Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4885, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849353

RESUMO

Inherited cardiomyopathies are common cardiac diseases worldwide, leading in the late stage to heart failure and death. The most promising treatments against these diseases are small molecules directly modulating the force produced by ß-cardiac myosin, the molecular motor driving heart contraction. Omecamtiv mecarbil and Mavacamten are two such molecules that completed phase 3 clinical trials, and the inhibitor Mavacamten is now approved by the FDA. In contrast to Mavacamten, Omecamtiv mecarbil acts as an activator of cardiac contractility. Here, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All-atom molecular dynamics simulations reveal how these molecules produce distinct effects in motor allostery thus impacting force production in opposite way. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.


Assuntos
Simulação de Dinâmica Molecular , Contração Miocárdica , Ureia , Contração Miocárdica/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Ureia/análogos & derivados , Ureia/farmacologia , Ureia/química , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Miosinas Ventriculares/metabolismo , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Animais , Benzilaminas , Uracila/análogos & derivados
2.
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
3.
Nat Commun ; 14(1): 3166, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37258552

RESUMO

To save energy and precisely regulate cardiac contractility, cardiac muscle myosin heads are sequestered in an 'off' state that can be converted to an 'on' state when exertion is increased. The 'off' state is equated with a folded-back structure known as the interacting-heads motif (IHM), which is a regulatory feature of all class-2 muscle and non-muscle myosins. We report here the human ß-cardiac myosin IHM structure determined by cryo-electron microscopy to 3.6 Å resolution, providing details of all the interfaces stabilizing the 'off' state. The structure shows that these interfaces are hot spots of hypertrophic cardiomyopathy mutations that are thought to cause hypercontractility by destabilizing the 'off' state. Importantly, the cardiac and smooth muscle myosin IHM structures dramatically differ, providing structural evidence for the divergent physiological regulation of these muscle types. The cardiac IHM structure will facilitate development of clinically useful new molecules that modulate IHM stability.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Humanos , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Microscopia Crioeletrônica , Coração , Cardiomiopatia Hipertrófica/genética
4.
Elife ; 112022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35767336

RESUMO

Mutations in the lever arm of ß-cardiac myosin are a frequent cause of hypertrophic cardiomyopathy, a disease characterized by hypercontractility and eventual hypertrophy of the left ventricle. Here, we studied five such mutations: three in the pliant region of the lever arm (D778V, L781P, and S782N) and two in the light chain-binding region (A797T and F834L). We investigated their effects on both motor function and myosin subfragment 2 (S2) tail-based autoinhibition. The pliant region mutations had varying effects on the motor function of a myosin construct lacking the S2 tail: overall, D778V increased power output, L781P reduced power output, and S782N had little effect on power output, while all three reduced the external force sensitivity of the actin detachment rate. With a myosin containing the motor domain and the proximal S2 tail, the pliant region mutations also attenuated autoinhibition in the presence of filamentous actin but had no impact in the absence of actin. By contrast, the light chain-binding region mutations had little effect on motor activity but produced marked reductions in autoinhibition in both the presence and absence of actin. Thus, mutations in the lever arm of ß-cardiac myosin have divergent allosteric effects on myosin function, depending on whether they are in the pliant or light chain-binding regions.


Assuntos
Cardiomiopatia Hipertrófica , Miosinas Ventriculares , Actinas/genética , Actinas/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Humanos , Mutação , Relação Estrutura-Atividade , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
5.
J Mol Biol ; 433(23): 167295, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34627791

RESUMO

In addition to a conventional relaxed state, a fraction of myosins in the cardiac muscle exists in a low-energy consuming super-relaxed (SRX) state, which is kept as a reserve pool that may be engaged under sustained increased cardiac demand. The conventional relaxed and the super-relaxed states are widely assumed to correspond to a structure where myosin heads are in an open configuration, free to interact with actin, and a closed configuration, inhibiting binding to actin, respectively. Disruption of the myosin SRX population is an emerging model in different heart diseases, such as hypertrophic cardiomyopathy, which results in excessive muscle contraction, and stabilizing them using myosin inhibitors is budding as an attractive therapeutic strategy. Here we examined the structure-function relationships of two myosin ATPase inhibitors, mavacamten and para-nitroblebbistatin, and found that binding of mavacamten at a site different than para-nitroblebbistatin populates myosin into the SRX state. Para-nitroblebbistatin, binding to a distal pocket to the myosin lever arm near the nucleotide-binding site, does not affect the usual myosin SRX state but instead appears to render myosin into a new, perhaps much more inhibited, 'ultra-relaxed' state. X-ray scattering-based rigid body modeling shows that both mavacamten and para-nitroblebbistatin induce novel conformations in human ß-cardiac heavy meromyosin that diverge significantly from the hypothetical open and closed states, and furthermore, mavacamten treatment causes greater compaction than para-nitroblebbistatin. Taken together, we conclude that mavacamten and para-nitroblebbistatin stabilize myosin in different structural states, and such states may give rise to different functional energy-sparing states.


Assuntos
Benzilaminas/química , Modelos Moleculares , Conformação Proteica , Uracila/análogos & derivados , Miosinas Ventriculares/química , Benzilaminas/farmacologia , Miosinas/antagonistas & inibidores , Miosinas/química , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Análise Espectral , Relação Estrutura-Atividade , Uracila/química , Uracila/farmacologia , Miosinas Ventriculares/antagonistas & inibidores
6.
Elife ; 102021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33605878

RESUMO

Hypertrophic cardiomyopathies (HCMs) are the leading cause of acute cardiac failure in young individuals. Over 300 mutations throughout ß-cardiac myosin, including in the motor domain, are associated with HCM. A ß-cardiac myosin motor mutation (R712L) leads to a severe form of HCM. Actin-gliding motility of R712L-myosin is inhibited, despite near-normal ATPase kinetics. By optical trapping, the working stroke of R712L-myosin was decreased 4-fold, but actin-attachment durations were normal. A prevalent hypothesis that HCM mutants are hypercontractile is thus not universal. R712 is adjacent to the binding site of the heart failure drug omecamtiv mecarbil (OM). OM suppresses the working stroke of normal ß-cardiac myosin, but remarkably, OM rescues the R712L-myosin working stroke. Using a flow chamber to interrogate a single molecule during buffer exchange, we found OM rescue to be reversible. Thus, the R712L mutation uncouples lever arm rotation from ATPase activity and this inhibition is rescued by OM.


Assuntos
Cardiomegalia/tratamento farmacológico , Cardiotônicos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Mutação , Ureia/análogos & derivados , Miosinas Ventriculares/genética , Humanos , Ureia/farmacologia , Miosinas Ventriculares/química
7.
J Biol Chem ; 294(46): 17451-17462, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31582565

RESUMO

Hypertrophic cardiomyopathy (HCM) is a common genetic disorder characterized by left ventricular hypertrophy and cardiac hyper-contractility. Mutations in the ß-cardiac myosin heavy chain gene (ß-MyHC) are a major cause of HCM, but the specific mechanistic changes to myosin function that lead to this disease remain incompletely understood. Predicting the severity of any ß-MyHC mutation is hindered by a lack of detailed examinations at the molecular level. Moreover, because HCM can take ≥20 years to develop, the severity of the mutations must be somewhat subtle. We hypothesized that mutations that result in early onset disease would have more severe changes in function than do later onset mutations. Here, we performed steady-state and transient kinetic analyses of myosins carrying one of seven missense mutations in the motor domain. Of these seven, four were previously identified in early onset cardiomyopathy screens. We used the parameters derived from these analyses to model the ATP-driven cross-bridge cycle. Contrary to our hypothesis, the results indicated no clear differences between early and late onset HCM mutations. Despite the lack of distinction between early and late onset HCM, the predicted occupancy of the force-holding actin·myosin·ADP complex at [Actin] = 3 Kapp along with the closely related duty ratio (the fraction of myosin in strongly attached force-holding states), and the measured ATPases all changed in parallel (in both sign and degree of change) compared with wildtype (WT) values. Six of the seven HCM mutations were clearly distinct from a set of previously characterized DCM mutations.


Assuntos
Adenosina Trifosfatases/genética , Cardiomiopatia Hipertrófica/genética , Miosinas/genética , Miosinas Ventriculares/genética , Citoesqueleto de Actina/genética , Actinas/química , Actinas/genética , Adenosina Trifosfatases/química , Idade de Início , Cardiomiopatia Hipertrófica/patologia , Feminino , Humanos , Cinética , Masculino , Mutação de Sentido Incorreto/genética , Contração Miocárdica/genética , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/genética , Miosinas/química , Índice de Gravidade de Doença , Miosinas Ventriculares/química
8.
J Biol Chem ; 294(46): 17314-17325, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31578282

RESUMO

We investigated a dilated cardiomyopathy (DCM) mutation (F764L) in human ß-cardiac myosin by determining its motor properties in the presence and absence of the heart failure drug omecamtive mecarbil (OM). The mutation is located in the converter domain, a key region of communication between the catalytic motor and lever arm in myosins, and is nearby but not directly in the OM-binding site. We expressed and purified human ß-cardiac myosin subfragment 1 (M2ß-S1) containing the F764L mutation, and compared it to WT with in vitro motility as well as steady-state and transient kinetics measurements. In the absence of OM we demonstrate that the F764L mutation does not significantly change maximum actin-activated ATPase activity but slows actin sliding velocity (15%) and the actomyosin ADP release rate constant (25%). The transient kinetic analysis without OM demonstrates that F764L has a similar duty ratio as WT in unloaded conditions. OM is known to enhance force generation in cardiac muscle while it inhibits the myosin power stroke and enhances actin-attachment duration. We found that OM has a reduced impact on F764L ATPase and sliding velocity compared with WT. Specifically, the EC50 for OM induced inhibition of in vitro motility was 3-fold weaker in F764L. Also, OM reduces maximum actin-activated ATPase 2-fold in F764L, compared with 4-fold with WT. Overall, our results suggest that F764L attenuates the impact of OM on actin-attachment duration and/or the power stroke. Our work highlights the importance of mutation-specific considerations when pursuing small molecule therapies for cardiomyopathies.


Assuntos
Cardiomiopatia Dilatada/genética , Insuficiência Cardíaca/genética , Ureia/análogos & derivados , Miosinas Ventriculares/genética , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/genética , Actinas/metabolismo , Actomiosina/genética , Adenosina Trifosfatases/genética , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/patologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/patologia , Humanos , Cinética , Atividade Motora/genética , Mutação , Contração Miocárdica/efeitos dos fármacos , Domínios Proteicos/genética , Ureia/farmacologia , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(35): E8143-E8152, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30104387

RESUMO

Mutations in ß-cardiac myosin, the predominant motor protein for human heart contraction, can alter power output and cause cardiomyopathy. However, measurements of the intrinsic force, velocity, and ATPase activity of myosin have not provided a consistent mechanism to link mutations to muscle pathology. An alternative model posits that mutations in myosin affect the stability of a sequestered, super relaxed state (SRX) of the protein with very slow ATP hydrolysis and thereby change the number of myosin heads accessible to actin. Here we show that purified human ß-cardiac myosin exists partly in an SRX and may in part correspond to a folded-back conformation of myosin heads observed in muscle fibers around the thick filament backbone. Mutations that cause hypertrophic cardiomyopathy destabilize this state, while the small molecule mavacamten promotes it. These findings provide a biochemical and structural link between the genetics and physiology of cardiomyopathy with implications for therapeutic strategies.


Assuntos
Benzilaminas/química , Uracila/análogos & derivados , Miosinas Ventriculares/química , Animais , Benzilaminas/farmacologia , Cardiomegalia/enzimologia , Cardiomegalia/genética , Humanos , Músculo Esquelético/enzimologia , Mutação , Suínos , Porco Miniatura , Uracila/química , Uracila/farmacologia , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
10.
Open Biol ; 8(4)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29669825

RESUMO

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.


Assuntos
Miosinas Cardíacas/fisiologia , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiologia , Animais , Miosinas Cardíacas/química , Miosinas Cardíacas/genética , Humanos , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/genética , Miosinas Ventriculares/química , Miosinas Ventriculares/genética , Miosinas Ventriculares/fisiologia
11.
J Struct Biol ; 200(3): 219-228, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28743637

RESUMO

X-ray structural determination of segments of the myosin rod has proved difficult because of the strong salt-dependent aggregation properties and repeating pattern of charges on the surface of the coiled-coil that lead to the formation of paracrystals. This problem has been resolved in part through the use of globular assembly domains that improve protein folding and prevent aggregation. The primary consideration now in designing coiled-coil fusion constructs for myosin is deciding where to truncate the coiled-coil and which amino acid residues to include from the folding domain. This is especially important for myosin that contains numerous regions of low predicted coiled-coil propensity. Here we describe the strategy adopted to determine the structure of the region that extends from Arg1677 - Leu1797 that included two areas that do not show a strong sequence signature of a conventional left-handed coiled coil or canonical heptad repeat. This demonstrates again that, with careful choice of fusion constructs, overlapping structures exhibit very similar conformations for the myosin rod fragments in the canonical regions. However, conformational variability is seen around Leu1706 which is a hot spot for cardiomyopathy mutations suggesting that this might be important for function.


