Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 184(8): 2135-2150.e13, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33765442

RESUMO

Sarcomeres are force-generating and load-bearing devices of muscles. A precise molecular picture of how sarcomeres are built underpins understanding their role in health and disease. Here, we determine the molecular architecture of native vertebrate skeletal sarcomeres by electron cryo-tomography. Our reconstruction reveals molecular details of the three-dimensional organization and interaction of actin and myosin in the A-band, I-band, and Z-disc and demonstrates that α-actinin cross-links antiparallel actin filaments by forming doublets with 6-nm spacing. Structures of myosin, tropomyosin, and actin at ~10 Å further reveal two conformations of the "double-head" myosin, where the flexible orientation of the lever arm and light chains enable myosin not only to interact with the same actin filament, but also to split between two actin filaments. Our results provide unexpected insights into the fundamental organization of vertebrate skeletal muscle and serve as a strong foundation for future investigations of muscle diseases.


Assuntos
Músculo Esquelético/metabolismo , Sarcômeros/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinina/química , Actinina/metabolismo , Actomiosina/química , Actomiosina/metabolismo , Animais , Microscopia Crioeletrônica , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Ligação Proteica , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Tropomiosina/química , Tropomiosina/metabolismo
2.
Nature ; 623(7988): 863-871, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37914933

RESUMO

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.


Assuntos
Miosinas Cardíacas , Miocárdio , Sarcômeros , Conectina/química , Conectina/metabolismo , Conectina/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Miocárdio/química , Miocárdio/citologia , Miocárdio/ultraestrutura , Sarcômeros/química , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/ultraestrutura
3.
J Biol Chem ; 300(1): 105511, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042491

RESUMO

Myosin binding protein-C (MyBP-C) is a multidomain protein that regulates muscle contraction. Mutations in MYBPC3, the gene encoding for the cardiac variant (henceforth called cMyBP-C), are amongst the most frequent causes of hypertrophic cardiomyopathy. Most mutations lead to a truncated version of cMyBP-C, which is most likely unstable. However, missense mutations have also been reported, which tend to cluster in the central domains of the cMyBP-C molecule. This suggests that these central domains are more than just a passive spacer between the better characterized N- and C-terminal domains. Here, we investigated the potential impact of four different missense mutations, E542Q, G596R, N755K, and R820Q, which are spread over the domains C3 to C6, on the function of MyBP-C on both the isolated protein level and in cardiomyocytes in vitro. Effect on domain stability, interaction with thin filaments, binding to myosin, and subcellular localization behavior were assessed. Our studies show that these missense mutations result in slightly different phenotypes at the molecular level, which are mutation specific. The expected functional readout of each mutation provides a valid explanation for why cMyBP-C fails to work as a brake in the regulation of muscle contraction, which eventually results in a hypertrophic cardiomyopathy phenotype. We conclude that missense mutations in cMyBP-C must be evaluated in context of their domain localization, their effect on interaction with thin filaments and myosin, and their effect on protein stability to explain how they lead to disease.


Assuntos
Cardiomiopatia Hipertrófica , Proteínas de Transporte , Mutação de Sentido Incorreto , Humanos , Cardiomiopatia Hipertrófica/genética , Proteínas de Transporte/genética , Domínios Proteicos/genética , Estabilidade Proteica
4.
Acta Neuropathol ; 141(3): 431-453, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33449170

RESUMO

Mutations in the sarcomeric protein titin, encoded by TTN, are emerging as a common cause of myopathies. The diagnosis of a TTN-related myopathy is, however, often not straightforward due to clinico-pathological overlap with other myopathies and the prevalence of TTN variants in control populations. Here, we present a combined clinico-pathological, genetic and biophysical approach to the diagnosis of TTN-related myopathies and the pathogenicity ascertainment of TTN missense variants. We identified 30 patients with a primary TTN-related congenital myopathy (CM) and two truncating variants, or one truncating and one missense TTN variant, or homozygous for one TTN missense variant. We found that TTN-related myopathies show considerable overlap with other myopathies but are strongly suggested by a combination of certain clinico-pathological features. Presentation was typically at birth with the clinical course characterized by variable progression of weakness, contractures, scoliosis and respiratory symptoms but sparing of extraocular muscles. Cardiac involvement depended on the variant position. Our biophysical analyses demonstrated that missense mutations associated with CMs are strongly destabilizing and exert their effect when expressed on a truncating background or in homozygosity. We hypothesise that destabilizing TTN missense mutations phenocopy truncating variants and are a key pathogenic feature of recessive titinopathies that might be amenable to therapeutic intervention.


Assuntos
Conectina/genética , Miotonia Congênita/diagnóstico , Miotonia Congênita/genética , Miotonia Congênita/patologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Adulto Jovem
5.
Hum Mol Genet ; 23(4): 980-91, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24105469

RESUMO

Core myopathies (CM), the main non-dystrophic myopathies in childhood, remain genetically unexplained in many cases. Heart disease is not considered part of the typical CM spectrum. No congenital heart defect has been reported, and childhood-onset cardiomyopathy has been documented in only two CM families with homozygous mutations of the TTN gene. TTN encodes titin, a giant protein of striated muscles. Recently, heterozygous TTN truncating mutations have also been reported as a major cause of dominant dilated cardiomyopathy. However, relatively few TTN mutations and phenotypes are known, and titin pathophysiological role in cardiac and skeletal muscle conditions is incompletely understood. We analyzed a series of 23 families with congenital CM and primary heart disease using TTN M-line-targeted sequencing followed in selected patients by whole-exome sequencing and functional studies. We identified seven novel homozygous or compound heterozygous TTN mutations (five in the M-line, five truncating) in 17% patients. Heterozygous parents were healthy. Phenotype analysis identified four novel titinopathies, including cardiac septal defects, left ventricular non-compaction, Emery-Dreifuss muscular dystrophy or arthrogryposis. Additionally, in vitro studies documented the first-reported absence of a functional titin kinase domain in humans, leading to a severe antenatal phenotype. We establish that CM are associated with a large range of heart conditions of which TTN mutations are a major cause, thereby expanding the TTN mutational and phenotypic spectrum. Additionally, our results suggest titin kinase implication in cardiac morphogenesis and demonstrate that heterozygous TTN truncating mutations may not manifest unless associated with a second mutation, reassessing the paradigm of their dominant expression.


Assuntos
Códon sem Sentido , Conectina/genética , Cardiopatias/genética , Miopatia da Parte Central/genética , Adolescente , Conectina/metabolismo , Consanguinidade , Feminino , Genes Recessivos , Estudos de Associação Genética , Predisposição Genética para Doença , Cardiopatias/metabolismo , Cardiopatias/patologia , Heterozigoto , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miopatia da Parte Central/metabolismo , Miopatia da Parte Central/patologia , Linhagem , Fenótipo , Adulto Jovem
6.
PLoS One ; 18(4): e0284453, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37079638

RESUMO

Obscurin is a giant muscle protein (>800 kDa) featuring multiple signalling domains, including an SH3-DH-PH domain triplet from the Trio-subfamily of guanosine nucleotide exchange factors (GEFs). While previous research suggests that these domains can activate the small GTPases RhoA and RhoQ in cells, in vitro characterization of these interactions using biophysical techniques has been hampered by the intrinsic instability of obscurin GEF domains. To study substrate specificity, mechanism and regulation of obscurin GEF function by individual domains, we successfully optimized recombinant production of obscurin GEF domains and found that MST-family kinases phosphorylate the obscurin DH domain at Thr5798. Despite extensive testing of multiple GEF domain fragments, we did not detect any nucleotide exchange activity in vitro against 9 representative small GTPases. Bioinformatic analyses show that obscurin differs from other Trio-subfamily GEFs in several important aspects. While further research is necessary to evaluate obscurin GEF activity in vivo, our results indicate that obscurin has atypical GEF domains that, if catalytically active at all, are subject to complex regulation.


Assuntos
Nucleotídeos , Proteínas rho de Ligação ao GTP , Proteínas rho de Ligação ao GTP/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Transdução de Sinais , Proteínas Musculares
7.
Science ; 375(6582): eabn1934, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35175800

RESUMO

In skeletal muscle, nebulin stabilizes and regulates the length of thin filaments, but the underlying mechanism remains nebulous. In this work, we used cryo-electron tomography and subtomogram averaging to reveal structures of native nebulin bound to thin filaments within intact sarcomeres. This in situ reconstruction provided high-resolution details of the interaction between nebulin and actin, demonstrating the stabilizing role of nebulin. Myosin bound to the thin filaments exhibited different conformations of the neck domain, highlighting its inherent structural variability in muscle. Unexpectedly, nebulin did not interact with myosin or tropomyosin, but it did interact with a troponin T linker through two potential binding motifs on nebulin, explaining its regulatory role. Our structures support the role of nebulin as a thin filament "molecular ruler" and provide a molecular basis for studying nemaline myopathies.


Assuntos
Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Miofibrilas/ultraestrutura , Actinas/química , Actinas/metabolismo , Animais , Tomografia com Microscopia Eletrônica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Proteínas Musculares/genética , Mutação , Miocárdio/química , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Miofibrilas/química , Miofibrilas/metabolismo , Miopatias da Nemalina/genética , Miopatias da Nemalina/metabolismo , Miosinas/química , Miosinas/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Músculos Psoas/química , Músculos Psoas/metabolismo , Músculos Psoas/ultraestrutura , Sarcômeros/química , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura
8.
Proc Natl Acad Sci U S A ; 105(36): 13385-90, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18765796

RESUMO

Biological responses to mechanical stress require strain-sensing molecules, whose mechanically induced conformational changes are relayed to signaling cascades mediating changes in cell and tissue properties. In vertebrate muscle, the giant elastic protein titin is involved in strain sensing via its C-terminal kinase domain (TK) at the sarcomeric M-band and contributes to the adaptation of muscle in response to changes in mechanical strain. TK is regulated in a unique dual autoinhibition mechanism by a C-terminal regulatory tail, blocking the ATP binding site, and tyrosine autoinhibition of the catalytic base. For access to the ATP binding site and phosphorylation of the autoinhibitory tyrosine, the C-terminal autoinhibitory tail needs to be removed. Here, we use AFM-based single-molecule force spectroscopy, molecular dynamics simulations, and enzymatics to study the conformational changes during strain-induced activation of human TK. We show that mechanical strain activates ATP binding before unfolding of the structural titin domains, and that TK can thus act as a biological force sensor. Furthermore, we identify the steps in which the autoinhibition of TK is mechanically relieved at low forces, leading to binding of the cosubstrate ATP and priming the enzyme for subsequent autophosphorylation and substrate turnover.


Assuntos
Proteínas Musculares/química , Proteínas Quinases/química , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Simulação por Computador , Conectina , Ativação Enzimática , Cinética , Microscopia de Força Atômica , Modelos Moleculares , Proteínas Musculares/metabolismo , Proteínas Musculares/ultraestrutura , Fosforilação , Dobramento de Proteína , Proteínas Quinases/metabolismo , Proteínas Quinases/ultraestrutura , Estrutura Terciária de Proteína , Spodoptera , Estresse Mecânico
9.
Cell Rep ; 36(4): 109448, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320358

RESUMO

Phospholamban (PLN) is an important regulator of cardiac calcium handling due to its ability to inhibit the calcium ATPase SERCA. ß-Adrenergic stimulation reverses SERCA inhibition via PLN phosphorylation and facilitates fast calcium reuptake. PLN also forms pentamers whose physiological significance has remained elusive. Using mathematical modeling combined with biochemical and cell biological experiments, we show that pentamers regulate both the dynamics and steady-state levels of monomer phosphorylation. Substrate competition by pentamers and a feed-forward loop involving inhibitor-1 can delay monomer phosphorylation by protein kinase A (PKA), whereas cooperative pentamer dephosphorylation enables bistable PLN steady-state phosphorylation. Simulations show that phosphorylation delay and bistability act as complementary filters that reduce the effect of random fluctuations in PKA activity, thereby ensuring consistent monomer phosphorylation and SERCA activity despite noisy upstream signals. Preliminary analyses suggest that the PLN mutation R14del could impair noise filtering, offering a new perspective on how this mutation causes cardiac arrhythmias.


Assuntos
Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/metabolismo , Multimerização Proteica , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Animais , Soluções Tampão , Proteínas de Ligação ao Cálcio/genética , Redes Reguladoras de Genes , Células HEK293 , Humanos , Modelos Biológicos , Mutação/genética , Fosforilação , Ratos Wistar
10.
Circulation ; 113(9): 1235-43, 2006 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-16505175

RESUMO

BACKGROUND: Nonenzymatic glycation that results in the production of early-glycation Amadori-modified proteins and advanced-glycation end products may be important in the pathogenesis of diabetic complications. However, the effects of early-glycated proteins, such as glycated serum albumin (Gly-BSA), are poorly defined. In this study, we investigated the effects of Gly-BSA on reactive oxygen species (ROS) production by cardiomyocytes. METHODS AND RESULTS: Cultured neonatal rat cardiomyocytes were incubated with Gly-BSA or vehicle (bovine serum albumin [BSA]) for up to 48 hours. Gly-BSA dose-dependently increased in situ ROS production (whole-cell dichlorodihydrofluorescein fluorescence), with an optimum effect at 400 microg/mL after 24-hour incubation (152+/-10% versus BSA 100%; P<0.01). Treatment with the NADPH oxidase inhibitor apocynin, a Nox2 (gp91phox) antisense oligonucleotide (Nox2 AS), or the peptide gp91ds-tat significantly reduced Gly-BSA-induced ROS production at 24 hours (68.5+/-2.2%, 61.4+/-8.3%, and 53.2+/-5.4% reduction, respectively). NADPH-dependent activity in cell homogenates was also significantly increased by Gly-BSA at 24 hours (161+/-8% versus BSA) and was inhibited by diphenyleneiodonium, apocynin, NOX2AS, and the protein kinase C inhibitor bisindolylmaleimide I but not by a nitric oxide synthase inhibitor or mitochondrial inhibitors. Furthermore, bisindolylmaleimide I prevented Gly-BSA-stimulated Rac1 translocation, an essential step for NADPH oxidase activation. Gly-BSA-induced increases in ROS were associated with apocynin-inhibitable nuclear translocation of nuclear factor-kappaB and an increase in atrial natriuretic factor mRNA expression. CONCLUSIONS: Gly-BSA stimulates cardiomyocyte ROS production through a protein kinase C-dependent activation of a Nox2-containing NADPH oxidase, which results in nuclear factor-kappaB activation and upregulation of atrial natriuretic factor mRNA. These findings suggest that early-glycated Amadori products may play a role in the development of diabetic heart disease.


Assuntos
Glicoproteínas/farmacologia , Miócitos Cardíacos/metabolismo , NADPH Oxidases/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Fator Natriurético Atrial/genética , Células Cultivadas , Complicações do Diabetes , Cardiopatias/etiologia , Humanos , Glicoproteínas de Membrana , Miócitos Cardíacos/citologia , NADPH Oxidase 2 , NF-kappa B/metabolismo , Ratos , Soroalbumina Bovina/farmacologia , Regulação para Cima
11.
PLoS One ; 10(12): e0144614, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26656831

RESUMO

Ms1 (also known as STARS and ABRA) has been shown to act as an early stress response gene in processes as different as hypertrophy in skeletal and cardiac muscle and growth of collateral blood vessels. It is important for cardiac development in zebrafish and is upregulated in mouse models for cardiac hypertrophy as well as in human failing hearts. Ms1 possesses actin binding sites at its C-terminus and is usually found in the cell bound to actin filaments in the cytosol or in sarcomeres. We determined the NMR structure of the only folded domain of Ms1 comprising the second actin binding site called actin binding domain 2 (ABD2, residues 294-375), and found that it is similar to the winged helix-turn-helix fold adopted mainly by DNA binding domains of transcriptional factors. In vitro experiments show specific binding of this domain, in combination with a newly discovered AT-hook motif located N-terminally, to the sequence (A/C/G)AAA(C/A). NMR and fluorescence titration experiments confirm that this motif is indeed bound specifically by the recognition helix. In neonatal rat cardiomyocytes endogenous Ms1 is found in the nucleus in a spotted pattern, reminiscent of PML bodies. In adult rat cardiomyocytes Ms1 is exclusively found in the sarcomere. A nuclear localisation site in the N-terminus of the protein is required for nuclear localisation. This suggests that Ms1 has the potential to act directly in the nucleus through specific interaction with DNA in development and potentially as a response to stress in adult tissues.


Assuntos
Núcleo Celular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miócitos Cardíacos/metabolismo , Motivos AT-Hook , Animais , Sítios de Ligação , Células HeLa , Humanos , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Ratos , Sarcômeros/metabolismo
12.
Nat Genet ; 45(1): 83-7, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23222957

RESUMO

Vici syndrome is a recessively inherited multisystem disorder characterized by callosal agenesis, cataracts, cardiomyopathy, combined immunodeficiency and hypopigmentation. To investigate the molecular basis of Vici syndrome, we carried out exome and Sanger sequence analysis in a cohort of 18 affected individuals. We identified recessive mutations in EPG5 (previously KIAA1632), indicating a causative role in Vici syndrome. EPG5 is the human homolog of the metazoan-specific autophagy gene epg-5, encoding a key autophagy regulator (ectopic P-granules autophagy protein 5) implicated in the formation of autolysosomes. Further studies showed a severe block in autophagosomal clearance in muscle and fibroblasts from individuals with mutant EPG5, resulting in the accumulation of autophagic cargo in autophagosomes. These findings position Vici syndrome as a paradigm of human multisystem disorders associated with defective autophagy and suggest a fundamental role of the autophagy pathway in the immune system and the anatomical and functional formation of organs such as the brain and heart.


Assuntos
Agenesia do Corpo Caloso/genética , Antígenos de Neoplasias/genética , Autofagia/genética , Catarata/genética , Genes Recessivos , Mutação , Proteínas Relacionadas à Autofagia , Biópsia , Consanguinidade , Exoma , Família , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana Lisossomal , Lisossomos/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Proteínas/metabolismo , Proteínas de Transporte Vesicular
13.
Curr Opin Pharmacol ; 12(3): 347-54, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22483604

RESUMO

Many diseases of heart and skeletal muscle, from heart failure to muscle atrophy, pose unmet needs for specific and effective treatments. Recent advances suggest that sarcomeres, the smallest contractile units of heart and skeletal muscles, can be viable pharmacological targets. In sarcomeres, the contractile actin and myosin filaments are organised by a network of proteins combining structural and signalling functions, forming the sarcomeric cytoskeleton. This includes the giant proteins titin, obscurin and nebulin, which contain protein-binding sites along with signalling domains such as protein kinase, Rho activator, and Src-homology domains. These signalling domains have recently been implicated in sarcomere assembly, and the regulation of muscle contractile and metabolic adaptation. Although many functions of sarcomeric proteins remain to be discovered, their potential as pharmacological targets is now emerging. Here, we will review recent insight into the physiological and pathological signalling functions of sarcomeric cytoskeletal proteins and discuss new aspects and strategies in skeletal muscle signalling, pathomechanisms and therapy.


Assuntos
Citoesqueleto de Actina/efeitos dos fármacos , Cardiopatias/tratamento farmacológico , Terapia de Alvo Molecular , Proteínas Musculares/antagonistas & inibidores , Doenças Musculares/tratamento farmacológico , Sarcômeros/efeitos dos fármacos , Animais , Acoplamento Excitação-Contração/efeitos dos fármacos , Cardiopatias/metabolismo , Humanos , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Doenças Musculares/metabolismo
14.
J Cell Biol ; 191(6): 1159-72, 2010 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-21149568

RESUMO

Members of the formin family are important for actin filament nucleation and elongation. We have identified a novel striated muscle-specific splice variant of the formin FHOD3 that introduces a casein kinase 2 (CK2) phosphorylation site. The specific targeting of muscle FHOD3 to the myofibrils in cardiomyocytes is abolished in phosphomutants or by the inhibition of CK2. Phosphorylation of muscle FHOD3 also prevents its interaction with p62/sequestosome 1 and its recruitment to autophagosomes. Furthermore, we show that muscle FHOD3 efficiently promotes the polymerization of actin filaments in cardiomyocytes and that the down-regulation of its expression severely affects myofibril integrity. In murine and human cardiomyopathy, we observe reduced FHOD3 expression with a concomitant isoform switch and change of subcellular targeting. Collectively, our data suggest that a muscle-specific isoform of FHOD3 is required for the maintenance of the contractile structures in heart muscle and that its function is regulated by posttranslational modification.


Assuntos
Caseína Quinase II/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miocárdio/metabolismo , Miofibrilas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Forminas , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Miócitos Cardíacos/metabolismo , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA