Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 19(5): e1011099, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37200380

RESUMO

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.


Assuntos
Miosinas Cardíacas , Miosinas Ventriculares , Humanos , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo , Coração , Miocárdio/metabolismo , Miosinas/química , Ureia/metabolismo
2.
Biophys J ; 122(1): 54-62, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36451546

RESUMO

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.


Assuntos
Coração , Miosinas , Miosinas/metabolismo , Miocárdio/metabolismo , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Domínios Proteicos , Ureia/metabolismo
3.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35163146

RESUMO

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.


Assuntos
Actomiosina/química , Actomiosina/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Sítios de Ligação , Humanos , Cinética , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica
4.
J Med Microbiol ; 70(5)2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33956590

RESUMO

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.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Febre Reumática/imunologia , Cardiopatia Reumática/imunologia , Autoanticorpos/sangue , Autoantígenos/química , Miosinas Cardíacas/química , Miosinas Cardíacas/imunologia , Humanos , Queratinas/imunologia , Laminina/imunologia , Estudos Longitudinais , Peptídeos/química , Peptídeos/imunologia , Febre Reumática/sangue , Cardiopatia Reumática/sangue , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Tropomiosina/imunologia
5.
ACS Infect Dis ; 7(6): 1483-1502, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34019767

RESUMO

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.


Assuntos
Papaína , Peptídeo Hidrolases , SARS-CoV-2/enzimologia , Proteases Virais/metabolismo , Sequência de Aminoácidos , Miosinas Cardíacas/química , Fatores de Transcrição Forkhead/química , Humanos , Cadeias Pesadas de Miosina/química , Papaína/metabolismo , Peptídeo Hidrolases/metabolismo , Proteína S/química , Receptor ErbB-4/química
6.
Proc Natl Acad Sci U S A ; 115(32): E7486-E7494, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30018063

RESUMO

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.


Assuntos
Actinas/metabolismo , Benzilaminas/farmacologia , Miosinas Cardíacas/metabolismo , Cardiomiopatia Hipertrófica Familiar/tratamento farmacológico , Subfragmentos de Miosina/metabolismo , Uracila/análogos & derivados , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/efeitos dos fármacos , Benzilaminas/uso terapêutico , Miosinas Cardíacas/química , Cardiomiopatia Hipertrófica Familiar/etiologia , Humanos , Cinética , Subfragmentos de Miosina/química , Estabilidade Proteica/efeitos dos fármacos , Uracila/farmacologia , Uracila/uso terapêutico
7.
J Biol Chem ; 293(23): 9017-9029, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29666183

RESUMO

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.


Assuntos
Miosinas Cardíacas/genética , Cardiomiopatia Dilatada/genética , Cadeias Pesadas de Miosina/genética , Mutação Puntual , Actinas/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Linhagem Celular , Humanos , Cinética , Camundongos , Modelos Moleculares , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Domínios Proteicos
8.
J Mol Biol ; 430(10): 1459-1478, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29660325

RESUMO

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.


Assuntos
Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Miopatias Distais/genética , Fibras Musculares Esqueléticas/citologia , Mutação , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Animais , Miosinas Cardíacas/metabolismo , Linhagem Celular , Técnicas In Vitro , Camundongos , Microscopia Eletrônica , Simulação de Dinâmica Molecular , Fibras Musculares Esqueléticas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Estrutura Secundária de Proteína , Ratos , Sarcômeros/química , Sarcômeros/metabolismo
9.
J Biol Chem ; 292(40): 16571-16577, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28808052

RESUMO

Mavacamten, formerly known as MYK-461 is a recently discovered novel small-molecule modulator of cardiac myosin that targets the underlying sarcomere hypercontractility of hypertrophic cardiomyopathy, one of the most prevalent heritable cardiovascular disorders. Studies on isolated cells and muscle fibers as well as intact animals have shown that mavacamten inhibits sarcomere force production, thereby reducing cardiac contractility. Initial mechanistic studies have suggested that mavacamten primarily reduces the steady-state ATPase activity by inhibiting the rate of phosphate release of ß-cardiac myosin-S1, but the molecular mechanism of action of mavacamten has not been described. Here we used steady-state and presteady-state kinetic analyses to investigate the mechanism of action of mavacamten. Transient kinetic analyses revealed that mavacamten modulates multiple steps of the myosin chemomechanical cycle. In addition to decreasing the rate-limiting step of the cycle (phosphate release), mavacamten reduced the number of myosin-S1 heads that can interact with the actin thin filament during transition from the weakly to the strongly bound state without affecting the intrinsic rate. Mavacamten also decreased the rate of myosin binding to actin in the ADP-bound state and the ADP-release rate from myosin-S1 alone. We, therefore, conclude that mavacamten acts on multiple stages of the myosin chemomechanical cycle. Although the primary mechanism of mavacamten-mediated inhibition of cardiac myosin is the decrease of phosphate release from ß-cardiac myosin-S1, a secondary mechanism decreases the number of actin-binding heads transitioning from the weakly to the strongly bound state, which occurs before phosphate release and may provide an additional method to modulate myosin function.


Assuntos
Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Benzilaminas/química , Miosinas Cardíacas/química , Subfragmentos de Miosina/química , Sarcômeros/química , Uracila/análogos & derivados , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Miosinas Cardíacas/metabolismo , Cardiomegalia/metabolismo , Bovinos , Subfragmentos de Miosina/metabolismo , Sarcômeros/metabolismo , Uracila/química
10.
Biophys J ; 113(1): 91-100, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28700929

RESUMO

We have used site-directed time-resolved fluorescence resonance energy transfer to determine the effect of a pathological mutation in the human ventricular essential light chain (hVELC) of myosin, on the structural dynamics of the actin-myosin complex. The hVELC modulates the function of actomyosin, through the interaction of its N-terminal extension with actin and its C-terminal lobe with the myosin heavy chain. Several mutations in hVELC are associated with hypertrophic cardiomyopathy (HCM). Some biochemical effects of these mutations are known, but further insight is needed about their effects on the structural dynamics of functioning actomyosin. Therefore, we introduced the HCM mutation E56G into a single-cysteine (C16) hVELC construct and substituted it for the VELC of bovine cardiac myosin subfragment 1. Using a donor fluorescent probe on actin (at C374) and an acceptor probe on C16 of hVELC, we performed time-resolved fluorescence resonance energy transfer, directly detecting structural changes within the bound actomyosin complex during function. The E56G mutation has no significant effect on actin-activated ATPase activity or actomyosin affinity in the presence of ATP, or on the structure of the strong-binding S complex in the absence of ATP. However, in the presence of saturating ATP, where both W (prepowerstroke) and S (postpowerstroke) structural states are observed, the mutant increases the mole fraction of the S complex (increasing the duty ratio), while shifting the structure of the remaining W complex toward that of S, indicating a structural redistribution toward the strongly bound (force-generating) complex. We propose that this effect is responsible for the hypercontractile phenotype induced by this HCM mutation in myosin.


Assuntos
Actomiosina/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Mutação , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Actinas/química , Actinas/metabolismo , Actomiosina/química , Actomiosina/genética , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Animais , Miosinas Cardíacas/química , Bovinos , Escherichia coli , Transferência Ressonante de Energia de Fluorescência , Humanos , Modelos Moleculares , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Cadeias Leves de Miosina/química , Coelhos
11.
Biophys J ; 112(5): 984-996, 2017 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-28297657

RESUMO

Modeling the complete actin.myosin ATPase cycle has always been limited by the lack of experimental data concerning key steps of the cycle, because these steps can only be defined at very low ionic strength. Here, using human ß-cardiac myosin-S1, we combine published data from transient and steady-state kinetics to model a minimal eight-state ATPase cycle. The model illustrates the occupancy of each intermediate around the cycle and how the occupancy is altered by changes in actin concentration for [actin] = 1-20Km. The cycle can be used to predict the maximal velocity of contraction (by motility assay or sarcomeric shortening) at different actin concentrations (which is consistent with experimental velocity data) and predict the effect of a 5 pN load on a single motor. The same exercise was repeated for human α-cardiac myosin S1 and rabbit fast skeletal muscle S1. The data illustrates how the motor domain properties can alter the ATPase cycle and hence the occupancy of the key states in the cycle. These in turn alter the predicted mechanical response of the myosin independent of other factors present in a sarcomere, such as filament stiffness and regulatory proteins. We also explore the potential of this modeling approach for the study of mutations in human ß-cardiac myosin using the hypertrophic myopathy mutation R453C. Our modeling, using the transient kinetic data, predicts mechanical properties of the motor that are compatible with the single-molecule study. The modeling approach may therefore be of wide use for predicting the properties of myosin mutations.


Assuntos
Actinas/metabolismo , Miosinas Cardíacas/metabolismo , Modelos Moleculares , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Trifosfato de Adenosina/metabolismo , Miosinas Cardíacas/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Ligação Proteica , Isoformas de Proteínas/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(10): E1796-E1804, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28223517

RESUMO

Omecamtiv mecarbil (OM), a putative heart failure therapeutic, increases cardiac contractility. We hypothesize that it does this by changing the structural kinetics of the myosin powerstroke. We tested this directly by performing transient time-resolved FRET on a ventricular cardiac myosin biosensor. Our results demonstrate that OM stabilizes myosin's prepowerstroke structural state, supporting previous measurements showing that the drug shifts the equilibrium constant for myosin-catalyzed ATP hydrolysis toward the posthydrolysis biochemical state. OM slowed the actin-induced powerstroke, despite a twofold increase in the rate constant for actin-activated phosphate release, the biochemical step in myosin's ATPase cycle associated with force generation and the conversion of chemical energy into mechanical work. We conclude that OM alters the energetics of cardiac myosin's mechanical cycle, causing the powerstroke to occur after myosin weakly binds to actin and releases phosphate. We discuss the physiological implications for these changes.


Assuntos
Miosinas Cardíacas/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Miosinas/efeitos dos fármacos , Ureia/análogos & derivados , Animais , Técnicas Biossensoriais , Miosinas Cardíacas/química , Miosinas Cardíacas/isolamento & purificação , Fármacos Cardiovasculares/administração & dosagem , Fármacos Cardiovasculares/química , Bovinos , Galinhas , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Cinética , Contração Miocárdica/efeitos dos fármacos , Miocárdio/enzimologia , Miocárdio/patologia , Miosinas/química , Fosfatos/química , Fosfatos/metabolismo , Coelhos , Ureia/administração & dosagem , Ureia/química
13.
Nano Lett ; 17(2): 1262-1268, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28112520

RESUMO

Despite recent advances in thermometry, determination of temperature at the nanometer scale in single molecules to live cells remains a challenge that holds great promise in disease detection among others. In the present study, we use a new approach to nanometer scale thermometry with a spatial and thermal resolution of 80 nm and 1 mK respectively, by directly associating 2 nm cadmium telluride quantum dots (CdTe QDs) to the subject under study. The 2 nm CdTe QDs physically adhered to bovine cardiac and rabbit skeletal muscle myosin, enabling the determination of heat released when ATP is hydrolyzed by both myosin motors. Greater heat loss reflects less work performed by the motor, hence decreased efficiency. Surprisingly, we found rabbit skeletal myosin to be more efficient than bovine cardiac. We have further extended this approach to demonstrate the gain in efficiency of Drosophila melanogaster skeletal muscle overexpressing the PGC-1α homologue spargel, a known mediator of improved exercise performance in humans. Our results establish a novel approach to determine muscle efficiency with promise for early diagnosis and treatment of various metabolic disorders including cancer.


Assuntos
Compostos de Cádmio/química , Miosinas Cardíacas/química , Músculo Esquelético/fisiologia , Pontos Quânticos/química , Miosinas de Músculo Esquelético/química , Telúrio/química , Trifosfato de Adenosina/química , Animais , Bovinos , Drosophila melanogaster/fisiologia , Fluorescência , Hidrólise , Masculino , Nanotecnologia , Tamanho da Partícula , Coelhos , Miosinas de Músculo Esquelético/fisiologia , Propriedades de Superfície , Temperatura , Termometria
14.
Arch Biochem Biophys ; 618: 1-8, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28131772

RESUMO

Cardiac myosin is the motor powering the heart. It moves actin with 3 step-size varieties generated by torque from the myosin heavy chain lever-arm rotation under the influence of myosin essential light chain whose N-terminal extension binds actin. Proposed mechanisms adapting myosin mechanochemical characteristics on the fly sometimes involve modulation of step-size selection probability via motor strain sensitivity. Strain following the power stroke, hypothetically imposed by the finite actin detachment rate 1/ton, is shown to have no effect on unloaded velocity when multiple myosins are simultaneously strongly actin bound in an in vitro motility assay. Actin filaments slide ∼2 native step-sizes while more than 1 myosin strongly binds actin probably ruling out an actin detachment limited model for imposing strain. It suggests that single myosin estimates for ton are too large, not applicable to the ensemble situation, or both. Parallel motility data quantitation involving instantaneous particle velocities (frame velocity) and actin filament track averaged velocities (track velocity) give an estimate of the random walk step-size, δ. Comparing δ for slow and fast motility components suggests the higher speed component has cardiac myosin upshifting to longer steps. Variable step-size characteristics imply cardiac myosin maintains a velocity dynamic range not involving strain.


Assuntos
Actinas/química , Miosinas Cardíacas/química , Ventrículos do Coração/metabolismo , Citoesqueleto de Actina/química , Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Adsorção , Animais , Movimento Celular , Reagentes de Ligações Cruzadas/química , Concentração de Íons de Hidrogênio , Cinética , Probabilidade , Domínios Proteicos , Solubilidade , Suínos , Temperatura
15.
J Biol Chem ; 290(49): 29270-80, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26446785

RESUMO

Our molecular modeling studies suggest a charge-dependent interaction between residues Glu-497 in the relay domain and Arg-712 in the converter domain of human ß-cardiac myosin. To test the significance of this putative interaction, we generated transgenic Drosophila expressing indirect flight muscle myosin with charge reversal mutations in the relay (E496R) or converter (R713E). Each mutation yielded dramatic reductions in myosin Ca-ATPase activity (~80%) as well as in basal (~67%) and actin-activated (~84%) Mg-ATPase activity. E496R myosin-induced in vitro actin-sliding velocity was reduced by 71% and R713E myosin permitted no actin motility. Indirect flight muscles of late pupae from each mutant displayed disrupted myofibril assembly, with adults having severely abnormal myofibrils and no flight ability. To understand the molecular basis of these defects, we constructed a putative compensatory mutant that expresses myosin with both E496R and R713E. Intriguingly, ATPase values were restored to ~73% of wild-type and actin-sliding velocity increased to 40%. The double mutation suppresses myofibril assembly defects in pupal indirect flight muscles and dramatically reduces myofibril disruption in young adults. Although sarcomere organization is not sustained in older flies and flight ability is not restored in homozygotes, young heterozygotes fly well. Our results indicate that this charge-dependent interaction between the myosin relay and converter domains is essential to the mechanochemical cycle and sarcomere assembly. Furthermore, the same inter-domain interaction is disrupted when modeling human ß-cardiac myosin heavy chain cardiomyopathy mutations E497D or R712L, implying that abolishing this salt bridge is one cause of the human disease.


Assuntos
Miosinas Cardíacas/química , Cardiomiopatia Hipertrófica/fisiopatologia , Cadeias Pesadas de Miosina/química , Actinas/química , Adenosina Trifosfatases/química , Aminoácidos/química , Animais , Animais Geneticamente Modificados , Galinhas , Cruzamentos Genéticos , Modelos Animais de Doenças , Drosophila melanogaster , Feminino , Humanos , Modelos Moleculares , Músculo Esquelético/metabolismo , Mutação , Miofibrilas/metabolismo , Pectinidae , Fenótipo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Sais/química , Sarcômeros/metabolismo , Transgenes
16.
FEBS J ; 282(12): 2379-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25825243

RESUMO

Dilated cardiomyopathy (DCM) is a disease of the myocardium characterized by left ventricular dilatation and diminished contractile function. Here we describe a novel DCM mutation in the myosin regulatory light chain (RLC), in which aspartic acid at position 94 is replaced by alanine (D94A). The mutation was identified by exome sequencing of three adult first-degree relatives who met formal criteria for idiopathic DCM. To obtain insight into the functional significance of this pathogenic MYL2 variant, we cloned and purified the human ventricular RLC wild-type (WT) and D94A mutant proteins, and performed in vitro experiments using RLC-mutant or WT-reconstituted porcine cardiac preparations. The mutation induced a reduction in the α-helical content of the RLC, and imposed intra-molecular rearrangements. The phosphorylation of RLC by Ca²âº/calmodulin-activated myosin light chain kinase was not affected by D94A. The mutation was seen to impair binding of RLC to the myosin heavy chain, and its incorporation into RLC-depleted porcine myosin. The actin-activated ATPase activity of mutant-reconstituted porcine cardiac myosin was significantly higher compared with ATPase of wild-type. No changes in the myofibrillar ATPase-pCa relationship were observed in wild-type- or D94A-reconstituted preparations. Measurements of contractile force showed a slightly reduced maximal tension per cross-section of muscle, with no change in the calcium sensitivity of force in D94A-reconstituted skinned porcine papillary muscle strips compared with wild-type. Our data indicate that subtle structural rearrangements in the RLC molecule, followed by its impaired interaction with the myosin heavy chain, may trigger functional abnormalities contributing to the DCM phenotype.


Assuntos
Miosinas Cardíacas/genética , Cardiomiopatia Dilatada/genética , Mutação , Cadeias Leves de Miosina/genética , Actinas/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Adulto , Substituição de Aminoácidos , Animais , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Dicroísmo Circular , Análise Mutacional de DNA , Feminino , Humanos , Masculino , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/metabolismo , Linhagem , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sus scrofa
17.
Proc Natl Acad Sci U S A ; 112(15): 4660-5, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825773

RESUMO

We have used time-resolved fluorescence resonance energy transfer (TR-FRET) to determine the role of myosin essential light chains (ELCs) in structural transitions within the actomyosin complex. Skeletal muscle myosins have two ELC isoforms, A1 and A2, which differ by an additional 40-45 residues at the N terminus of A1, and subfragment 1 (S1) containing A1 (S1A1) has higher catalytic efficiency and higher affinity for actin than S1A2. ELC's location at the junction between the catalytic and light-chain domains gives it the potential to play a central role in the force-generating power stroke. Therefore, we measured site-directed TR-FRET between a donor on actin and an acceptor near the C terminus of ELC, detecting directly the rotation of the light-chain domain (lever arm) relative to actin (power stroke), induced by the interaction of ATP-bound myosin with actin. TR-FRET resolved the weakly bound (W) and strongly bound (S) states of actomyosin during the W-to-S transition (power stroke). We found that the W states are essentially the same for the two isoenzymes, but the S states are quite different, indicating a much larger movement of S1A1. FRET from actin to a probe on the N-terminal extension of A1 showed close proximity to actin. We conclude that the N-terminal extension of A1-ELC modulates the W-to-S structural transition of acto-S1, so that the light-chain domain undergoes a much larger power stroke in S1A1 than in S1A2. These results have profound implications for understanding the contractile function of actomyosin, as needed in therapeutic design for muscle disorders.


Assuntos
Actomiosina/metabolismo , Miosinas Cardíacas/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Músculo Esquelético/metabolismo , Cadeias Leves de Miosina/metabolismo , Actinas/química , Actinas/metabolismo , Actomiosina/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Algoritmos , Animais , Miosinas Cardíacas/química , Cinética , Modelos Moleculares , Contração Muscular , Cadeias Leves de Miosina/química , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Coelhos , Rotação , Fatores de Tempo
18.
Biochemistry ; 54(10): 1963-75, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25680381

RESUMO

We determined the effect of Omecamtiv Mecarbil, a novel allosteric effector of cardiac muscle myosin, on the kinetic and "in vitro" motility properties of the porcine ventricular heavy meromyosin (PV-HMM). Omecamtiv Mecarbil increases the equilibrium constant of the hydrolysis step (M-ATP ⇄ M-ADP-Pi) from 2.4 to 6 as determined by quench flow, but the maximal rates of both the hydrolysis step and tryptophan fluorescence increase are unchanged by the drug. OM also increases the amplitude of the fast phase of phosphate dissociation (AM-ADP-Pi → AM-ADP + Pi) that is associated with force production in muscle by 4-fold. These results suggest a mechanism in which hydrolysis of M-ATP to M-ADP-Pi occurs both before and after the recovery stroke, but rapid acceleration of phosphate dissociation by actin occurs only on post-recovery stroke A-M-ADP-Pi. One of the more dramatic effects of OM on PV-HMM is a 14-fold decrease in the unloaded shortening velocity measured by the in vitro motility assay. The increase in flux through phosphate dissociation and the unchanged rate of ADP dissociation (AM-ADP → AM + ADP) by the drug produce a higher duty ratio motor in which a larger fraction of myosin heads are strongly bound to actin filaments. The increased internal load produced by a larger fraction of strongly attached crossbridges explains the reduced rate of in vitro motility velocity in the presence of OM and predicts that the drug will produce slower and stronger contraction of cardiac muscle.


Assuntos
Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Miosinas Cardíacas/química , Miocárdio/química , Ureia/análogos & derivados , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Miosinas Cardíacas/metabolismo , Cinética , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Suínos , Ureia/química
19.
Eur J Cardiothorac Surg ; 47(3): 416-25; discussion 425, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24778452

RESUMO

OBJECTIVES: Cross-talk between organ-specific extracellular matrix (ECM) and stem cells is often assumed but has not been directly demonstrated. We developed a protocol for the preparation of human cardiac ECM (cECM) and studied whether cECM has effects on pluripotent stem cell differentiation that may be useful for future cardiac regeneration strategies in patients with end-stage heart failure. METHODS: Of note, 0.3 mm-thick cECM slices were prepared from samples of myocardium from patients with end-stage non-ischaemic dilated cardiomyopathy, using a three-step protocol involving hypotonic lysis buffer, sodium dodecyl sulphate (SDS) and foetal bovine serum (FBS). Murine embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and mesenchymal stromal cells (MSCs) were seeded and grown in standard culture, on cECM or on non-specific ECM preparations (Matrigel® or Geltrex®). Cell attachment, apoptosis induction (Caspase 3/7 activity) and metabolic activity (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium conversion) were followed. Transcriptional activation of genes involved in pluripotency; early and late myocardial development; and endothelial, ectodermal or endodermal commitment were monitored by quantitative real-time polymerase chain reaction (rtPCR). Protein expression of selected markers was confirmed by immunohistology. RESULTS: cECM supported the proliferation of ESCs and iPSCs, and Caspase 3/7 activity was significantly lower compared with standard culture. Cardiac lineage commitment was favoured when ESCs or iPSCs were grown on cECM, as evidenced by the significantly increased mRNA expression of cardiac alpha myosin heavy polypeptide 6 (Myh6), cardiac troponin T2 (Tnnt2) and NK2 homeobox 5 (Nkx2.5) as well as positive immunohistology for cardiac troponin T and heavy-chain cardiac myosin protein. In contrast, Matrigel or Geltrex did not induce cardiac-specific markers. MSCs showed no evidence of cardiomyocyte differentiation. CONCLUSIONS: Human cardiac ECM seems to direct differentiation of pluripotent stem cells towards a cardiomyocyte phenotype. This phenomenon supports the use of cardiac ECM preparations for guided stem cell differentiation and myocardial repair, and may ultimately increase the therapeutic efficacy of cell therapy in heart failure patients.


Assuntos
Matriz Extracelular/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Apoptose , Biomarcadores/análise , Biomarcadores/metabolismo , Miosinas Cardíacas/análise , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Matriz Extracelular/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/química , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/química , Células-Tronco Pluripotentes/fisiologia , Troponina T/análise , Troponina T/química , Troponina T/metabolismo
20.
Biochemistry ; 53(42): 6717-24, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25265574

RESUMO

We have used enzyme kinetics to investigate the molecular mechanism by which the N-terminal domains of human and mouse cardiac MyBP-C (C0C1, C1C2, and C0C2) affect the activation of myosin ATP hydrolysis by F-actin and by native porcine thin filaments. N-Terminal domains of cMyBP-C inhibit the activation of myosin-S1 ATPase by F-actin. However, mouse and human C1C2 and C0C2 produce biphasic activating and inhibitory effects on the activation of myosin ATP hydrolysis by native cardiac thin filaments. Low ratios of MyBP-C N-terminal domains to thin filaments activate myosin-S1 ATP hydrolysis, but higher ratios inhibit ATP hydrolysis, as is observed with F-actin alone. These data suggest that low concentrations of C1C2 and C0C2 activate thin filaments by a mechanism similar to that of rigor myosin-S1, whereas higher concentrations inhibit the ATPase rate by competing with myosin-S1-ADP-Pi for binding to actin and thin filaments. In contrast to C0C2 and C1C2, the activating effects of the C0C1 domain are species-dependent: human C0C1 activates actomyosin-S1 ATPase rates, but mouse C0C1 does not produce significant activation or inhibition. Phosphorylation of serine residues in the m-linker between the C1 and C2 domains by protein kinase-A decreases the activation of thin filaments by huC0C2 at pCa > 8 but has little effect on the activation mechanism at pCa = 4. In sarcomeres, the low ratio of cMyBP-C to actin is expected to favor the activating effects of cMyBP-C while minimizing inhibition produced by competition with myosin heads.


Assuntos
Citoesqueleto de Actina/química , Trifosfato de Adenosina/química , Proteínas de Transporte/química , Miocárdio/química , Miosinas/química , Actinas/química , Animais , Cálcio/química , Miosinas Cardíacas/química , Humanos , Hidrólise , Cinética , Camundongos , Fosforilação , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/química , Especificidade da Espécie , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA