Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
2.
Nature ; 623(7988): 863-871, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914933

RESUMEN

The thick filament is a key component of sarcomeres, the basic units of striated muscle1. Alterations in thick filament proteins are associated with familial hypertrophic cardiomyopathy and other heart and muscle diseases2. Despite the central importance of the thick filament, its molecular organization remains unclear. Here we present the molecular architecture of native cardiac sarcomeres in the relaxed state, determined by cryo-electron tomography. Our reconstruction of the thick filament reveals the three-dimensional organization of myosin, titin and myosin-binding protein C (MyBP-C). The arrangement of myosin molecules is dependent on their position along the filament, suggesting specialized capacities in terms of strain susceptibility and force generation. Three pairs of titin-α and titin-ß chains run axially along the filament, intertwining with myosin tails and probably orchestrating the length-dependent activation of the sarcomere. Notably, whereas the three titin-α chains run along the entire length of the thick filament, titin-ß chains do not. The structure also demonstrates that MyBP-C bridges thin and thick filaments, with its carboxy-terminal region binding to the myosin tails and directly stabilizing the OFF state of the myosin heads in an unforeseen manner. These results provide a foundation for future research investigating muscle disorders involving sarcomeric components.


Asunto(s)
Miosinas Cardíacas , Miocardio , Sarcómeros , Conectina/química , Conectina/metabolismo , Conectina/ultraestructura , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Miocardio/química , Miocardio/citología , Miocardio/ultraestructura , Sarcómeros/química , Sarcómeros/metabolismo , Sarcómeros/ultraestructura , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/ultraestructura
3.
Nature ; 623(7988): 853-862, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914935

RESUMEN

Pumping of the heart is powered by filaments of the motor protein myosin that pull on actin filaments to generate cardiac contraction. In addition to myosin, the filaments contain cardiac myosin-binding protein C (cMyBP-C), which modulates contractility in response to physiological stimuli, and titin, which functions as a scaffold for filament assembly1. Myosin, cMyBP-C and titin are all subject to mutation, which can lead to heart failure. Despite the central importance of cardiac myosin filaments to life, their molecular structure has remained a mystery for 60 years2. Here we solve the structure of the main (cMyBP-C-containing) region of the human cardiac filament using cryo-electron microscopy. The reconstruction reveals the architecture of titin and cMyBP-C and shows how myosin's motor domains (heads) form three different types of motif (providing functional flexibility), which interact with each other and with titin and cMyBP-C to dictate filament architecture and function. The packing of myosin tails in the filament backbone is also resolved. The structure suggests how cMyBP-C helps to generate the cardiac super-relaxed state3; how titin and cMyBP-C may contribute to length-dependent activation4; and how mutations in myosin and cMyBP-C might disturb interactions, causing disease5,6. The reconstruction resolves past uncertainties and integrates previous data on cardiac muscle structure and function. It provides a new paradigm for interpreting structural, physiological and clinical observations, and for the design of potential therapeutic drugs.


Asunto(s)
Miosinas Cardíacas , Microscopía por Crioelectrón , Miocardio , Humanos , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/ultraestructura , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas Portadoras/ultraestructura , Conectina/química , Conectina/metabolismo , Conectina/ultraestructura , Miocardio/química , Miocardio/ultraestructura
4.
PLoS Comput Biol ; 19(5): e1011099, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37200380

RESUMEN

The druggability of small-molecule binding sites can be significantly affected by protein motions and conformational changes. Ligand binding, protein dynamics and protein function have been shown to be closely interconnected in myosins. The breakthrough discovery of omecamtiv mecarbil (OM) has led to an increased interest in small molecules that can target myosin and modulate its function for therapeutic purposes (myosin modulators). In this work, we use a combination of computational methods, including steered molecular dynamics, umbrella sampling and binding pocket tracking tools, to follow the evolution of the OM binding site during the recovery stroke transition of human ß-cardiac myosin. We found that steering two internal coordinates of the motor domain can recapture the main features of the transition and in particular the rearrangements of the binding site, which shows significant changes in size, shape and composition. Possible intermediate conformations were also identified, in remarkable agreement with experimental findings. The differences in the binding site properties observed along the transition can be exploited for the future development of conformation-selective myosin modulators.


Asunto(s)
Miosinas Cardíacas , Miosinas Ventriculares , Humanos , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo , Corazón , Miocardio/metabolismo , Miosinas/química , Urea/metabolismo
5.
Biophys J ; 122(1): 54-62, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36451546

RESUMEN

The development of small molecule myosin modulators has seen an increased effort in recent years due to their possible use in the treatment of cardiac and skeletal myopathies. Omecamtiv mecarbil (OM) is the first-in-class cardiac myotrope and the first to enter clinical trials. Its selectivity toward slow/beta-cardiac myosin lies at the heart of its function; however, little is known about the underlying reasons for selectivity to this isoform as opposed to other closely related ones such as fast-type skeletal myosins. In this work, we compared the structure and dynamics of the OM binding site in cardiac and in fasttype IIa skeletal myosin to identify possible reasons for OM selectivity. We found that the different shape, size, and composition of the binding pocket in skeletal myosin directly affects the binding mode and related affinity of OM, which is potentially a result of weaker interactions and less optimal molecular recognition. Moreover, we identified a side pocket adjacent to the OM binding site that shows increased accessibility in skeletal myosin compared with the cardiac isoform. These findings could pave the way to the development of skeletal-selective compounds that can target this region of the protein and potentially be used to treat congenital myopathies where muscle weakness is related to myosin loss of function.


Asunto(s)
Corazón , Miosinas , Miosinas/metabolismo , Miocardio/metabolismo , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Dominios Proteicos , Urea/metabolismo
6.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35163146

RESUMEN

Two isoforms of human cardiac myosin, alpha and beta, share significant sequence similarities but show different kinetics. The alpha isoform is a faster motor; it spends less time being strongly bound to actin during the actomyosin cycle. With alpha isoform, actomyosin dissociates faster upon ATP binding, and the affinity of ADP to actomyosin is weaker. One can suggest that the isoform-specific actomyosin kinetics is regulated at the nucleotide binding site of human cardiac myosin. Myosin is a P-loop ATPase; the nucleotide-binding site consists of P-loop and loops switch 1 and 2. All three loops position MgATP for successful hydrolysis. Loops sequence is conserved in both myosin isoforms, and we hypothesize that the isoform-specific structural element near the active site regulates the rate of nucleotide binding and release. Previously we ran molecular dynamics simulations and found that loop S291-E317 near loop switch 1 is more compact and exhibits larger fluctuations of the position of amino acid residues in beta isoform than in alpha. In alpha isoform, the loop forms a salt bridge with loop switch 1, the bridge is not present in beta isoform. Two isoleucines I303 and I313 of loop S291-E317 are replaced with valines in alpha isoform. We introduced a double mutation I303V:I313V in beta isoform background and studied how the mutation affects the rate of ATP binding and ADP dissociation from actomyosin. We found that ATP-induced actomyosin dissociation occurs faster in the mutant, but the rate of ADP release remains the same as in the wild-type beta isoform. Due to the proximity of loop S291-E317 and loop switch 1, a faster rate of ATP-induced actomyosin dissociation indicates that loop S291-E317 affects structural dynamics of loop switch 1, and that loop switch 1 controls ATP binding to the active site. A similar rate of ADP dissociation from actomyosin in the mutant and wild-type myosin constructs indicates that loop switch 1 does not control ADP release from actomyosin.


Asunto(s)
Actomiosina/química , Actomiosina/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Sitios de Unión , Humanos , Cinética , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
7.
Eur J Med Genet ; 64(11): 104314, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34481090

RESUMEN

OBJECTIVE: Atrial septal defect, secundum (ASD Ⅱ, OMIM: 603642) is the second common congenital heart defect (CHD) in China. However, the genetic etiology of familial ASD II remains elusive. METHODS AND RESULTS: Using whole-exome sequencing (WES) and Sanger sequencing, we identified a novel myosin heavy chain 6 (MYH6) gene insertion variation, NM_002471.3: c.5465_5470dup (Arg1822_Glu1823dup), in a large Chinese Han family with ASD II. The variant Arg1822_Glu1823dup co-segregated with the disease in this family with autosomal dominant inheritance. The insertion variant located in the coiled-coil domain of the MYH6 protein, which is highly conserved across homologous myosin proteins and species. In transfected myoblast C2C12 cell lines, the MYH6 Arg1822_Glu1823dup variant significantly impaired myofibril formation and increased apoptosis but did not significantly reduce cell viability. Furthermore, molecular simulations revealed that the Arg1822_Glu1823dup variant impaired the myosin α-helix, increasing the stability of the coiled-coil myosin dimer, suggesting that this variant has an effect on the coiled-coil domain self-aggregation. These findings indicate that Arg1822_Glu1823dup variant plays a crucial role in the pathogenesis of ASD II. CONCLUSION: Our findings expand the spectrum of MYH6 variations associated with familial ASD II and may provide a molecular basis in genetic counseling and prenatal diagnosis for this Chinses family.


Asunto(s)
Miosinas Cardíacas/genética , Defectos del Tabique Interatrial/genética , Mutagénesis Insercional , Cadenas Pesadas de Miosina/genética , Adulto , Animales , Apoptosis , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Línea Celular , Supervivencia Celular , Niño , Femenino , Defectos del Tabique Interatrial/metabolismo , Defectos del Tabique Interatrial/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mioblastos/metabolismo , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/metabolismo , Linaje , Conformación Proteica en Hélice alfa , Estabilidad Proteica
8.
J Med Microbiol ; 70(5)2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33956590

RESUMEN

Introduction. Group A streptococci can trigger autoimmune responses that lead to acute rheumatic fever (ARF) and rheumatic heart disease (RHD).Gap Statement. Some autoantibodies generated in ARF/RHD target antigens in the S2 subfragment region of cardiac myosin. However, little is known about the kinetics of these antibodies during the disease process.Aim. To determine the antibody responses over time in patients and healthy controls against host tissue proteins - cardiac myosin and peptides from its S2 subfragment, tropomyosin, laminin and keratin.Methodology. We used enzyme-linked immunosorbent assays (ELISA) to determine antibody responses in: (1) healthy controls; (2) patients with streptococcal pharyngitis; (3) patients with ARF with carditis and (4) patients with RHD on penicillin prophylaxis.Results. We observed significantly higher antibody responses against extracellular proteins - laminin and keratin in pharyngitis group, patients with ARF and patients with RHD when compared to healthy controls. The antibody responses against intracellular proteins - cardiac myosin and tropomyosin were elevated only in the group of patients with ARF with active carditis. While the reactivity to S2 peptides S2-1-3, 8-11, 14, 16-18, 21-22 and 32 was higher in patients with ARF, the reactivity in the RHD group was high only against S2-1, 9, 11, 12 when compared to healthy controls. The reactivity against S2 peptides reduced as the disease condition stabilized in the ARF group whereas the reactivity remained unaltered in the RHD group. By contrast antibodies against laminin and keratin persisted in patients with RHD.Conclusion. Our findings of antibody responses against host proteins support the multistep hypothesis in the development of rheumatic carditis. The differential kinetics of serum antibody responses against S2 peptides may have potential use as markers of ongoing cardiac damage that can be used to monitor patients with ARF/RHD.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Fiebre Reumática/inmunología , Cardiopatía Reumática/inmunología , Autoanticuerpos/sangre , Autoantígenos/química , Miosinas Cardíacas/química , Miosinas Cardíacas/inmunología , Humanos , Queratinas/inmunología , Laminina/inmunología , Estudios Longitudinales , Péptidos/química , Péptidos/inmunología , Fiebre Reumática/sangre , Cardiopatía Reumática/sangre , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Tropomiosina/inmunología
9.
ACS Infect Dis ; 7(6): 1483-1502, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34019767

RESUMEN

Viral proteases are highly specific and recognize conserved cleavage site sequences of ∼6-8 amino acids. Short stretches of homologous host-pathogen sequences (SSHHPS) can be found spanning the viral protease cleavage sites. We hypothesized that these sequences corresponded to specific host protein targets since >40 host proteins have been shown to be cleaved by Group IV viral proteases and one Group VI viral protease. Using PHI-BLAST and the viral protease cleavage site sequences, we searched the human proteome for host targets and analyzed the hit results. Although the polyprotein and host proteins related to the suppression of the innate immune responses may be the primary targets of these viral proteases, we identified other cleavable host proteins. These proteins appear to be related to the virus-induced phenotype associated with Group IV viruses, suggesting that information about viral pathogenesis may be extractable directly from the viral genome sequence. Here we identify sequences cleaved by the SARS-CoV-2 papain-like protease (PLpro) in vitro within human MYH7 and MYH6 (two cardiac myosins linked to several cardiomyopathies), FOXP3 (an X-linked Treg cell transcription factor), ErbB4 (HER4), and vitamin-K-dependent plasma protein S (PROS1), an anticoagulation protein that prevents blood clots. Zinc inhibited the cleavage of these host sequences in vitro. Other patterns emerged from multispecies sequence alignments of the cleavage sites, which may have implications for the selection of animal models and zoonosis. SSHHPS/nsP is an example of a sequence-specific post-translational silencing mechanism.


Asunto(s)
Papaína , Péptido Hidrolasas , SARS-CoV-2/enzimología , Proteasas Virales/metabolismo , Secuencia de Aminoácidos , Miosinas Cardíacas/química , Factores de Transcripción Forkhead/química , Humanos , Cadenas Pesadas de Miosina/química , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , Proteína S/química , Receptor ErbB-4/química
10.
J Biol Chem ; 296: 100694, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33895132

RESUMEN

Myosin heavy chain 7b (MYH7b) is an ancient member of the myosin heavy chain motor protein family that is expressed in striated muscles. In mammalian cardiac muscle, MYH7b RNA is expressed along with two other myosin heavy chains, ß-myosin heavy chain (ß-MyHC) and α-myosin heavy chain (α-MyHC). However, unlike ß-MyHC and α-MyHC, which are maintained in a careful balance at the protein level, the MYH7b locus does not produce a full-length protein in the heart due to a posttranscriptional exon-skipping mechanism that occurs in a tissue-specific manner. Whether this locus has a role in the heart beyond producing its intronic microRNA, miR-499, was unclear. Using cardiomyocytes derived from human induced pluripotent stem cells as a model system, we found that the noncoding exon-skipped RNA (lncMYH7b) affects the transcriptional landscape of human cardiomyocytes, independent of miR-499. Specifically, lncMYH7b regulates the ratio of ß-MyHC to α-MyHC, which is crucial for cardiac contractility. We also found that lncMYH7b regulates beat rate and sarcomere formation in cardiomyocytes. This regulation is likely achieved through control of a member of the TEA domain transcription factor family (TEAD3, which is known to regulate ß-MyHC). Therefore, we conclude that this ancient gene has been repurposed by alternative splicing to produce a regulatory long-noncoding RNA in the human heart that affects cardiac myosin composition.


Asunto(s)
Miosinas Cardíacas/metabolismo , Miocardio/metabolismo , Cadenas Pesadas de Miosina/metabolismo , ARN Largo no Codificante/genética , Miosinas Cardíacas/química , Humanos , Células Madre Pluripotentes Inducidas , MicroARNs/genética , Simulación de Dinámica Molecular , Miocardio/citología , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/química , Conformación Proteica
11.
Genetics ; 216(1): 177-190, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32753389

RESUMEN

Myosin is an essential motor protein, which in muscle is comprised of two molecules each of myosin heavy-chain (MHC), the essential or alkali myosin light-chain 1 (MLC1), and the regulatory myosin light-chain 2 (MLC2). It has been shown previously that MLC2 phosphorylation at two canonical serine residues is essential for proper flight muscle function in Drosophila; however, MLC2 is also phosphorylated at additional residues for which the mechanism and functional significance is not known. We found that a hypomorphic allele of Pkcδ causes a flightless phenotype; therefore, we hypothesized that PKCδ phosphorylates MLC2. We rescued flight disability by duplication of the wild-type Pkcδ gene. Moreover, MLC2 is hypophosphorylated in Pkcδ mutant flies, but it is phosphorylated in rescued animals. Myosin isolated from Pkcδ mutant flies shows a reduced actin-activated ATPase activity, and MLC2 in these myosin preparations can be phosphorylated directly by recombinant human PKCδ. The flightless phenotype is characterized by a shortened and disorganized sarcomere phenotype that becomes apparent following eclosion. We conclude that MLC2 is a direct target of phosphorylation by PKCδ, and that this modification is necessary for flight muscle maturation and function.


Asunto(s)
Miosinas Cardíacas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Drosophila melanogaster , Humanos , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , Sarcómeros/metabolismo
12.
Mol Med Rep ; 20(6): 5229-5238, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31638223

RESUMEN

ß­myosin heavy chain (MHC) 7 (MYH7) is the dominant pathogenic gene that harbors mutations in 20­30% of cases of familial hypertrophic cardiomyopathy (HCM). The aim of this study was to elucidate the distribution and type of genetic variations among Chinese HCM families. From 2013 to 2017, the clinical data of 387 HCM probands and their families were collected. Targeted exome­sequencing technology was used in all probands, and the selected mutations were subsequently verified by Sanger sequencing in the probands, family members and 300 healthy ethnic­matched volunteers. Three­dimensional models were created using Swiss­PdbViewer 4.1, and further genetic analyses were performed to determine sequence conservation and frequency of the mutations. Among the 5 probands with double MYH7 mutations, 4 carried compound heterozygous mutations, and 1 carried monoallelic double mutations (A934V and E1387K). Four family members of the proband with monoallelic double mutations had the same mutation as the proband. Echocardiography and 12­lead electrocardiography revealed abnormalities in the proband and 3 of the 4 carriers. The probands with compound heterozygous mutation had a higher left ventricular mass as revealed by echocardiography and higher QRS, SV1 and RV5+SV1 amplitudes than those with monoallelic double mutations (P<0.05). Simulation of the 3D structure of mutated proteins showed that the replacement of alanine by valine affected the flexibility of the MHC neck domain in case of the A934V mutation, whereas reactivity of the MHC rod domain was affected in the case of the E1387K mutation. In conclusion, we identified several novel HCM­causing MYH7 mutations. More importantly, this is the first study to report a rare HCM family with monoallelic double mutations.


Asunto(s)
Alelos , Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Cadenas Pesadas de Miosina/genética , Adolescente , Adulto , Sustitución de Aminoácidos , Miosinas Cardíacas/química , Cardiomiopatía Hipertrófica Familiar/mortalidad , Niño , Preescolar , Toma de Decisiones Clínicas , Análisis Mutacional de ADN , Manejo de la Enfermedad , Ecocardiografía , Electrocardiografía , Femenino , Pruebas Genéticas , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Cadenas Pesadas de Miosina/química , Linaje , Pronóstico , Conformación Proteica , Adulto Joven
13.
Sci Rep ; 9(1): 12580, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467300

RESUMEN

Cardiac contractility is enhanced by phosphorylation of myosin light chain 2 (MLC2) by cardiac-specific MLC kinase (cMLCK), located at the neck region of myosin heavy chain. In normal mouse and human hearts, the level of phosphorylation is maintained relatively constant, at around 30-40% of total MLC2, likely by well-balanced phosphorylation and phosphatase-dependent dephosphorylation. Overexpression of cMLCK promotes sarcomere organization, while the loss of cMLCK leads to cardiac atrophy in vitro and in vivo. In this study, we showed that cMLCK is predominantly expressed at the Z-disc with additional diffuse cytosolic expression in normal adult mouse and human hearts. cMLCK interacts with the Z-disc protein, α-actinin2, with a high-affinity kinetic value of 13.4 ± 0.1 nM through the N-terminus region of cMLCK unique to cardiac-isoform. cMLCK mutant deficient for interacting with α-actinin2 did not promote sarcomeric organization and reduced cardiomyocyte cell size. In contrast, a cMLCK kinase-deficient mutant showed effects similar to wild-type cMLCK on sarcomeric organization and cardiomyocyte cell size. Our results suggest that cMLCK plays a role in sarcomere organization, likely distinct from its role in phosphorylating MLC2, both of which will contribute to the enhancement of cardiac contractility.


Asunto(s)
Actinina/metabolismo , Miosinas Cardíacas/metabolismo , Miocardio/enzimología , Cadenas Ligeras de Miosina/metabolismo , Adulto , Animales , Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Humanos , Recién Nacido , Ratones , Mutación , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , Especificidad por Sustrato
14.
Open Biol ; 8(11)2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30463911

RESUMEN

Cardiac ventricular myosin (ßmys) translates actin by transducing ATP free energy into mechanical work during muscle contraction. Unitary ßmys translation of actin is the step-size. In vitro and in vivo ßmys regulates contractile force and velocity autonomously by remixing three different step-sizes with adaptive stepping frequencies. Cardiac and skeletal actin isoforms have a specific 1 : 4 stoichiometry in normal adult human ventriculum. Human adults with inheritable hypertrophic cardiomyopathy (HCM) upregulate skeletal actin in ventriculum probably compensating the diseased muscle's inability to meet demand by adjusting ßmys force-velocity characteristics. ßmys force-velocity characteristics were compared for skeletal versus cardiac actin substrates using ensemble in vitro motility and single myosin assays. Two competing myosin strain-sensitive mechanisms regulate step-size choices dividing single ßmys mechanics into low- and high-force regimes. The actin isoforms alter myosin strain-sensitive regulation such that onset of the high-force regime, where a short step-size is a large or major contributor, is offset to higher loads probably by the unique cardiac essential light chain (ELC) N-terminus/cardiac actin contact at Glu6/Ser358. It modifies ßmys force-velocity by stabilizing the ELC N-terminus/cardiac actin association. Uneven onset of the high-force regime for skeletal versus cardiac actin modulates force-velocity characteristics as skeletal/cardiac actin fractional content increases in diseased muscle.


Asunto(s)
Actinas/química , Miosinas Cardíacas/química , Miosinas del Músculo Esquelético/química , Actinas/metabolismo , Animales , Miosinas Cardíacas/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Conejos , Miosinas del Músculo Esquelético/metabolismo
15.
Nat Commun ; 9(1): 4019, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30275503

RESUMEN

Hypertrophic cardiomyopathies (HCM) result from distinct single-point mutations in sarcomeric proteins that lead to muscle hypercontractility. While different models account for a pathological increase in the power output, clear understanding of the molecular basis of dysfunction in HCM is the mandatory next step to improve current treatments. Here, we present an optimized quasi-atomic model of the sequestered state of cardiac myosin coupled to X-ray crystallography and in silico analysis of the mechanical compliance of the lever arm, allowing the systematic study of a large set of HCM mutations and the definition of different mutation classes based on their effects on lever arm compliance, sequestered state stability, and motor functions. The present work reconciles previous models and explains how distinct HCM mutations can have disparate effects on the motor mechano-chemical parameters and yet lead to the same disease. The framework presented here can guide future investigations aiming at finding HCM treatments.


Asunto(s)
Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Animales , Miosinas Cardíacas/química , Cardiomiopatía Hipertrófica/genética , Bovinos , Simulación por Computador , Cristalografía por Rayos X , Modelos Cardiovasculares , Modelos Moleculares , Mutación , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Conformación Proteica , Mapeo de Interacción de Proteínas , Sarcómeros/genética , Sarcómeros/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(32): E7486-E7494, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30018063

RESUMEN

We used transient biochemical and structural kinetics to elucidate the molecular mechanism of mavacamten, an allosteric cardiac myosin inhibitor and a prospective treatment for hypertrophic cardiomyopathy. We find that mavacamten stabilizes an autoinhibited state of two-headed cardiac myosin not found in the single-headed S1 myosin motor fragment. We determined this by measuring cardiac myosin actin-activated and actin-independent ATPase and single-ATP turnover kinetics. A two-headed myosin fragment exhibits distinct autoinhibited ATP turnover kinetics compared with a single-headed fragment. Mavacamten enhanced this autoinhibition. It also enhanced autoinhibition of ADP release. Furthermore, actin changes the structure of the autoinhibited state by forcing myosin lever-arm rotation. Mavacamten slows this rotation in two-headed myosin but does not prevent it. We conclude that cardiac myosin is regulated in solution by an interaction between its two heads and propose that mavacamten stabilizes this state.


Asunto(s)
Actinas/metabolismo , Bencilaminas/farmacología , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica Familiar/tratamiento farmacológico , Subfragmentos de Miosina/metabolismo , Uracilo/análogos & derivados , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Regulación Alostérica/efectos de los fármacos , Bencilaminas/uso terapéutico , Miosinas Cardíacas/química , Cardiomiopatía Hipertrófica Familiar/etiología , Humanos , Cinética , Subfragmentos de Miosina/química , Estabilidad Proteica/efectos de los fármacos , Uracilo/farmacología , Uracilo/uso terapéutico
17.
BMC Cardiovasc Disord ; 18(1): 137, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29969989

RESUMEN

BACKGROUND: Myosin VI, encoded by MYH6, is expressed dominantly in human cardiac atria and plays consequential roles in cardiac muscle contraction and comprising the cardiac muscle thick filament. It has been reported that the mutations in the MYH6 gene associated with sinus venosus atrial septal defect (ASD type III), hypertrophic (HCM) and dilated (DCM) cardiomyopathies. METHODS: Two patients in an Iranian family have been identified who affected to Congenital Heart Disease (CHD). The male patient, besides CHD, shows that the thyroglossal sinus, refractive errors of the eye and mitral stenosis. The first symptoms emerged at the birth and diagnosis based on clinical features was made at about 5 years. The family had a history of ASD. For recognizing mutated gene (s), whole exome sequencing (WES) was performed for the male patient and variants were analyzed by autosomal dominant inheritance mode. RESULTS: Eventually, by several filtering processes, a mutation in MYH6 gene (NM_002471.3), c.3835C > T; R1279X, was identified as the most likely disease-susceptibility variant and then confirmed by Sanger sequencing in the family. The mutation frequency was checked out in the local databases. This mutation results in the elimination of the 660 amino acids in the C-terminal of Myosin VI protein, including the vital parts of the coiled-coil structure of the tail domain. CONCLUSIONS: Our study represents the first case of Sinus venosus defect caused directly by MYH6 stop codon mutation. Our data indicate that by increase haploinsufficiency of myosin VI, c.3835C > T mutation with reduced penetrance could be associated with CHD.


Asunto(s)
Miosinas Cardíacas/genética , Codón sin Sentido , Análisis Mutacional de ADN/métodos , Secuenciación del Exoma/métodos , Defectos del Tabique Interatrial/genética , Cadenas Pesadas de Miosina/genética , Adulto , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Niño , Codón de Terminación , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Defectos del Tabique Interatrial/diagnóstico , Herencia , Humanos , Irán , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/metabolismo , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Conformación Proteica , Factores de Riesgo , Relación Estructura-Actividad
18.
J Biol Chem ; 293(23): 9017-9029, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29666183

RESUMEN

Dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) can cause arrhythmias, heart failure, and cardiac death. Here, we functionally characterized the motor domains of five DCM-causing mutations in human ß-cardiac myosin. Kinetic analyses of the individual events in the ATPase cycle revealed that each mutation alters different steps in this cycle. For example, different mutations gave enhanced or reduced rate constants of ATP binding, ATP hydrolysis, or ADP release or exhibited altered ATP, ADP, or actin affinity. Local effects dominated, no common pattern accounted for the similar mutant phenotype, and there was no distinct set of changes that distinguished DCM mutations from previously analyzed HCM myosin mutations. That said, using our data to model the complete ATPase contraction cycle revealed additional critical insights. Four of the DCM mutations lowered the duty ratio (the ATPase cycle portion when myosin strongly binds actin) because of reduced occupancy of the force-holding A·M·D complex in the steady state. Under load, the A·M·D state is predicted to increase owing to a reduced rate constant for ADP release, and this effect was blunted for all five DCM mutations. We observed the opposite effects for two HCM mutations, namely R403Q and R453C. Moreover, the analysis predicted more economical use of ATP by the DCM mutants than by WT and the HCM mutants. Our findings indicate that DCM mutants have a deficit in force generation and force-holding capacity due to the reduced occupancy of the force-holding state.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Dilatada/genética , Cadenas Pesadas de Miosina/genética , Mutación Puntual , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Cardiomiopatía Dilatada/metabolismo , Línea Celular , Humanos , Cinética , Ratones , Modelos Moleculares , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/metabolismo , Dominios Proteicos
19.
J Mol Biol ; 430(10): 1459-1478, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29660325

RESUMEN

Over 20 mutations in ß-cardiac myosin heavy chain (ß-MHC), expressed in cardiac and slow muscle fibers, cause Laing early-onset distal myopathy (MPD-1), a skeletal muscle myopathy. Most of these mutations are in the coiled-coil tail and commonly involve a mutation to a proline or a single-residue deletion, both of which are predicted to strongly affect the secondary structure of the coiled coil. To test this, we characterized the effects of two MPD-1 causing mutations: A1603P and K1617del in vitro and in cells. Both mutations affected secondary structure, decreasing the helical content of 15 heptad and light meromyosin constructs. Both mutations also severely disrupted the ability of glutathione S-transferase-light meromyosin fusion proteins to form minifilaments in vitro, as demonstrated by negative stain electron microscopy. Mutant eGFP-tagged ß-MHC accumulated abnormally into the M-line of sarcomeres in cultured skeletal muscle myotubes. Incorporation of eGFP-tagged ß-MHC into sarcomeres in adult rat cardiomyocytes was reduced. Molecular dynamics simulations using a composite structure of part of the coiled coil demonstrated that both mutations affected the structure, with the mutation to proline (A1603P) having a smaller effect compared to K1617del. Taken together, it seems likely that the MPD-1 mutations destabilize the coiled coil, resulting in aberrant myosin packing in thick filaments in muscle sarcomeres, providing a potential mechanism for the disease.


Asunto(s)
Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Miopatías Distales/genética , Fibras Musculares Esqueléticas/citología , Mutación , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Animales , Miosinas Cardíacas/metabolismo , Línea Celular , Técnicas In Vitro , Ratones , Microscopía Electrónica , Simulación de Dinámica Molecular , Fibras Musculares Esqueléticas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Estructura Secundaria de Proteína , Ratas , Sarcómeros/química , Sarcómeros/metabolismo
20.
Open Biol ; 8(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29669825

RESUMEN

Myosin transduces ATP free energy into mechanical work in muscle. Cardiac muscle has dynamically wide-ranging power demands on the motor as the muscle changes modes in a heartbeat from relaxation, via auxotonic shortening, to isometric contraction. The cardiac power output modulation mechanism is explored in vitro by assessing single cardiac myosin step-size selection versus load. Transgenic mice express human ventricular essential light chain (ELC) in wild- type (WT), or hypertrophic cardiomyopathy-linked mutant forms, A57G or E143K, in a background of mouse α-cardiac myosin heavy chain. Ensemble motility and single myosin mechanical characteristics are consistent with an A57G that impairs ELC N-terminus actin binding and an E143K that impairs lever-arm stability, while both species down-shift average step-size with increasing load. Cardiac myosin in vivo down-shifts velocity/force ratio with increasing load by changed unitary step-size selections. Here, the loaded in vitro single myosin assay indicates quantitative complementarity with the in vivo mechanism. Both have two embedded regulatory transitions, one inhibiting ADP release and a second novel mechanism inhibiting actin detachment via strain on the actin-bound ELC N-terminus. Competing regulators filter unitary step-size selection to control force-velocity modulation without myosin integration into muscle. Cardiac myosin is muscle in a molecule.


Asunto(s)
Miosinas Cardíacas/fisiología , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiología , Animales , Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Miosinas Ventriculares/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...