Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(2): 105652, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38224947

RESUMEN

The physiological importance of cardiac myosin regulatory light chain (RLC) phosphorylation by its dedicated cardiac myosin light chain kinase has been established in both humans and mice. Constitutive RLC-phosphorylation, regulated by the balanced activities of cardiac myosin light chain kinase and myosin light chain phosphatase (MLCP), is fundamental to the biochemical and physiological properties of myofilaments. However, limited information is available on cardiac MLCP. In this study, we hypothesized that the striated muscle-specific MLCP regulatory subunit, MYPT2, targets the phosphatase catalytic subunit to cardiac myosin, contributing to the maintenance of cardiac function in vivo through the regulation of RLC-phosphorylation. To test this hypothesis, we generated a floxed-PPP1R12B mouse model crossed with a cardiac-specific Mer-Cre-Mer to conditionally ablate MYPT2 in adult cardiomyocytes. Immunofluorescence microscopy using the gene-ablated tissue as a control confirmed the localization of MYPT2 to regions where it overlaps with a subset of RLC. Biochemical analysis revealed an increase in RLC-phosphorylation in vivo. The loss of MYPT2 demonstrated significant protection against pressure overload-induced hypertrophy, as evidenced by heart weight, qPCR of hypertrophy-associated genes, measurements of myocyte diameters, and expression of ß-MHC protein. Furthermore, mantATP chase assays revealed an increased ratio of myosin heads distributed to the interfilament space in MYPT2-ablated heart muscle fibers, confirming that RLC-phosphorylation regulated by MLCP, enhances cardiac performance in vivo. Our findings establish MYPT2 as the regulatory subunit of cardiac MLCP, distinct from the ubiquitously expressed canonical smooth muscle MLCP. Targeting MYPT2 to increase cardiac RLC-phosphorylation in vivo may improve baseline cardiac performance, thereby attenuating pathological hypertrophy.


Asunto(s)
Miocitos Cardíacos , Quinasa de Cadena Ligera de Miosina , Animales , Humanos , Ratones , Hipertrofia/metabolismo , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Ratones Endogámicos C57BL
2.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35177471

RESUMEN

In this study, we investigated the role of the super-relaxed (SRX) state of myosin in the structure-function relationship of sarcomeres in the hearts of mouse models of cardiomyopathy-bearing mutations in the human ventricular regulatory light chain (RLC, MYL2 gene). Skinned papillary muscles from hypertrophic (HCM-D166V) and dilated (DCM-D94A) cardiomyopathy models were subjected to small-angle X-ray diffraction simultaneously with isometric force measurements to obtain the interfilament lattice spacing and equatorial intensity ratios (I11/I10) together with the force-pCa relationship over a full range of [Ca2+] and at a sarcomere length of 2.1 µm. In parallel, we studied the effect of mutations on the ATP-dependent myosin energetic states. Compared with wild-type (WT) and DCM-D94A mice, HCM-D166V significantly increased the Ca2+ sensitivity of force and left shifted the I11/I10-pCa relationship, indicating an apparent movement of HCM-D166V cross-bridges closer to actin-containing thin filaments, thereby allowing for their premature Ca2+ activation. The HCM-D166V model also disrupted the SRX state and promoted an SRX-to-DRX (super-relaxed to disordered relaxed) transition that correlated with an HCM-linked phenotype of hypercontractility. While this dysregulation of SRX ↔ DRX equilibrium was consistent with repositioning of myosin motors closer to the thin filaments and with increased force-pCa dependence for HCM-D166V, the DCM-D94A model favored the energy-conserving SRX state, but the structure/function-pCa data were similar to WT. Our results suggest that the mutation-induced redistribution of myosin energetic states is one of the key mechanisms contributing to the development of complex clinical phenotypes associated with human HCM-D166V and DCM-D94A mutations.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatías/metabolismo , Cadenas Ligeras de Miosina/genética , Actinas/metabolismo , Animales , Miosinas Cardíacas/metabolismo , Cardiomiopatías/genética , Cardiomiopatía Hipertrófica/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertrofia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Contracción Miocárdica/genética , Cadenas Ligeras de Miosina/metabolismo , Miosinas/metabolismo , Miosinas/fisiología , Fenotipo , Fosforilación , Sarcómeros/metabolismo , Relación Estructura-Actividad , Difracción de Rayos X/métodos
3.
Medicina (Kaunas) ; 60(7)2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39064613

RESUMEN

Background and Objectives: Biodex System® is an advanced dynamometer used for testing various biomechanical parameters of muscles. Test outcomes allow for the identification of muscle pathology and consequently lead to a clinical diagnosis. Despite being widely used for the testing and rehabilitation of the human musculoskeletal system, no universal and acceptable protocol for wrist examination has been proposed for patients with wrist pathology. In this study, the authors aim to identify the most appropriate protocol for testing the biomechanical parameters of flexors and extensors of the wrist. Materials and Methods: A group of 20 patients with symptomatic tennis elbow and 26 healthy volunteers were examined using three different protocols: isokinetic, isometric and isotonic. Protocol order for each study participant was assigned at random with a minimum of a 24 h break between protocols. All protocol parameters were set according to data obtained from a literature review and an earlier pilot study. Following completion of each protocol, participants filled out a questionnaire-based protocol, assessing pain intensity during the exam, difficulty with exam performance and post-exam muscle fatigue. Results: The isotonic protocol showed the best patient tolerance and the highest questionnaire score. There was a significant difference (p < 0.05) between the three protocols in average pain intensity reported by study participants. All participants completed the isotonic protocol, but not all patients with symptomatic tennis elbow were able to complete the isometric and isokinetic protocols. The isotonic protocol was deemed "difficult but possible to complete" by study participants. Conclusions: The isotonic protocol is most suitable for testing the flexors and extensors of the wrist. It gives the most biomechanical data of all protocols, is well tolerated by patients and rarely causes pain during examination even in symptomatic participants.


Asunto(s)
Dinamómetro de Fuerza Muscular , Codo de Tenista , Muñeca , Humanos , Masculino , Adulto , Femenino , Fenómenos Biomecánicos , Codo de Tenista/fisiopatología , Codo de Tenista/diagnóstico , Muñeca/fisiología , Muñeca/fisiopatología , Persona de Mediana Edad , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Contracción Isométrica/fisiología , Encuestas y Cuestionarios , Contracción Isotónica/fisiología
4.
Am J Physiol Heart Circ Physiol ; 324(6): H739-H750, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36897749

RESUMEN

Heart failure (HF) with preserved ejection fraction (HFpEF) represents a major unmet medical need owing to its diverse pathophysiology and lack of effective therapies. Potent synthetic, agonists (MR-356 and MR-409) of growth hormone-releasing hormone (GHRH) improve the phenotype of models of HF with reduced ejection fraction (HFrEF) and in cardiorenal models of HFpEF. Endogenous GHRH exhibits a broad range of regulatory influences in the cardiovascular (CV) system and aging and plays a role in several cardiometabolic conditions including obesity and diabetes. Whether agonists of GHRH can improve the phenotype of cardiometabolic HFpEF remains untested and unknown. Here we tested the hypothesis that MR-356 can mitigate/reverse the cardiometabolic HFpEF phenotype. C57BL6N mice received a high-fat diet (HFD) plus the nitric oxide synthase inhibitor (l-NAME) for 9 wk. After 5 wk of HFD + l-NAME regimen, animals were randomized to receive daily injections of MR-356 or placebo during a 4-wk period. Control animals received no HFD + l-NAME or agonist treatment. Our results showed the unique potential of MR-356 to treat several HFpEF-like features including cardiac hypertrophy, fibrosis, capillary rarefaction, and pulmonary congestion. MR-356 improved cardiac performance by improving diastolic function, global longitudinal strain (GLS), and exercise capacity. Importantly, the increased expression of cardiac pro-brain natriuretic peptide (pro-BNP), inducible nitric oxide synthase (iNOS), and vascular endothelial growth factor-A (VEGF-A) was restored to normal levels suggesting that MR-356 reduced myocardial stress associated with metabolic inflammation in HFpEF. Thus, agonists of GHRH may be an effective therapeutic strategy for the treatment of cardiometabolic HFpEF phenotype.NEW & NOTEWORTHY This randomized study used rigorous hemodynamic tools to test the efficacy of a synthetic GHRH agonist to improve cardiac performance in a cardiometabolic HFpEF. Daily injection of the GHRH agonist, MR-356, reduced the HFpEF-like effects as evidenced by improved diastolic dysfunction, reduced cardiac hypertrophy, fibrosis, and pulmonary congestion. Notably, end-diastolic pressure and end-diastolic pressure-volume relationship were reset to control levels. Moreover, treatment with MR-356 increased exercise capacity and reduced myocardial stress associated with metabolic inflammation in HFpEF.


Asunto(s)
Insuficiencia Cardíaca , Animales , Ratones , Cardiomegalia , Modelos Animales de Enfermedad , Fibrosis , Hormona Liberadora de Hormona del Crecimiento , Inflamación , NG-Nitroarginina Metil Éster , Volumen Sistólico/fisiología , Factor A de Crecimiento Endotelial Vascular , Función Ventricular Izquierda
5.
Int J Mol Sci ; 23(24)2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36555229

RESUMEN

In this study, we aimed to investigate whether short-term and low-dose treatment with hydroxychloroquine (HCQ), an antimalarial drug, can modulate heart function in a preclinical model of dilated cardiomyopathy (DCM) expressing the D94A mutation in cardiac myosin regulatory light chain (RLC) compared with healthy non-transgenic (NTg) littermates. Increased interest in HCQ came with the COVID-19 pandemic, but the risk of cardiotoxic side effects of HCQ raised concerns, especially in patients with an underlying heart condition, e.g., cardiomyopathy. Effects of HCQ treatment vs. placebo (H2O), administered in Tg-D94A vs. NTg mice over one month, were studied by echocardiography and muscle contractile mechanics. Global longitudinal strain analysis showed the HCQ-mediated improvement in heart performance in DCM mice. At the molecular level, HCQ promoted the switch from myosin's super-relaxed (SRX) to disordered relaxed (DRX) state in DCM-D94A hearts. This result indicated more myosin cross-bridges exiting a hypocontractile SRX-OFF state and assuming the DRX-ON state, thus potentially enhancing myosin motor function in DCM mice. This bottom-up investigation of the pharmacological use of HCQ at the level of myosin molecules, muscle fibers, and whole hearts provides novel insights into mechanisms by which HCQ therapy mitigates some abnormal phenotypes in DCM-D94A mice and causes no harm in healthy NTg hearts.


Asunto(s)
COVID-19 , Cardiomiopatía Dilatada , Ratones , Humanos , Animales , Ratones Transgénicos , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/genética , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Pandemias , Tratamiento Farmacológico de COVID-19 , Mutación , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Fenotipo , Contracción Miocárdica
6.
Am J Physiol Heart Circ Physiol ; 320(2): H881-H890, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337957

RESUMEN

Morbidity and mortality associated with heart disease is a growing threat to the global population, and novel therapies are needed. Mavacamten (formerly called MYK-461) is a small molecule that binds to cardiac myosin and inhibits myosin ATPase. Mavacamten is currently in clinical trials for the treatment of obstructive hypertrophic cardiomyopathy (HCM), and it may provide benefits for treating other forms of heart disease. We investigated the effect of mavacamten on cardiac muscle contraction in two transgenic mouse lines expressing the human isoform of cardiac myosin regulatory light chain (RLC) in their hearts. Control mice expressed wild-type RLC (WT-RLC), and HCM mice expressed the N47K RLC mutation. In the absence of mavacamten, skinned papillary muscle strips from WT-RLC mice produced greater isometric force than strips from N47K mice. Adding 0.3 µM mavacamten decreased maximal isometric force and reduced Ca2+ sensitivity of contraction for both genotypes, but this reduction in pCa50 was nearly twice as large for WT-RLC versus N47K. We also used stochastic length-perturbation analysis to characterize cross-bridge kinetics. The cross-bridge detachment rate was measured as a function of [MgATP] to determine the effect of mavacamten on myosin nucleotide handling rates. Mavacamten increased the MgADP release and MgATP binding rates for both genotypes, thereby contributing to faster cross-bridge detachment, which could speed up myocardial relaxation during diastole. Our data suggest that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction via decreased strong cross-bridge binding. Mavacamten may become a useful therapy for patients with heart disease, including some forms of HCM.NEW & NOTEWORTHY Mavacamten is a pharmaceutical that binds to myosin, and it is under investigation as a therapy for some forms of heart disease. We show that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction in skinned myocardial strips from a mouse model of hypertrophic cardiomyopathy that expresses the N47K mutation in cardiac myosin regulatory light chain. Mavacamten reduces contractility by decreasing strong cross-bridge binding, partially due to faster cross-bridge nucleotide handling rates that speed up myosin detachment.


Asunto(s)
Bencilaminas/farmacología , Señalización del Calcio/efectos de los fármacos , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Contracción Miocárdica/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Músculos Papilares/efectos de los fármacos , Uracilo/análogos & derivados , Miosinas Ventriculares/antagonistas & inhibidores , Animales , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Humanos , Cinética , Masculino , Ratones Transgénicos , Mutación , Cadenas Ligeras de Miosina/genética , Músculos Papilares/enzimología , Músculos Papilares/fisiopatología , Uracilo/farmacología , Miosinas Ventriculares/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(10): E2338-E2347, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463717

RESUMEN

Dilated cardiomyopathy (DCM) is a devastating heart disease that affects about 1 million people in the United States, but the underlying mechanisms remain poorly understood. In this study, we aimed to determine the biomechanical and structural causes of DCM in transgenic mice carrying a novel mutation in the MYL2 gene, encoding the cardiac myosin regulatory light chain. Transgenic D94A (aspartic acid-to-alanine) mice were created and investigated by echocardiography and invasive hemodynamic and molecular structural and functional assessments. Consistent with the DCM phenotype, a significant reduction of the ejection fraction (EF) was observed in ∼5- and ∼12-mo-old male and female D94A lines compared with respective WT controls. Younger male D94A mice showed a more pronounced left ventricular (LV) chamber dilation compared with female counterparts, but both sexes of D94A lines developed DCM by 12 mo of age. The hypocontractile activity of D94A myosin motors resulted in the rightward shift of the force-pCa dependence and decreased actin-activated myosin ATPase activity. Consistent with a decreased Ca2+ sensitivity of contractile force, a small-angle X-ray diffraction study, performed in D94A fibers at submaximal Ca2+ concentrations, revealed repositioning of the D94A cross-bridge mass toward the thick-filament backbone supporting the hypocontractile state of D94A myosin motors. Our data suggest that structural perturbations at the level of sarcomeres result in aberrant cardiomyocyte cytoarchitecture and lead to LV chamber dilation and decreased EF, manifesting in systolic dysfunction of D94A hearts. The D94A-induced development of DCM in mice closely follows the clinical phenotype and suggests that MYL2 may serve as a new therapeutic target for dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Sarcómeros/metabolismo , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación Missense , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/genética , Sarcómeros/genética
8.
FASEB J ; 33(3): 3152-3166, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30365366

RESUMEN

Myosin light chain 2 ( MYL2) gene encodes the myosin regulatory light chain (RLC) simultaneously in heart ventricles and in slow-twitch skeletal muscle. Using transgenic mice with cardiac-specific expression of the human R58Q-RLC mutant, we sought to determine whether the hypertrophic cardiomyopathy phenotype observed in papillary muscles (PMs) of R58Q mice is also manifested in slow-twitch soleus (SOL) muscles. Skinned SOL muscles and ventricular PMs of R58Q animals exhibited lower contractile force that was not observed in the fast-twitch extensor digitorum longus muscles of R58Q vs. wild-type-RLC mice, but mutant animals did not display gross muscle weakness in vivo. Consistent with SOL muscle abnormalities in R58Q vs. wild-type mice, myosin ATPase staining revealed a decreased proportion of fiber type I/type II only in SOL muscles but not in the extensor digitorum longus muscles. The similarities between SOL muscles and PMs of R58Q mice were further supported by quantitative proteomics. Differential regulation of proteins involved in energy metabolism, cell-cell interactions, and protein-protein signaling was concurrently observed in the hearts and SOL muscles of R58Q mice. In summary, even though R58Q expression was restricted to the heart of mice, functional similarities were clearly observed between the hearts and slow-twitch skeletal muscle, suggesting that MYL2 mutated models of hypertrophic cardiomyopathy may be useful research tools to study the molecular, structural, and energetic mechanisms of cardioskeletal myopathy associated with myosin RLC.-Kazmierczak, K., Liang, J., Yuan, C.-C., Yadav, S., Sitbon, Y. H., Walz, K., Ma, W., Irving, T. C., Cheah, J. X., Gomes, A. V., Szczesna-Cordary, D. Slow-twitch skeletal muscle defects accompany cardiac dysfunction in transgenic mice with a mutation in the myosin regulatory light chain.


Asunto(s)
Miosinas Cardíacas/genética , Miosinas Cardíacas/fisiología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Fibras Musculares de Contracción Lenta/fisiología , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/fisiología , Sustitución de Aminoácidos , Animales , Cardiomiopatía Hipertrófica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Contracción Muscular/genética , Contracción Muscular/fisiología , Fibras Musculares de Contracción Lenta/patología , Mutación Missense , Contracción Miocárdica/genética , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Miocardio/patología , Músculos Papilares/patología , Músculos Papilares/fisiopatología , Proteómica
9.
J Muscle Res Cell Motil ; 41(4): 313-327, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31131433

RESUMEN

The activity of cardiac and skeletal muscles depends upon the ATP-coupled actin-myosin interactions to execute the power stroke and muscle contraction. The goal of this review article is to provide insight into the function of myosin II, the molecular motor of the heart and skeletal muscles, with a special focus on the role of myosin II light chain (MLC) components. Specifically, we focus on the involvement of myosin regulatory (RLC) and essential (ELC) light chains in striated muscle development, isoform appearance and their function in normal and diseased muscle. We review the consequences of isoform switching and knockout of specific MLC isoforms on cardiac and skeletal muscle function in various animal models. Finally, we discuss how dysregulation of specific RLC/ELC isoforms can lead to cardiac and skeletal muscle diseases and summarize the effects of most studied mutations leading to cardiac or skeletal myopathies.


Asunto(s)
Músculo Esquelético/metabolismo , Miocardio/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Animales , Humanos , Ratones
10.
Pflugers Arch ; 471(5): 683-699, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30706179

RESUMEN

Genetic cardiomyopathies, a group of cardiovascular disorders based on ventricular morphology and function, are among the leading causes of morbidity and mortality worldwide. Such genetically driven forms of hypertrophic (HCM), dilated (DCM), and restrictive (RCM) cardiomyopathies are chronic, debilitating diseases that result from biomechanical defects in cardiac muscle contraction and frequently progress to heart failure (HF). Locus and allelic heterogeneity, as well as clinical variability combined with genetic and phenotypic overlap between different cardiomyopathies, have challenged proper clinical prognosis and provided an incentive for identification of pathogenic variants. This review attempts to provide an overview of inherited cardiomyopathies with a focus on their genetic etiology in myosin regulatory (RLC) and essential (ELC) light chains, which are EF-hand protein family members with important structural and regulatory roles. From the clinical discovery of cardiomyopathy-linked light chain mutations in patients to an array of exploratory studies in animals, and reconstituted and recombinant systems, we have summarized the current state of knowledge on light chain mutations and how they induce physiological disease states via biochemical and biomechanical alterations at the molecular, tissue, and organ levels. Cardiac myosin RLC phosphorylation and the N-terminus ELC have been discussed as two important emerging modalities with important implications in the regulation of myosin motor function, and thus cardiac performance. A comprehensive understanding of such triggers is absolutely necessary for the development of target-specific rescue strategies to ameliorate or reverse the effects of myosin light chain-related inherited cardiomyopathies.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Restrictiva/genética , Cadenas Ligeras de Miosina/genética , Animales , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Restrictiva/etiología , Cardiomiopatía Restrictiva/patología , Modelos Animales de Enfermedad , Humanos , Mutación
11.
Proc Natl Acad Sci U S A ; 112(30): E4138-46, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26124132

RESUMEN

Myosin light chain kinase (MLCK)-dependent phosphorylation of the regulatory light chain (RLC) of cardiac myosin is known to play a beneficial role in heart disease, but the idea of a phosphorylation-mediated reversal of a hypertrophic cardiomyopathy (HCM) phenotype is novel. Our previous studies on transgenic (Tg) HCM-RLC mice revealed that the D166V (Aspartate166 → Valine) mutation-induced changes in heart morphology and function coincided with largely reduced RLC phosphorylation in situ. We hypothesized that the introduction of a constitutively phosphorylated Serine15 (S15D) into the hearts of D166V mice would prevent the development of a deleterious HCM phenotype. In support of this notion, MLCK-induced phosphorylation of D166V-mutated hearts was found to rescue some of their abnormal contractile properties. Tg-S15D-D166V mice were generated with the human cardiac RLC-S15D-D166V construct substituted for mouse cardiac RLC and were subjected to functional, structural, and morphological assessments. The results were compared with Tg-WT and Tg-D166V mice expressing the human ventricular RLC-WT or its D166V mutant, respectively. Echocardiography and invasive hemodynamic studies demonstrated significant improvements of intact heart function in S15D-D166V mice compared with D166V, with the systolic and diastolic indices reaching those monitored in WT mice. A largely reduced maximal tension and abnormally high myofilament Ca(2+) sensitivity observed in D166V-mutated hearts were reversed in S15D-D166V mice. Low-angle X-ray diffraction study revealed that altered myofilament structures present in HCM-D166V mice were mitigated in S15D-D166V rescue mice. Our collective results suggest that expression of pseudophosphorylated RLC in the hearts of HCM mice is sufficient to prevent the development of the pathological HCM phenotype.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Animales , Calcio/química , Cristalografía por Rayos X , Progresión de la Enfermedad , Ecocardiografía , Femenino , Corazón/fisiopatología , Hemodinámica , Humanos , Hipertrofia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Contracción Miocárdica , Miofibrillas/metabolismo , Fenotipo , Fosforilación , Estructura Secundaria de Proteína , Difracción de Rayos X
12.
Biochemistry ; 55(1): 186-98, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26671638

RESUMEN

Muscle myosin cyclically hydrolyzes ATP to translate actin. Ventricular cardiac myosin (ßmys) moves actin with three distinct unitary step-sizes resulting from its lever-arm rotation and with step-frequencies that are modulated in a myosin regulation mechanism. The lever-arm associated essential light chain (vELC) binds actin by its 43 residue N-terminal extension. Unitary steps were proposed to involve the vELC N-terminal extension with the 8 nm step engaging the vELC/actin bond facilitating an extra ∼19 degrees of lever-arm rotation while the predominant 5 nm step forgoes vELC/actin binding. A minor 3 nm step is the unlikely conversion of the completed 5 to the 8 nm step. This hypothesis was tested using a 17 residue N-terminal truncated vELC in porcine ßmys (Δ17ßmys) and a 43 residue N-terminal truncated human vELC expressed in transgenic mouse heart (Δ43αmys). Step-size and step-frequency were measured using the Qdot motility assay. Both Δ17ßmys and Δ43αmys had significantly increased 5 nm step-frequency and coincident loss in the 8 nm step-frequency compared to native proteins suggesting the vELC/actin interaction drives step-size preference. Step-size and step-frequency probability densities depend on the relative fraction of truncated vELC and relate linearly to pure myosin species concentrations in a mixture containing native vELC homodimer, two truncated vELCs in the modified homodimer, and one native and one truncated vELC in the heterodimer. Step-size and step-frequency, measured for native homodimer and at two or more known relative fractions of truncated vELC, are surmised for each pure species by using a new analytical method.


Asunto(s)
Miosinas Cardíacas/metabolismo , Miocardio/metabolismo , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Miosinas Cardíacas/química , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Miocardio/química , Conformación Proteica , Multimerización de Proteína , Porcinos
13.
Arch Biochem Biophys ; 601: 121-32, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26906074

RESUMEN

Using microarray and bioinformatics, we examined the gene expression profiles in transgenic mouse hearts expressing mutations in the myosin regulatory light chain shown to cause hypertrophic cardiomyopathy (HCM). We focused on two malignant RLC-mutations, Arginine 58→Glutamine (R58Q) and Aspartic Acid 166 â†’ Valine (D166V), and one benign, Lysine 104 â†’ Glutamic Acid (K104E)-mutation. Datasets of differentially expressed genes for each of three mutants were compared to those observed in wild-type (WT) hearts. The changes in the mutant vs. WT samples were shown as fold-change (FC), with stringency FC ≥ 2. Based on the gene profiles, we have identified the major signaling pathways that underlie the R58Q-, D166V- and K104E-HCM phenotypes. The correlations between different genotypes were also studied using network-based algorithms. Genes with strong correlations were clustered into one group and the central gene networks were identified for each HCM mutant. The overall gene expression patterns in all mutants were distinct from the WT profiles. Both malignant mutations shared certain classes of genes that were up or downregulated, but most similarities were noted between D166V and K104E mice, with R58Q hearts showing a distinct gene expression pattern. Our data suggest that all three HCM mice lead to cardiomyopathy in a mutation-specific manner and thus develop HCM through diverse mechanisms.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Regulación de la Expresión Génica , Mutación , Cadenas Ligeras de Miosina/metabolismo , Algoritmos , Animales , Arginina/química , Biología Computacional , Perfilación de la Expresión Génica , Ácido Glutámico/química , Glutamina/química , Lisina/química , Ratones , Ratones Transgénicos , Familia de Multigenes , Miocardio/metabolismo , Cadenas Ligeras de Miosina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Análisis de Componente Principal , Valina/química
14.
Arch Biochem Biophys ; 580: 14-21, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26116789

RESUMEN

Familial hypertrophic cardiomyopathy (HCM) is characterized by left ventricular hypertrophy and myofibrillar disarray, and often results in sudden cardiac death. Two HCM mutations, N47K and R58Q, are located in the myosin regulatory light chain (RLC). The RLC mechanically stabilizes the myosin lever arm, which is crucial to myosin's ability to transmit contractile force. The N47K and R58Q mutations have previously been shown to reduce actin filament velocity under load, stemming from a more compliant lever arm (Greenberg, 2010). In contrast, RLC phosphorylation was shown to impart stiffness to the myosin lever arm (Greenberg, 2009). We hypothesized that phosphorylation of the mutant HCM-RLC may mitigate distinct mutation-induced structural and functional abnormalities. In vitro motility assays were utilized to investigate the effects of RLC phosphorylation on the HCM-RLC mutant phenotype in the presence of an α-actinin frictional load. Porcine cardiac ß-myosin was depleted of its native RLC and reconstituted with mutant or wild-type human RLC in phosphorylated or non-phosphorylated form. Consistent with previous findings, in the presence of load, myosin bearing the HCM mutations reduced actin sliding velocity compared to WT resulting in 31-41% reductions in force production. Myosin containing phosphorylated RLC (WT or mutant) increased sliding velocity and also restored mutant myosin force production to near WT unphosphorylated values. These results point to RLC phosphorylation as a general mechanism to increase force production of the individual myosin motor and as a potential target to ameliorate the HCM-induced phenotype at the molecular level.


Asunto(s)
Actinina/química , Actinas/química , Cadenas Ligeras de Miosina/química , Quinasa de Cadena Ligera de Miosina/química , Miosinas Ventriculares/química , Actinina/genética , Actinas/genética , Animales , Pollos , Expresión Génica , Ventrículos Cardíacos/química , Humanos , Cinética , Movimiento (Física) , Músculo Esquelético/química , Músculo Liso/química , Mutación , Cadenas Ligeras de Miosina/genética , Quinasa de Cadena Ligera de Miosina/genética , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Porcinos , Miosinas Ventriculares/genética
15.
J Muscle Res Cell Motil ; 36(6): 447-61, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26668058

RESUMEN

In this study we aimed to provide an in-depth proteomic analysis of differentially expressed proteins in the hearts of transgenic mouse models of pathological and physiological cardiac hypertrophy using tandem mass tag labeling and liquid chromatography tandem mass spectrometry. The Δ43 mouse model, expressing the 43-amino-acid N-terminally truncated myosin essential light chain (ELC) served as a tool to study the mechanisms of physiological cardiac remodeling, while the pathological hypertrophy was investigated in A57G (Alanine 57 â†’ Glycine) ELC mice. The results showed that 30 proteins were differentially expressed in Δ43 versus A57G hearts as determined by multiple pair comparisons of the mutant versus wild-type (WT) samples with P < 0.05. The A57G hearts showed differential expression of nine mitochondrial proteins involved in metabolic processes compared to four proteins for ∆43 hearts when both mutants were compared to WT hearts. Comparisons between ∆43 and A57G hearts showed an upregulation of three metabolically important mitochondrial proteins but downregulation of nine proteins in ∆43 hearts. The physiological model of cardiac hypertrophy (∆43) showed no changes in the levels of Ca(2+)-binding proteins relative to WT, while the pathologic model (A57G) showed the upregulation of three Ca(2+)-binding proteins, including sarcalumenin. Unique differences in chaperone and fatty acid metabolism proteins were also observed in Δ43 versus A57G hearts. The proteomics data support the results from functional studies performed previously on both animal models of cardiac hypertrophy and suggest that the A57G- and not ∆43- mediated alterations in fatty acid metabolism and Ca(2+) homeostasis may contribute to pathological cardiac remodeling in A57G hearts.


Asunto(s)
Corazón/fisiología , Mutación/genética , Miocardio/metabolismo , Cadenas Ligeras de Miosina/genética , Proteoma/metabolismo , Remodelación Ventricular/fisiología , Animales , Calcio/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Femenino , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Proteómica/métodos , Regulación hacia Arriba/fisiología , Remodelación Ventricular/genética
16.
J Mol Cell Cardiol ; 74: 318-29, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24992035

RESUMEN

We have examined, for the first time, the effects of the familial hypertrophic cardiomyopathy (HCM)-associated Lys104Glu mutation in the myosin regulatory light chain (RLC). Transgenic mice expressing the Lys104Glu substitution (Tg-MUT) were generated and the results were compared to Tg-WT (wild-type human ventricular RLC) mice. Echocardiography with pulse wave Doppler in 6month-old Tg-MUT showed early signs of diastolic disturbance with significantly reduced E/A transmitral velocities ratio. Invasive hemodynamics in 6month-old Tg-MUT mice also demonstrated a borderline significant prolonged isovolumic relaxation time (Tau) and a tendency for slower rate of pressure decline, suggesting alterations in diastolic function in Tg-MUT. Six month-old mutant animals had no LV hypertrophy; however, at >13months they displayed significant hypertrophy and fibrosis. In skinned papillary muscles from 5 to 6month-old mice a mutation induced reduction in maximal tension and slower muscle relaxation rates were observed. Mutated cross-bridges showed increased rates of binding to the thin filaments and a faster rate of the power stroke. In addition, ~2-fold lower level of RLC phosphorylation was observed in the mutant compared to Tg-WT. In line with the higher mitochondrial content seen in Tg-MUT hearts, the MUT-myosin ATPase activity was significantly higher than WT-myosin, indicating increased energy consumption. In the in vitro motility assay, MUT-myosin produced higher actin sliding velocity under zero load, but the velocity drastically decreased with applied load in the MUT vs. WT myosin. Our results suggest that diastolic disturbance (impaired muscle relaxation, lower E/A) and inefficiency of energy use (reduced contractile force and faster ATP consumption) may underlie the Lys104Glu-mediated HCM phenotype.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Mutación , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/genética , Músculos Papilares/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Diástole , Regulación de la Expresión Génica , Frecuencia Cardíaca , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Relajación Muscular , Contracción Miocárdica , Miocitos Cardíacos/patología , Cadenas Ligeras de Miosina/metabolismo , Músculos Papilares/diagnóstico por imagen , Músculos Papilares/patología , Cultivo Primario de Células , Transducción de Señal , Técnicas de Cultivo de Tejidos , Ultrasonografía Doppler de Pulso
17.
Artículo en Inglés | MEDLINE | ID: mdl-38494592

RESUMEN

We investigated the impact of the phosphomimetic (Ser15 → Asp15) myosin regulatory light chain (S15D-RLC) on the Super-Relaxed (SRX) state of myosin using previously characterized transgenic (Tg) S15D-D166V rescue mice, comparing them to the Hypertrophic Cardiomyopathy (HCM) Tg-D166V model and wild-type (WT) RLC mice. In the Tg-D166V model, we observed a disruption of the SRX state, resulting in a transition from SRX to DRX (Disordered Relaxed) state, which explains the hypercontractility of D166V-mutated myosin motors. The presence of the S15D moiety in Tg-S15D-D166V mice restored the SRX/DRX balance to levels comparable to Tg-WT, thus mitigating the hypercontractile behavior associated with the HCM-D166V mutation. Additionally, we investigated the impact of delivering the S15D-RLC molecule to the hearts of Tg-D166V mice via adeno-associated virus (AAV9) and compared their condition to AAV9-empty vector-injected or non-injected Tg-D166V animals. Tg-D166V mice injected with AAV9 S15D-RLC exhibited a significantly higher proportion of myosin heads in the SRX state compared to those injected with AAV9 empty vector or left non-injected. No significant effect was observed in Tg-WT hearts treated similarly. These findings suggest that AAV9-delivered phosphomimetic S15D-RLC modality mitigates the abnormal Tg-D166V phenotype without impacting the normal function of Tg-WT hearts. Global longitudinal strain analysis supported these observations, indicating that the S15D moiety can alleviate the HCM-D166V phenotype by restoring SRX stability and the SRX ↔ DRX equilibrium.

18.
Am J Physiol Heart Circ Physiol ; 304(2): H253-9, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23144314

RESUMEN

The role of cardiac myosin essential light chain (ELC) in the sarcomere length (SL) dependency of myofilament contractility is unknown. Therefore, mechanical and dynamic contractile properties were measured at SL 1.9 and 2.2 µm in cardiac muscle fibers from two groups of transgenic (Tg) mice: 1) Tg-wild-type (WT) mice that expressed WT human ventricular ELC and 2) Tg-Δ43 mice that expressed a mutant ELC lacking 1-43 amino acids. In agreement with previous studies, Ca(2+)-activated maximal tension decreased significantly in Tg-Δ43 fibers. pCa(50) (-log(10) [Ca(2+)](free) required for half maximal activation) values at SL of 1.9 µm were 5.64 ± 0.02 and 5.70 ± 0.02 in Tg-WT and Tg-Δ43 fibers, respectively. pCa(50) values at SL of 2.2 µm were 5.70 ± 0.01 and 5.71 ± 0.01 in Tg-WT and Tg-Δ43 fibers, respectively. The SL-mediated increase in the pCa(50) value was statistically significant only in Tg-WT fibers (P < 0.01), indicating that the SL dependency of myofilament Ca(2+) sensitivity was blunted in Tg-Δ43 fibers. The SL dependency of cross-bridge (XB) detachment kinetics was also blunted in Tg-Δ43 fibers because the decrease in XB detachment kinetics was significant (P < 0.001) only at SL 1.9 µm. Thus the increased XB dwell time at the short SL augments Ca(2+) sensitivity at short SL and thus blunts SL-mediated increase in myofilament Ca(2+) sensitivity. Our data suggest that the NH(2)-terminal extension of cardiac ELC not only augments the amplitude of force generation, but it also may play a role in mediating the SL dependency of XB detachment kinetics and myofilament Ca(2+) sensitivity.


Asunto(s)
Calcio/metabolismo , Acoplamiento Excitación-Contracción , Eliminación de Gen , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Miofibrillas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Secuencia de Aminoácidos , Animales , Fenómenos Biomecánicos , Femenino , Humanos , Cinética , Ratones , Ratones Transgénicos , Fuerza Muscular , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Sarcómeros/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 305(4): H575-89, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23748425

RESUMEN

The functional consequences of the familial hypertrophic cardiomyopathy A57G (alanine-to-glycine) mutation in the myosin ventricular essential light chain (ELC) were assessed in vitro and in vivo using previously generated transgenic (Tg) mice expressing A57G-ELC mutant vs. wild-type (WT) of human cardiac ELC and in recombinant A57G- or WT-protein-exchanged porcine cardiac muscle strips. Compared with the Tg-WT, there was a significant increase in the Ca²âº sensitivity of force (ΔpCa50 ≅ 0.1) and an ~1.3-fold decrease in maximal force per cross section of muscle observed in the mutant preparations. In addition, a significant increase in passive tension in response to stretch was monitored in Tg-A57G vs. Tg-WT strips indicating a mutation-induced myocardial stiffness. Consistently, the hearts of Tg-A57G mice demonstrated a high level of fibrosis and hypertrophy manifested by increased heart weight-to-body weight ratios and a decreased number of nuclei indicating an increase in the two-dimensional size of Tg-A57G vs. Tg-WT myocytes. Echocardiography examination showed a phenotype of eccentric hypertrophy in Tg-A57G mice, enhanced left ventricular (LV) cavity dimension without changes in LV posterior/anterior wall thickness. Invasive hemodynamics data revealed significantly increased end-systolic elastance, defined by the slope of the pressure-volume relationship, indicating a mutation-induced increase in cardiac contractility. Our results suggest that the A57G allele causes disease by means of a discrete modulation of myofilament function, increased Ca²âº sensitivity, and decreased maximal tension followed by compensatory hypertrophy and enhanced contractility. These and other contributing factors such as increased myocardial stiffness and fibrosis most likely activate cardiomyopathic signaling pathways leading to pathologic cardiac remodeling.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/metabolismo , Mutación , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Músculos Papilares/metabolismo , Animales , Fenómenos Biomecánicos , Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Modelos Animales de Enfermedad , Acoplamiento Excitación-Contracción , Fibrosis , Predisposición Genética a la Enfermedad , Hemodinámica , Humanos , Cinética , Ratones , Ratones Transgénicos , Contracción Miocárdica , Miofibrillas/metabolismo , Músculos Papilares/patología , Fenotipo , Fosforilación , Porcinos , Ultrasonografía , Función Ventricular Izquierda , Remodelación Ventricular
20.
Biochem J ; 442(1): 95-103, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22091967

RESUMEN

FHC (familial hypertrophic cardiomyopathy) is a heritable form of cardiac hypertrophy caused by mutations in genes encoding sarcomeric proteins. The present study focuses on the A13T mutation in the human ventricular myosin RLC (regulatory light chain) that is associated with a rare FHC variant defined by mid-ventricular obstruction and septal hypertrophy. We generated heart-specific Tg (transgenic) mice with ~10% of human A13T-RLC mutant replacing the endogenous mouse cardiac RLC. Histopathological examinations of longitudinal heart sections from Tg-A13T mice showed enlarged interventricular septa and profound fibrotic lesions compared with Tg-WT (wild-type), expressing the human ventricular RLC, or non-Tg mice. Functional studies revealed an abnormal A13T mutation-induced increase in isometric force production, no change in the force-pCa relationship and a decreased Vmax of the acto-myosin ATPase. In addition, a fluorescence-based assay showed a 3-fold lower binding affinity of the recombinant A13T mutant for the RLC-depleted porcine myosin compared with WT-RLC. These results suggest that the A13T mutation triggers a hypertrophic response through changes in cardiac sarcomere organization and myosin cross-bridge function leading to abnormal remodelling of the heart. The significant functional changes observed, despite a low level of A13T mutant incorporation into myofilaments, suggest a 'poison-peptide' mechanism of disease.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Contracción Miocárdica/fisiología , Cadenas Ligeras de Miosina/genética , Animales , Cardiomiopatía Hipertrófica Familiar/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Contracción Miocárdica/genética , Miocardio/patología , Miofibrillas/genética , Cadenas Ligeras de Miosina/fisiología , Músculos Papilares/patología , Conejos , Sarcómeros/ultraestructura , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA