cMyBP-C and the presence of degradation products in the infarct tissue. Phosphorylation levels of cMyBP-C were greatly reduced in ischemic areas of the MI heart compared to non-ischemic regions and sham control hearts. Post-MI plasma samples from these rats, as well as humans, were assayed for cMyBP-C and its fragments by sandwich ELISA and immunoprecipitation analyses. Results showed significantly elevated levels of cMyBP-C in the plasma of all post-MI samples. Overall, this study suggests that cMyBP-C is an easily releasable myofilament protein that is dephosphorylated, degraded and released into the circulation post-MI. The presence of elevated levels of cMyBP-C in the blood provides a promising novel biomarker able to accurately rule in MI, thus aiding in the further assessment of ischemic heart disease.


Assuntos
Proteínas de Transporte/metabolismo , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Biomarcadores/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Infarto do Miocárdio/genética , Miocárdio/metabolismo , Miocárdio/patologia , Fosforilação , Proteólise , Ratos , Ratos Sprague-Dawley , Sarcômeros/metabolismo , Fatores de Tempo
3.
Circulation ; 119(11): 1473-83, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19273718

RESUMO

BACKGROUND: Mutations in the MYBPC3 gene, encoding cardiac myosin-binding protein C (cMyBP-C), are a frequent cause of familial hypertrophic cardiomyopathy. In the present study, we investigated whether protein composition and function of the sarcomere are altered in a homogeneous familial hypertrophic cardiomyopathy patient group with frameshift mutations in MYBPC3 (MYBPC3(mut)). METHODS AND RESULTS: Comparisons were made between cardiac samples from MYBPC3 mutant carriers (c.2373dupG, n=7; c.2864_2865delCT, n=4) and nonfailing donors (n=13). Western blots with the use of antibodies directed against cMyBP-C did not reveal truncated cMyBP-C in MYBPC3(mut). Protein expression of cMyBP-C was significantly reduced in MYBPC3(mut) by 33+/-5%. Cardiac MyBP-C phosphorylation in MYBPC3(mut) samples was similar to the values in donor samples, whereas the phosphorylation status of cardiac troponin I was reduced by 84+/-5%, indicating divergent phosphorylation of the 2 main contractile target proteins of the beta-adrenergic pathway. Force measurements in mechanically isolated Triton-permeabilized cardiomyocytes demonstrated a decrease in maximal force per cross-sectional area of the myocytes in MYBPC3(mut) (20.2+/-2.7 kN/m(2)) compared with donor (34.5+/-1.1 kN/m(2)). Moreover, Ca(2+) sensitivity was higher in MYBPC3(mut) (pCa(50)=5.62+/-0.04) than in donor (pCa(50)=5.54+/-0.02), consistent with reduced cardiac troponin I phosphorylation. Treatment with exogenous protein kinase A, to mimic beta-adrenergic stimulation, did not correct reduced maximal force but abolished the initial difference in Ca(2+) sensitivity between MYBPC3(mut) (pCa(50)=5.46+/-0.03) and donor (pCa(50)=5.48+/-0.02). CONCLUSIONS: Frameshift MYBPC3 mutations cause haploinsufficiency, deranged phosphorylation of contractile proteins, and reduced maximal force-generating capacity of cardiomyocytes. The enhanced Ca(2+) sensitivity in MYBPC3(mut) is due to hypophosphorylation of troponin I secondary to mutation-induced dysfunction.


Assuntos
Cardiomiopatia Hipertrófica Familiar/genética , Cardiomiopatia Hipertrófica Familiar/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Adulto , Idoso , Biópsia , Cálcio/metabolismo , Feminino , Mutação da Fase de Leitura , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação/fisiologia , RNA Mensageiro/metabolismo , Sarcômeros/metabolismo
4.
Circ Res ; 101(9): 928-38, 2007 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17823372

RESUMO

The role of cardiac myosin-binding protein C (cMyBP-C) in cardiac contraction is still not fully resolved. Experimental ablation of cMyBP-C by various means resulted in inconsistent changes in Ca2+ sensitivity and increased velocity of force of skinned preparations. To evaluate how these effects are integrated in an intact, living myocyte context, we investigated consequences of cMyBP-C ablation in ventricular myocytes and left atria from cMyBP-C knock-out (KO) mice compared with wild-type (WT). At 6 weeks, KO myocytes exhibited mild hypertrophy that became more pronounced by 30 weeks. Isolated cells from KO exhibited markedly lower diastolic sarcomere length (SL) without change in diastolic Ca2+. The lower SL in KO was partly abolished by the actin-myosin ATPase inhibitors 2,3-butanedione monoxime or blebbistatin, indicating residual actin-myosin interaction in diastole. The relationship between cytosolic Ca2+ and SL showed that KO cells started to contract at lower Ca2+ without reaching a higher maximum, yielding a smaller area of the phase-plane diagram. Both sarcomere shortening and Ca2+ transient were prolonged in KO. Isolated KO left atria exhibited a marked increase in sensitivity to external Ca2+ and, in contrast to WT, continued to develop twitch force at low micromolar Ca2+. Taken together, the main consequence of cMyBP-C ablation was a defect in diastolic relaxation and a smaller dynamic range of cell shortening, both of which likely result from the increased myofilament Ca2+ sensitivity. Our findings indicate that cMyBP-C functions as a restraint on myosin-actin interaction at low Ca2+ and short SL to allow complete relaxation during diastole.


Assuntos
Cardiomiopatia Hipertrófica Familiar/fisiopatologia , Proteínas de Transporte/fisiologia , Diástole/fisiologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Actinas/metabolismo , Animais , Cálcio/metabolismo , Cardiomiopatia Hipertrófica Familiar/patologia , Proteínas de Transporte/genética , Citosol/metabolismo , Átrios do Coração/citologia , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Sarcômeros/fisiologia , Sístole/fisiologia
5.
J Gen Physiol ; 129(5): 419-28, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17470661

RESUMO

Although absence or abnormality of cardiac myosin binding protein C (cMyBP-C) produces serious structural and functional abnormalities of the heart, function of the protein itself is not clearly understood, and the cause of the abnormalities, unidentified. Here we report that a major function of cMyBP-C may be regulating the stability of the myosin-containing contractile filaments through phosphorylation of cMyBP-C. Antibodies were raised against three different regions of cMyBP-C to detect changes in structure within the molecule, and loss of myosin heavy chain was used to monitor degradation of the thick filament. Results from Western blotting and polyacrylamide gel electrophoresis indicate that cMyBP-C can exist in two different forms that produce, respectively, stable and unstable thick filaments. The stable form has well-ordered myosin heads and requires phosphorylation of the cMyBP-C. The unstable form has disordered myosin heads. In tissue with intact cardiac cells, the unstable unphosphorylated cMyBP-C is more easily proteolyzed, causing thick filaments first to release cMyBP-C and/or its proteolytic peptides and then myosin. Filaments deficient in cMyBP-C are fragmented by shear force well tolerated by the stable form. We hypothesize that modulation of filament stability can be coupled at the molecular level with the strength of contraction by the sensitivity of each to the concentration of calcium ions.


Assuntos
Miosinas Cardíacas/metabolismo , Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Miofibrilas/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Transporte/química , Citoesqueleto/ultraestrutura , Técnicas In Vitro , Peso Molecular , Cadeias Pesadas de Miosina/metabolismo , Peptídeo Hidrolases/metabolismo , Fosforilação , Conformação Proteica , Isoformas de Proteínas/metabolismo , Ratos
6.
Circulation ; 111(7): 906-12, 2005 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-15699252

RESUMO

BACKGROUND: Contractile dysfunction develops in the chronically instrumented canine myocardium after bouts of low-flow ischemia and persists after reperfusion. The objective of this study is to identify whether changes in the phosphorylation state of myosin-binding protein C (MyBP-C) are a potential cause of dysfunction. METHODS AND RESULTS: During low-flow ischemia, MyBP-C is dephosphorylated, and the number of actomyosin cross-bridges in the central core of the sarcomere decreases as thick filaments dissemble from the periphery of the myofibril. During reperfusion, MyBP-C remains dephosphorylated, and its degradation is accelerated. CONCLUSIONS: Dephosphorylation of MyBP-C may initiate changes in myofibril thick filament structure that decrease the interaction of myosin heads with actin thin filaments. Limiting the formation of actomyosin cross-bridges may contribute to the contractile dysfunction that is apparent after low-flow ischemia. Breakdown of MyBP-C during reperfusion may prolong myocardial stunning.


Assuntos
Proteínas de Transporte/metabolismo , Contração Miocárdica , Isquemia Miocárdica/fisiopatologia , Miofibrilas/ultraestrutura , Actinas/metabolismo , Actomiosina/metabolismo , Animais , Cães , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/metabolismo , Reperfusão Miocárdica , Miosinas/metabolismo , Fosforilação
7.
J Gen Physiol ; 122(6): 761-74, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14638934

RESUMO

In contrast to skeletal muscle isoforms of myosin binding protein C (MyBP-C), the cardiac isoform has 11 rather than 10 fibronectin or Ig modules (modules are identified as C0 to C10, NH2 to COOH terminus), 3 phosphorylation sites between modules C1 and C2, and 28 additional amino acids rich in proline in C5. Phosphorylation between C1 and C2 increases maximum Ca-activated force (Fmax), alters thick filament structure, and increases the probability of myosin heads on the thick filament binding to actin on the thin filament. Unphosphorylated C1C2 fragment binds to myosin, but phosphorylation inhibits the binding. MyBP-C also binds to actin. Using two types of immunoprecipitation and cosedimentation, we show that fragments of MyBP-C containing C0 bind to actin. In low concentrations C0-containing fragments bind to skinned fibers when the NH2 terminus of endogenous MyBP-C is bound to myosin, but not when MyBP-C is bound to actin. C1C2 fragments bind to skinned fibers when endogenous MyBP-C is bound to actin but not to myosin. Disruption of interactions of endogenous C0 with a high concentration of added C0C2 fragments produces the same effect on contractility as extraction of MyBP-C, namely decrease in Fmax and increase in Ca sensitivity. These results suggest that cardiac contractility can be regulated by shifting the binding of the NH2 terminus of MyBP-C between actin and myosin. This mechanism may have an effect on diastolic filling of the heart.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miosinas/metabolismo , Actinas/genética , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Dados de Sequência Molecular , Ligação Proteica/fisiologia , Ratos
8.
Adv Exp Med Biol ; 538: 31-40; discussion 40-1, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15098652

RESUMO

Both the MyBP-C motif between C1 and C2 and the C5 module are important regions for implementing the effect of MyBP-C on myosin and on contractility but in different ways. C5 may determine the folding of MyBP-C and the manner in which MyBP-C interacts with myosin. In spite of its apparent importance this interaction does not appear to be physiologically regulated. Its alteration by mutation however can have a major effect on contractility. On the other hand, the effect of the motif is regulated by phosphorylation and appears to be an important part of a physiological mechanism(s) for modulating contractility. Thick filaments isolated from cardiac muscle exist in one of three different structures (Levine et al 2001). Different degrees of phosphorylation of MyBP-C can produce transitions among the three structures. The combination of the binding results of Flashman et al (2002) with the data of McClellan et al (2001) suggests that the C5 interaction with C8 is critical in maintaining the normal structure of thick filaments and the normal function of the force generators in the filaments. The cardiac-specific sequence in C5 and its normal interaction with another part of the same or a different MyBP-C may be required for the correct 3 dimensional shape of the three MyBP-C molecules at each locus in the C zone and the normal structure of the thick filament. The normal interactions may be necessary to allow transitions in binding and filament structure that are associated with phosphorylation of the MyBP-C motif.


Assuntos
Proteínas de Transporte/fisiologia , Contração Muscular , Miocárdio/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Transporte/química , Cinética , Camundongos , Músculo Esquelético/metabolismo , Mutação , Miosinas/química , Fosforilação , Ligação Proteica , Dobramento de Proteína , Isoformas de Proteínas , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA