Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Struct Biol ; 214(2): 107856, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35427781

RESUMO

INTRODUCTION: The central C4 and C5 domains (C4C5) of cardiac myosin binding protein C (cMyBPC) contain a flexible interdomain linker and a cardiac-isoform specific loop. However, their importance in the functional regulation of cMyBPC has not been extensively studied. METHODS AND RESULTS: We expressed recombinant C4C5 proteins with deleted linker and loop regions and performed biophysical experiments to determine each of their structural and dynamic roles. We show that the linker and C5 loop regions modulate the secondary structure and thermal stability of C4C5. Furthermore, we provide evidence through extended molecular dynamics simulations and principle component analyses that C4C5 can adopt a completely bent or latched conformation. The simulation trajectory and interaction network analyses reveal that the completely bent conformation of C4C5 exhibits a specific pattern of residue-level interactions. Therefore, we propose a "hinge-and-latch" mechanism where the linker allows a great degree of flexibility and bending, while the loop aids in achieving a completely bent and latched conformation. Although this may be one of many bent positions that C4C5 can adopt, we illustrate for the first time in molecular detail that this type of large scale conformational change can occur in the central domains of cMyBPC. CONCLUSIONS: Our hinge-and-latch mechanism demonstrates that the linker and loop regions participate in dynamic modulation of cMyBPC's motion and global conformation. These structural and dynamic features may contribute to muscle isoform-specific regulation of actomyosin activity, and have potential implications regarding its ability to propagate or retract cMyBPC's regulatory N-terminal domains.


Assuntos
Citoesqueleto de Actina , Simulação de Dinâmica Molecular , Citoesqueleto de Actina/química , Conformação Proteica , Estrutura Secundária de Proteína
2.
J Gen Physiol ; 153(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33688929

RESUMO

Omecamtiv mecarbil (OM), a direct myosin motor activator, is currently being tested as a therapeutic replacement for conventional inotropes in heart failure (HF) patients. It is known that HF patients exhibit dysregulated ß-adrenergic signaling and decreased cardiac myosin-binding protein C (cMyBPC) phosphorylation, a critical modulator of myocardial force generation. However, the functional effects of OM in conditions of altered cMyBPC phosphorylation have not been established. Here, we tested the effects of OM on force generation and cross-bridge (XB) kinetics using murine myocardial preparations isolated from wild-type (WT) hearts and from hearts expressing S273A, S282A, and S302A substitutions (SA) in the M domain, between the C1 and C2 domains of cMyBPC, which cannot be phosphorylated. At submaximal Ca2+ activations, OM-mediated force enhancements were less pronounced in SA than in WT myocardial preparations. Additionally, SA myocardial preparations lacked the dose-dependent increases in force that were observed in WT myocardial preparations. Following OM incubation, the basal differences in the rate of XB detachment (krel) between WT and SA myocardial preparations were abolished, suggesting that OM differentially affects the XB behavior when cMyBPC phosphorylation is reduced. Similarly, in myocardial preparations pretreated with protein kinase A to phosphorylate cMyBPC, incubation with OM significantly slowed krel in both the WT and SA myocardial preparations. Collectively, our data suggest there is a strong interplay between the effects of OM and XB behavior, such that it effectively uncouples the sarcomere from cMyBPC phosphorylation levels. Our findings imply that OM may significantly alter the in vivo cardiac response to ß-adrenergic stimulation.


Assuntos
Contração Miocárdica , Ureia , Animais , Humanos , Camundongos , Miocárdio/metabolismo , Fosforilação , Ureia/análogos & derivados , Ureia/metabolismo
3.
JCI Insight ; 5(17)2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32750038

RESUMO

Decreased cardiac myosin-binding protein C (cMyBPC) expression due to inheritable mutations is thought to contribute to the hypertrophic cardiomyopathy (HCM) phenotype, suggesting that increasing cMyBPC content is of therapeutic benefit. In vitro assays show that cMyBPC N-terminal domains (NTDs) contain structural elements necessary and sufficient to modulate actomyosin interactions, but it is unknown if they can regulate in vivo myocardial function. To test whether NTDs can recapitulate the effects of full-length (FL) cMyBPC in rescuing cardiac function in a cMyBPC-null mouse model of HCM, we assessed the efficacy of AAV9 gene transfer of a cMyBPC NTD that contained domains C0C2 and compared its therapeutic potential with AAV9-FL gene replacement. AAV9 vectors were administered systemically at neonatal day 1, when early-onset disease phenotypes begin to manifest. A comprehensive analysis of in vivo and in vitro function was performed following cMyBPC gene transfer. Our results show that a systemic injection of AAV9-C0C2 significantly improved cardiac function (e.g., 52.24 ± 1.69 ejection fraction in the C0C2-treated group compared with 40.07 ± 1.97 in the control cMyBPC-/- group, P < 0.05) and reduced the histopathologic signs of cardiomyopathy. Furthermore, C0C2 significantly slowed and normalized the accelerated cross-bridge kinetics found in cMyBPC-/- control myocardium, as evidenced by a 32.41% decrease in the rate of cross-bridge detachment (krel). Results indicate that C0C2 can rescue biomechanical defects of cMyBPC deficiency and that the NTD may be a target region for therapeutic myofilament kinetic manipulation.


Assuntos
Cardiomiopatias/terapia , Proteínas de Transporte/genética , Terapia Genética/métodos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Camundongos , Miocárdio/metabolismo , Domínios Proteicos , Volume Sistólico
4.
Expert Opin Drug Discov ; 15(4): 457-469, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32067508

RESUMO

Introduction: Heart failure remains one of the largest clinical challenges in the United States. Researchers have continually searched for more effective heart failure treatments that target the cardiac sarcomere but have found few successes despite numerous expensive cardiovascular clinical trials. Among many reasons, the high failure rate of cardiovascular clinical trials may be partly due to incomplete characterization of a drug candidate's complex interaction with cardiac physiology.Areas covered: In this review, the authors address the issue of preclinical cardiovascular studies of sarcomere-targeting heart failure therapies. The authors consider inherent tradeoffs made between mechanistic transparency and physiological fidelity for several relevant preclinical techniques at the atomic, molecular, heart muscle fiber, whole heart, and whole-organism levels. Thus, the authors suggest a comprehensive, bottom-up approach to preclinical cardiovascular studies that fosters scientific rigor and hypothesis-driven drug discovery.Expert opinion: In the authors' opinion, the implementation of hypothesis-driven drug discovery practices, such as the bottom-up approach to preclinical cardiovascular studies, will be imperative for the successful development of novel heart failure treatments. However, additional changes to clinical definitions of heart failure and current drug discovery culture must accompany the bottom-up approach to maximize the effectiveness of hypothesis-driven drug discovery.


Assuntos
Fármacos Cardiovasculares/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Sarcômeros/metabolismo , Animais , Desenvolvimento de Medicamentos , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos , Insuficiência Cardíaca/fisiopatologia , Humanos
5.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 661-677, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30611859

RESUMO

Mutations in cardiac myosin binding protein C (cMyBPC) are a major cause of hypertrophic cardiomyopathy (HCM). In particular, a single amino acid substitution of tyrosine to serine at residue 237 in humans (residue 235 in mice) has been linked to HCM with strong disease association. Although cMyBPC truncations, deletions and insertions, and frame shift mutations have been studied, relatively little is known about the functional consequences of missense mutations in cMyBPC. In this study, we characterized the functional and structural effects of the HCM-causing Y235S mutation by performing mechanical experiments and molecular dynamics simulations (MDS). cMyBPC null mouse myocardium was virally transfected with wild-type (WT) or Y235S cMyBPC (KOY235S). We found that Y235S cMyBPC was properly expressed and incorporated into the cardiac sarcomere, suggesting that the mechanism of disease of the Y235S mutation is not haploinsufficiency or poison peptides. Mechanical experiments in detergent-skinned myocardium isolated from KOY235S hearts revealed hypercontractile behavior compared to KOWT hearts, evidenced by accelerated cross-bridge kinetics and increased Ca2+ sensitivity of force generation. In addition, MDS revealed that the Y235S mutation causes alterations in important intramolecular interactions, surface conformations, and electrostatic potential of the C1 domain of cMyBPC. Our combined in vitro and in silico data suggest that the Y235S mutation directly disrupts internal and surface properties of the C1 domain of cMyBPC, which potentially alters its ligand-binding interactions. These molecular changes may underlie the mechanism for hypercontractile cross-bridge behavior, which ultimately results in the development of cardiac hypertrophy and in vivo cardiac dysfunction.


Assuntos
Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Mutação de Sentido Incorreto , Contração Miocárdica/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Cardiomiopatia Hipertrófica/metabolismo , Proteínas de Transporte/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Proteínas Mutantes/fisiologia , Mutação de Sentido Incorreto/fisiologia , Miocárdio/metabolismo , Domínios Proteicos/genética , Sarcômeros/genética , Sarcômeros/metabolismo , Serina/genética , Tirosina/genética
6.
Arch Biochem Biophys ; 662: 213-218, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30576628

RESUMO

Current inotropic therapies improve systolic function in heart failure patients but also elicit undesirable side effects such as arrhythmias and increased intracellular Ca2+ transients. In order to maintain myocyte Ca2+ homeostasis, the increased cytosolic Ca2+ needs to be actively transported back to sarcoplasmic reticulum leading to depleted ATP reserves. Thus, an emerging approach is to design sarcomere-based treatments to correct impaired contractility via a direct and allosteric modulation of myosin's intrinsic force-generating behavior -a concept that potentially avoids the "off-target" effects. To achieve this goal, various biophysical approaches are utilized to investigate the mechanistic impact of sarcomeric modulators but information derived from diverse approaches is not fully integrated into therapeutic applications. This is in part due to the lack of information that provides a coherent connecting link between biophysical data to in vivo function. Hence, our ability to clearly discern the drug-mediated impact on whole-heart function is diminished. Reducing this translational barrier can significantly accelerate clinical progress related to sarcomere-based therapies by optimizing drug-dosing and treatment duration protocols based on information obtained from biophysical studies. Therefore, we attempt to link biophysical mechanical measurements obtained in isolated cardiac muscle and in vivo contractile function.


Assuntos
Contração Miocárdica/fisiologia , Miocárdio , Pesquisa Translacional Biomédica , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Humanos , Contração Miocárdica/efeitos dos fármacos , Sarcômeros/fisiologia
7.
Int J Cardiol ; 273: 168-176, 2018 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-30279005

RESUMO

Diminished cardiac contractile function is a characteristic feature of dilated cardiomyopathy (DCM) and many other heart failure (HF) causing etiologies. We tested the hypothesis that targeting the sarcomere to increase cardiac contractility can effectively prevent the DCM phenotype in muscle-LIM protein knockout (MLP-/-) mice. The ablation of cardiac myosin binding protein C (MYBPC3-/-) protected the MLP-/- mice from developing the DCM phenotype. We examined the in vivo cardiac function and morphology of the resultant mouse model lacking both MLP and MYBPC3 (DKO) by echocardiography and pressure-volume catheterization and found a significant reduction in hypertrophy, as evidenced by normalized wall thickness and chamber dimensions, and improved systolic function, as evidenced by enhanced ejection fraction (~26% increase compared MLP-/- mice) and rate of pressure development (DKO 7851.0 ±â€¯504.8 vs. MLP-/- 4496.4 ±â€¯196.8 mmHg/s). To investigate the molecular basis for the improved DKO phenotype we performed mechanical experiments in skinned myocardium isolated from WT and the individual KO mice. Skinned myocardium isolated from DKO mice displayed increased Ca2+ sensitivity of force generation, and significantly accelerated rate of cross-bridge detachment (+63% compared to MLP-/-) and rate of XB recruitment (+58% compared to MLP-/-) at submaximal Ca2+ activations. The in vivo and in vitro functional enhancement of DKO mice demonstrates that enhancing the sarcomeric contractility can be cardioprotective in HF characterized by reduced cardiac output, such as in cases of DCM.


Assuntos
Cardiomiopatia Dilatada/genética , Proteínas de Transporte/genética , Modelos Animais de Doenças , Proteínas com Domínio LIM/genética , Proteínas Musculares/genética , Sarcômeros/genética , Sístole/fisiologia , Animais , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/metabolismo , Proteínas de Transporte/metabolismo , Feminino , Proteínas com Domínio LIM/deficiência , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Proteínas Musculares/deficiência , Miócitos Cardíacos/fisiologia , Sarcômeros/metabolismo
8.
J Am Heart Assoc ; 7(17): e009627, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30371160

RESUMO

Background Recent studies suggest that mavacamten (Myk461), a small myosin-binding molecule, decreases hypercontractility in myocardium expressing hypertrophic cardiomyopathy-causing missense mutations in myosin heavy chain. However, the predominant feature of most mutations in cardiac myosin binding protein-C ( cMyBPC ) that cause hypertrophic cardiomyopathy is reduced total cMyBPC expression, and the impact of Myk461 on cMyBPC -deficient myocardium is currently unknown. Methods and Results We measured the impact of Myk461 on steady-state and dynamic cross-bridge ( XB ) behavior in detergent-skinned mouse wild-type myocardium and myocardium lacking cMyBPC (knockout (KO)). KO myocardium exhibited hypercontractile XB behavior as indicated by significant accelerations in rates of XB detachment (krel) and recruitment (kdf) at submaximal Ca2+ activations. Incubation of KO and wild-type myocardium with Myk461 resulted in a dose-dependent force depression, and this impact was more pronounced at low Ca2+ activations. Interestingly, Myk461-induced force depressions were less pronounced in KO myocardium, especially at low Ca2+ activations, which may be because of increased acto-myosin XB formation and potential disruption of super-relaxed XB s in KO myocardium. Additionally, Myk461 slowed krel in KO myocardium but not in wild-type myocardium, indicating increased XB " on" time. Furthermore, the greater degree of Myk461-induced slowing in kdf and reduction in XB recruitment magnitude in KO myocardium normalized the XB behavior back to wild-type levels. Conclusions This is the first study to demonstrate that Myk461-induced force depressions are modulated by cMyBPC expression levels in the sarcomere, and emphasizes that clinical use of Myk461 may need to be optimized based on the molecular trigger that underlies the hypertrophic cardiomyopathy phenotype.


Assuntos
Benzilaminas/farmacologia , Coração/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Uracila/análogos & derivados , Actinas/metabolismo , Animais , Cálcio/metabolismo , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Contração Miocárdica/genética , Miosinas/metabolismo , Uracila/farmacologia
9.
Circ Heart Fail ; 10(10)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29030372

RESUMO

BACKGROUND: Omecamtiv mecarbil (OM) enhances systolic function in vivo by directly binding the myosin cross-bridges (XBs) in the sarcomere. However, the mechanistic details governing OM-induced modulation of XB behavior in failing human myocardium are unclear. METHODS AND RESULTS: The effects of OM on steady state and dynamic XB behavior were measured in chemically skinned myocardial preparations isolated from human donor and heart failure (HF) left ventricle. HF myocardium exhibited impaired contractile function as evidenced by reduced maximal force, magnitude of XB recruitment (Pdf), and a slowed rate of XB detachment (krel) at submaximal Ca2+ activations. Ca2+ sensitivity of force generation (pCa50) was higher in HF myocardium when compared with donor myocardium, both prior to and after OM incubations. OM incubation (0.5 and 1.0 µmol/L) enhanced force generation at submaximal Ca2+ activations in a dose-dependent manner. Notably, OM induced a slowing in krel with 1.0 µmol/L OM but not with 0.5 µmol/L OM in HF myocardium. Additionally, OM exerted other differential effects on XB behavior in HF myocardium as evidenced by a greater enhancement in Pdf and slowing in the time course of cooperative XB recruitment (Trec), which collectively prolonged achievement of peak force development (Tpk), compared with donor myocardium. CONCLUSIONS: Our findings demonstrate that OM augments force generation but also prolongs the time course of XB transitions to force-bearing states in remodeled HF myocardium, which may extend the systolic ejection time in vivo. Optimal OM dosing is critical for eliciting enhanced systolic function without excessive prolongation of systolic ejection time, which may compromise diastolic filling.


Assuntos
Cardiotônicos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Força Muscular/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miosinas/metabolismo , Ureia/análogos & derivados , Cardiotônicos/metabolismo , Proteínas de Transporte/metabolismo , Estudos de Casos e Controles , Relação Dose-Resposta a Droga , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Técnicas In Vitro , Fosforilação , Ligação Proteica , Sarcômeros/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Troponina I/metabolismo , Troponina T/metabolismo , Ureia/metabolismo , Ureia/farmacologia , Remodelação Ventricular
10.
Sci Adv ; 3(3): e1602445, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28345052

RESUMO

Phosphorylation of cardiac myosin binding protein-C (MyBP-C) modulates cardiac contractile function; however, the specific roles of individual serines (Ser) within the M-domain that are targets for ß-adrenergic signaling are not known. Recently, we demonstrated that significant accelerations in in vivo pressure development following ß-agonist infusion can occur in transgenic (TG) mouse hearts expressing phospho-ablated Ser282 (that is, TGS282A) but not in hearts expressing phospho-ablation of all three serines [that is, Ser273, Ser282, and Ser302 (TG3SA)], suggesting an important modulatory role for other Ser residues. In this regard, there is evidence that Ser302 phosphorylation may be a key contributor to the ß-agonist-induced positive inotropic responses in the myocardium, but its precise functional role has not been established. Thus, to determine the in vivo and in vitro functional roles of Ser302 phosphorylation, we generated TG mice expressing nonphosphorylatable Ser302 (that is, TGS302A). Left ventricular pressure-volume measurements revealed that TGS302A mice displayed no accelerations in the rate of systolic pressure rise and an inability to maintain systolic pressure following dobutamine infusion similar to TG3SA mice, implicating Ser302 phosphorylation as a critical regulator of enhanced systolic performance during ß-adrenergic stress. Dynamic strain-induced cross-bridge (XB) measurements in skinned myocardium isolated from TGS302A hearts showed that the molecular basis for impaired ß-adrenergic-mediated enhancements in systolic function is due to the absence of protein kinase A-mediated accelerations in the rate of cooperative XB recruitment. These results demonstrate that Ser302 phosphorylation regulates cardiac contractile reserve by enhancing contractile responses during ß-adrenergic stress.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Proteínas de Transporte/metabolismo , Ventrículos do Coração/metabolismo , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Proteínas de Transporte/genética , Camundongos , Camundongos Transgênicos , Contração Miocárdica/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Função Ventricular Esquerda/genética
11.
J Appl Physiol (1985) ; 122(3): 520-530, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-27909224

RESUMO

Molecular adaptations to chronic neurohormonal stress, including sarcomeric protein cleavage and phosphorylation, provide a mechanism to increase ventricular contractility and enhance cardiac output, yet the link between sarcomeric protein modifications and changes in myocardial function remains unclear. To examine the effects of neurohormonal stress on posttranslational modifications of sarcomeric proteins, mice were administered combined α- and ß-adrenergic receptor agonists (isoproterenol and phenylephrine, IPE) for 14 days using implantable osmotic pumps. In addition to significant cardiac hypertrophy and increased maximal ventricular pressure, IPE treatment accelerated pressure development and relaxation (74% increase in dP/dtmax and 14% decrease in τ), resulting in a 52% increase in cardiac output compared with saline (SAL)-treated mice. Accelerated pressure development was maintained when accounting for changes in heart rate and preload, suggesting that myocardial adaptations contribute to enhanced ventricular contractility. Ventricular myocardium isolated from IPE-treated mice displayed a significant reduction in troponin I (TnI) and myosin-binding protein C (MyBP-C) expression and a concomitant increase in the phosphorylation levels of the remaining TnI and MyBP-C protein compared with myocardium isolated from saline-treated control mice. Skinned myocardium isolated from IPE-treated mice displayed a significant acceleration in the rate of cross-bridge (XB) detachment (46% increase) and an enhanced magnitude of XB recruitment (43% increase) at submaximal Ca2+ activation compared with SAL-treated mice but unaltered myofilament Ca2+ sensitivity of force generation. These findings demonstrate that sarcomeric protein modifications during neurohormonal stress are molecular adaptations that enhance in vivo ventricular contractility through accelerated XB kinetics to increase cardiac output.NEW & NOTEWORTHY Posttranslational modifications to sarcomeric regulatory proteins provide a mechanism to modulate cardiac function in response to stress. In this study, we demonstrate that neurohormonal stress produces modifications to myosin-binding protein C and troponin I, including a reduction in protein expression within the sarcomere and increased phosphorylation of the remaining protein, which serve to enhance cross-bridge kinetics and increase cardiac output. These findings highlight the importance of sarcomeric regulatory protein modifications in modulating ventricular function during cardiac stress.


Assuntos
Débito Cardíaco/fisiologia , Proteínas de Transporte/metabolismo , Contração Miocárdica/fisiologia , Sarcômeros/fisiologia , Estresse Fisiológico/fisiologia , Troponina I/metabolismo , Citoesqueleto de Actina/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Cinética , Masculino , Camundongos , Miofibrilas/fisiologia
12.
Front Physiol ; 7: 38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26913007

RESUMO

Cardiac myosin binding protein-C (cMyBP-C) phosphorylation is an important regulator of contractile function, however, its contributions to length-dependent changes in cross-bridge (XB) kinetics is unknown. Therefore, we performed mechanical experiments to quantify contractile function in detergent-skinned ventricular preparations isolated from wild-type (WT) hearts, and hearts expressing non-phosphorylatable cMyBP-C [Ser to Ala substitutions at residues Ser273, Ser282, and Ser302 (i.e., 3SA)], at sarcomere length (SL) 1.9 µm or 2.1µm, prior and following protein kinase A (PKA) treatment. Steady-state force generation measurements revealed a blunting in the length-dependent increase in myofilament Ca(2+)-sensitivity of force generation (pCa50) following an increase in SL in 3SA skinned myocardium compared to WT skinned myocardium. Dynamic XB behavior was assessed at submaximal Ca(2+)-activations by imposing an acute rapid stretch of 2% of initial muscle length, and measuring both the magnitudes and rates of resultant phases of force decay due to strain-induced XB detachment and delayed force rise due to recruitment of additional XBs with increased SL (i.e., stretch activation). The magnitude (P2) and rate of XB detachment (k rel) following stretch was significantly reduced in 3SA skinned myocardium compared to WT skinned myocardium at short and long SL, and prior to and following PKA treatment. Furthermore, the length-dependent acceleration of k rel due to decreased SL that was observed in WT skinned myocardium was abolished in 3SA skinned myocardium. PKA treatment accelerated the rate of XB recruitment (k df) following stretch at both SL's in WT but not in 3SA skinned myocardium. The amplitude of the enhancement in force generation above initial pre-stretch steady-state levels (P3) was not different between WT and 3SA skinned myocardium at any condition measured. However, the magnitude of the entire delayed force phase which can dip below initial pre-stretch steady-state levels (Pdf) was significantly lower in 3SA skinned myocardium under all conditions, in part due to a reduced magnitude of XB detachment (P2) in 3SA skinned myocardium compared to WT skinned myocardium. These findings demonstrate that cMyBP-C phospho-ablation regulates SL- and PKA-mediated effects on XB kinetics in the myocardium, which would be expected to contribute to the regulation of the Frank-Starling mechanism.

13.
J Mol Cell Cardiol ; 85: 262-72, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26100051

RESUMO

Decreased expression of cardiac myosin binding protein-C (cMyBP-C) in the myocardium is thought to be a contributing factor to hypertrophic cardiomyopathy in humans, and the initial molecular defect is likely abnormal cross-bridge (XB) function which leads to impaired force generation, decreased contractile performance, and hypertrophy in vivo. The myosin activator omecamtiv mecarbil (OM) is a pharmacological drug that specifically targets the myosin XB and recent evidence suggests that OM induces a significant decrease in the in vivo motility velocity and an increase in the XB duty cycle. Thus, the molecular effects of OM maybe beneficial in improving contractile function in skinned myocardium lacking cMyBP-C because absence of cMyBP-C in the sarcomere accelerates XB kinetics and enhances XB turnover rate, which presumably reduces contractile efficiency. Therefore, parameters of XB function were measured in skinned myocardium lacking cMyBP-C prior to and following OM incubation. We measured ktr, the rate of force redevelopment as an index of XB transition from both the weakly- to strongly-bound state and from the strongly- to weakly-bound states and performed stretch activation experiments to measure the rates of XB detachment (krel) and XB recruitment (kdf) in detergent-skinned ventricular preparations isolated from hearts of wild-type (WT) and cMyBP-C knockout (KO) mice. Samples from donor human hearts were also used to assess the effects of OM in cardiac muscle expressing a slow ß-myosin heavy chain (ß-MHC). Incubation of skinned myocardium with OM produced large enhancements in steady-state force generation which were most pronounced at low levels of [Ca(2+)] activations, suggesting that OM cooperatively recruits additional XB's into force generating states. Despite a large increase in steady-state force generation following OM incubation, parallel accelerations in XB kinetics as measured by ktr were not observed, and there was a significant OM-induced decrease in krel which was more pronounced in the KO skinned myocardium compared to WT skinned myocardium (58% in WT vs. 76% in KO at pCa 6.1), such that baseline differences in krel between KO and WT skinned myocardium were no longer apparent following OM-incubation. A significant decrease in the kdf was also observed following OM incubation in all groups, which may be related to the increase in the number of cooperatively recruited XB's at low Ca(2+)-activations which slows the overall rate of force generation. Our results indicate that OM may be a useful pharmacological approach to normalize hypercontractile XB kinetics in myocardium with decreased cMyBP-C expression due to its molecular effects on XB behavior.


Assuntos
Proteínas de Transporte/metabolismo , Ativadores de Enzimas/farmacologia , Contração Miocárdica/efeitos dos fármacos , Ureia/análogos & derivados , Animais , Cálcio/fisiologia , Proteínas de Transporte/genética , Feminino , Humanos , Cinética , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Miocárdio/metabolismo , Miosinas/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Ureia/farmacologia
14.
Front Physiol ; 5: 461, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25520665

RESUMO

Enhanced cardiac contractile function with increased sarcomere length (SL) is, in part, mediated by a decrease in the radial distance between myosin heads and actin. The radial disposition of myosin heads relative to actin is modulated by cardiac myosin binding protein-C (cMyBP-C), suggesting that cMyBP-C contributes to the length-dependent activation (LDA) in the myocardium. However, the precise roles of cMyBP-C in modulating cardiac LDA are unclear. To determine the impact of cMyBP-C on LDA, we measured isometric force, myofilament Ca(2+)-sensitivity (pCa50) and length-dependent changes in kinetic parameters of cross-bridge (XB) relaxation (k rel), and recruitment (k df) due to rapid stretch, as well as the rate of force redevelopment (k tr) in response to a large slack-restretch maneuver in skinned ventricular multicellular preparations isolated from the hearts of wild-type (WT) and cMyBP-C knockout (KO) mice, at SL's 1.9 µm or 2.1 µm. Our results show that maximal force was not significantly different between KO and WT preparations but length-dependent increase in pCa50 was attenuated in the KO preparations. pCa50 was not significantly different between WT and KO preparations at long SL (5.82 ± 0.02 in WT vs. 5.87 ± 0.02 in KO), whereas pCa50 was significantly different between WT and KO preparations at short SL (5.71 ± 0.02 in WT vs. 5.80 ± 0.01 in KO; p < 0.05). The k tr, measured at half-maximal Ca(2+)-activation, was significantly accelerated at short SL in WT preparations (8.74 ± 0.56 s(-1) at 1.9 µm vs. 5.71 ± 0.40 s(-1) at 2.1 µm, p < 0.05). Furthermore, k rel and k df were accelerated by 32% and 50%, respectively at short SL in WT preparations. In contrast, k tr was not altered by changes in SL in KO preparations (8.03 ± 0.54 s(-1) at 1.9 µm vs. 8.90 ± 0.37 s(-1) at 2.1 µm). Similarly, KO preparations did not exhibit length-dependent changes in k rel and k df. Collectively, our data implicate cMyBP-C as an important regulator of LDA via its impact on dynamic XB behavior due to changes in SL.

15.
J Physiol ; 592(17): 3747-65, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24951619

RESUMO

Cardiac myosin binding protein-C phosphorylation plays an important role in modulating cardiac muscle function and accelerating contraction. It has been proposed that Ser282 phosphorylation may serve as a critical molecular switch that regulates the phosphorylation of neighbouring Ser273 and Ser302 residues, and thereby govern myofilament contractile acceleration in response to protein kinase A (PKA). Therefore, to determine the regulatory roles of Ser282 we generated a transgenic (TG) mouse model expressing cardiac myosin binding protein-C with a non-phosphorylatable Ser282 (i.e. serine to alanine substitution, TG(S282A)). Myofibrils isolated from TG(S282A) hearts displayed robust PKA-mediated phosphorylation of Ser273 and Ser302, and the increase in phosphorylation was identical to TG wild-type (TG(WT)) controls. No signs of pathological cardiac hypertrophy were detected in TG(S282A) hearts by either histological examination of cardiac sections or echocardiography. Baseline fractional shortening, ejection fraction, isovolumic relaxation time, rate of pressure development and rate of relaxation (τ) were unaltered in TG(S282A) mice. However, the increase in cardiac contractility as well as the acceleration of pressure development observed in response to ß-adrenergic stimulation was attenuated in TG(S282A) mice. In agreement with our in vivo data, in vitro force measurements revealed that PKA-mediated acceleration of cross-bridge kinetics in TG(S282A) myocardium was significantly attenuated compared to TG(WT) myocardium. Taken together, our data suggest that while Ser282 phosphorylation does not regulate the phosphorylation of neighbouring Ser residues and basal cardiac function, full acceleration of cross-bridge kinetics and left ventricular pressure development cannot be achieved in its absence.


Assuntos
Proteínas de Transporte/metabolismo , Mutação , Contração Miocárdica , Serina/genética , Agonistas Adrenérgicos beta/farmacologia , Animais , Proteínas de Transporte/genética , Feminino , Coração/efeitos dos fármacos , Coração/fisiologia , Masculino , Camundongos , Miocárdio/metabolismo , Miofibrilas/metabolismo , Miofibrilas/fisiologia , Fosforilação
16.
Pflugers Arch ; 466(2): 225-30, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24310821

RESUMO

Through its ability to interact with both the thick and thin filament proteins within the sarcomere, cardiac myosin binding protein-C (cMyBP-C) regulates the contractile properties of the myocardium. The central regulatory role of cMyBP-C in heart function is emphasized by the fact that a large proportion of inherited hypertrophic cardiomyopathy cases in humans are caused by mutations in cMyBP-C. The primary dysfunction in cMyBP-C-related cardiomyopathies is likely to be abnormal myofilament contractile function; however, currently, there are no effective therapies for ameliorating these contractile defects. Thus, there is a compelling need to design novel therapies to restore normal contractile function in cMyBP-C-related cardiomyopathies. To this end, concepts gleaned from various structural, functional, and biochemical studies can now be utilized to engineer cMyBP-C proteins that, when incorporated into the sarcomere, can significantly improve contractile function. In this review, we discuss the rationale for cMyBP-C-based gene therapies that can be utilized to treat contractile dysfunction in inherited and acquired cardiomyopathies.


Assuntos
Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Terapia Genética/métodos , Contração Miocárdica/efeitos dos fármacos , Cardiomiopatia Hipertrófica/terapia , Proteínas de Transporte/biossíntese , Humanos , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miofibrilas/metabolismo , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo
17.
Biophys J ; 105(9): 2104-13, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24209855

RESUMO

The causal link between disparate tropomyosin (Tm) functions and the structural instability in Tm is unknown. To test the hypothesis that the structural instability in the central region of Tm modulates the function of the overlapping ends of contiguous Tm dimers, we used transgenic mice (Tm(DM)) that expressed a mutant α-Tm in the heart; S229E and H276N substitutions induce structural instability in the central region and the overlapping ends of Tm, respectively. In addition, two mouse cardiac troponin T mutants (TnT(1-44Δ) and TnT(45-74Δ)) that have a divergent effect on the overlapping ends of Tm were employed. The S229E-induced instability in the central region of Tm(DM) altered the overlapping ends of Tm(DM), thereby it negated the attenuating effect of H276N on Ca(2+)-activated maximal tension. The rate of cross-bridge detachment (g) decreased in Tm(DM)+TnT(WT) and Tm(H276N)+TnT(WT) fibers but increased in Tm(DM)+TnT(45-74Δ) fibers; however, TnT(45-74Δ) did not alter g, demonstrating that S229E in Tm(DM) had divergent effects on g. The S229E substitution in Tm(DM) ablated the H276N-induced desensitization of myofilament Ca(2+) sensitivity in Tm(DM)+TnT(1-44Δ) fibers. To our knowledge, novel findings from this study show that the structural instability in the central region of Tm modifies cardiac contractile function via its effect on the overlapping ends of contiguous Tm.


Assuntos
Tropomiosina/química , Tropomiosina/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Cálcio/metabolismo , Cinética , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estabilidade Proteica , Tropomiosina/genética
18.
J Gen Physiol ; 142(4): 413-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24043862

RESUMO

Divergent effects of α- and ß-myosin heavy chain (MHC) isoforms on contractile behavior arise mainly because of their impact on thin filament cooperativity. The N terminus of cardiac troponin T (cTnT) also modulates thin filament cooperativity. Our hypothesis is that the impact of the N terminus of cTnT on thin filament activation is modulated by a shift from α- to ß-MHC isoform. We engineered two recombinant proteins by deleting residues 1-43 and 44-73 in rat cTnT (RcTnT): RcTnT(1-43Δ) and RcTnT(44-73Δ), respectively. Dynamic and steady-state contractile parameters were measured at sarcomere length of 2.3 µm after reconstituting proteins into detergent-skinned muscle fibers from normal (α-MHC) and propylthiouracil-treated (ß-MHC) rat hearts. α-MHC attenuated Ca(2+)-activated maximal tension (∼46%) in RcTnT(1-43Δ) fibers. In contrast, ß-MHC decreased tension only by 19% in RcTnT(1-43Δ) fibers. Both α- and ß-MHC did not affect tension in RcTnT(44-73Δ) fibers. The instantaneous muscle fiber stiffness measurements corroborated the divergent impact of α- and ß-MHC on tension in RcTnT(1-43Δ) fibers. pCa50 (-log of [Ca(2+)]free required for half-maximal activation) decreased significantly by 0.13 pCa units in α-MHC + RcTnT(1-43Δ) fibers but remained unaltered in ß-MHC + RcTnT(1-43Δ) fibers, demonstrating that ß-MHC counteracted the attenuating effect of RcTnT(1-43Δ) on myofilament Ca(2+) sensitivity. ß-MHC did not alter the sudden stretch-mediated recruitment of new cross-bridges (ER) in RcTnT(1-43Δ) fibers, but α-MHC attenuated ER by 36% in RcTnT(1-43Δ) fibers. The divergent impact of α- and ß-MHC on how the N terminus of cTnT modulates contractile dynamics has implications for heart disease; alterations in cTnT and MHC are known to occur via changes in isoform expression or mutations.


Assuntos
Cadeias Pesadas de Miosina/metabolismo , Sarcômeros/fisiologia , Troponina T/metabolismo , Miosinas Ventriculares/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Masculino , Dados de Sequência Molecular , Contração Muscular , Mutação , Propiltiouracila/farmacologia , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Troponina T/química , Troponina T/genética
19.
FASEB J ; 27(9): 3848-59, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23748972

RESUMO

The functional significance of the molecular swivel at the head-to-tail overlapping ends of contiguous tropomyosin (Tm) dimers in striated muscle is unknown. Contractile measurements were made in muscle fibers from transgenic (TG) mouse hearts that expressed a mutant α-Tm (Tm(H276N)). We also reconstituted mouse cardiac troponin T (McTnT) N-terminal deletion mutants, McTnT(1-44Δ) and McTnT(45-74Δ), into muscle fibers from Tm(H276N). For controls, we used the wild-type (WT) McTnT because altered effects could be correlated with the mutant forms of McTnT. Tm(H276N) slowed crossbridge (XB) detachment rate (g) by 19%. McTnT(1-44Δ) attenuated Ca(2+)-activated maximal tension against Tm(WT) (36%) and Tm(H276N) (38%), but sped g only against Tm(H276N) by 35%. The rate of tension redevelopment decreased (17%) only in McTnT(1-44Δ) + Tm(H276N) fibers. McTnT(45-74Δ) attenuated tension (19%) and myofilament Ca(2+) sensitivity (pCa50=5.93 vs. 6.00 in the control fibers) against Tm(H276N), but not against Tm(WT) background. Thus, altered XB cycling kinetics decreased the fraction of strongly bound XBs in McTnT(1-44Δ) + Tm(H276N) fibers, whereas diminished thin-filament cooperativity attenuated tension in McTnT(45-74Δ) + Tm(H276N) fibers. In summary, our study is the first to show that the interplay between the N terminus of cTnT and the overlapping ends of contiguous Tm effectuates different states of Tm on the actin filament.


Assuntos
Actinas/metabolismo , Tropomiosina/metabolismo , Troponina T/metabolismo , Actinas/genética , Animais , Western Blotting , Cálcio/metabolismo , Eletroforese em Gel de Poliacrilamida , Camundongos , Camundongos Transgênicos , Músculo Estriado/metabolismo , Contração Miocárdica/genética , Contração Miocárdica/fisiologia , Tropomiosina/genética
20.
J Physiol ; 591(5): 1217-34, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23207592

RESUMO

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


Assuntos
Contração Miocárdica , Músculos Papilares/metabolismo , Troponina T/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Eletroforese em Gel de Poliacrilamida , Cinética , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Mutação , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Isoformas de Proteínas , Proteínas Recombinantes/metabolismo , Tropomiosina/genética , Tropomiosina/metabolismo , Troponina T/química , Troponina T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...