Assuntos
Subfragmentos de Miosina/química , Miosinas Ventriculares/química , Cristalografia por Raios X , Humanos , Leucina/genética , Modelos Moleculares , Subfragmentos de Miosina/genética , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Miosinas Ventriculares/genética
12.
Elife ; 62017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28606303

RESUMO

Cardiac ß-myosin variants cause hypertrophic (HCM) or dilated (DCM) cardiomyopathy by disrupting sarcomere contraction and relaxation. The locations of variants on isolated myosin head structures predict contractility effects but not the prominent relaxation and energetic deficits that characterize HCM. During relaxation, pairs of myosins form interacting-heads motif (IHM) structures that with other sarcomere proteins establish an energy-saving, super-relaxed (SRX) state. Using a human ß-cardiac myosin IHM quasi-atomic model, we defined interactions sites between adjacent myosin heads and associated protein partners, and then analyzed rare variants from 6112 HCM and 1315 DCM patients and 33,370 ExAC controls. HCM variants, 72% that changed electrostatic charges, disproportionately altered IHM interaction residues (expected 23%; HCM 54%, p=2.6×10-19; DCM 26%, p=0.66; controls 20%, p=0.23). HCM variant locations predict impaired IHM formation and stability, and attenuation of the SRX state - accounting for altered contractility, reduced diastolic relaxation, and increased energy consumption, that fully characterizes HCM pathogenesis.


Assuntos
Cardiomiopatia Dilatada/fisiopatologia , Cardiomiopatia Hipertrófica/fisiopatologia , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo , Humanos , Modelos Moleculares , Contração Miocárdica , Ligação Proteica
13.
Sci Adv ; 3(2): e1601959, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28246639

RESUMO

Hypertrophic cardiomyopathy (HCM) affects 1 in 500 individuals and is an important cause of arrhythmias and heart failure. Clinically, HCM is characterized as causing hypercontractility, and therapies are aimed toward controlling the hyperactive physiology. Mutations in the ß-cardiac myosin comprise ~40% of genetic mutations associated with HCM, and the converter domain of myosin is a hotspot for HCM-causing mutations; however, the underlying primary effects of these mutations on myosin's biomechanical function remain elusive. We hypothesize that these mutations affect the biomechanical properties of myosin, such as increasing its intrinsic force and/or its duty ratio and therefore the ensemble force of the sarcomere. Using recombinant human ß-cardiac myosin, we characterize the molecular effects of three severe HCM-causing converter domain mutations: R719W, R723G, and G741R. Contrary to our hypothesis, the intrinsic forces of R719W and R723G mutant myosins are decreased compared to wild type and unchanged for G741R. Actin and regulated thin filament gliding velocities are ~15% faster for R719W and R723G myosins, whereas there is no change in velocity for G741R. Adenosine triphosphatase activities and the load-dependent velocity change profiles of all three mutant proteins are very similar to those of wild type. These results indicate that the net biomechanical properties of human ß-cardiac myosin carrying these converter domain mutations are very similar to those of wild type or are even slightly hypocontractile, leading us to consider an alternative mechanism for the clinically observed hypercontractility. Future work includes how these mutations affect protein interactions within the sarcomere that increase the availability of myosin heads participating in force production.


Assuntos
Cardiomegalia , Doenças Genéticas Inatas , Miosinas Ventriculares/química , Substituição de Aminoácidos , Humanos , Mutação de Sentido Incorreto , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
14.
J Biol Chem ; 292(9): 3768-3778, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28082673

RESUMO

The small molecule drug omecamtiv mecarbil (OM) specifically targets cardiac muscle myosin and is known to enhance cardiac muscle performance, yet its impact on human cardiac myosin motor function is unclear. We expressed and purified human ß-cardiac myosin subfragment 1 (M2ß-S1) containing a C-terminal Avi tag. We demonstrate that the maximum actin-activated ATPase activity of M2ß-S1 is slowed more than 4-fold in the presence of OM, whereas the actin concentration required for half-maximal ATPase was reduced dramatically (30-fold). We find OM does not change the overall actin affinity. Transient kinetic experiments suggest that there are two kinetic pathways in the presence of OM. The dominant pathway results in a slow transition between actomyosin·ADP states and increases the time myosin is strongly bound to actin. However, OM also traps a population of myosin heads in a weak actin affinity state with slow product release. We demonstrate that OM can reduce the actin sliding velocity more than 100-fold in the in vitro motility assay. The ionic strength dependence of in vitro motility suggests the inhibition may be at least partially due to drag forces from weakly attached myosin heads. OM causes an increase in duty ratio examined in the motility assay. Experiments with permeabilized human myocardium demonstrate that OM increases calcium sensitivity and slows force development (ktr) in a concentration-dependent manner, whereas the maximally activated force is unchanged. We propose that OM increases the myosin duty ratio, which results in enhanced calcium sensitivity but slower force development in human myocardium.


Assuntos
Cálcio/química , Miocárdio/metabolismo , Ureia/análogos & derivados , Miosinas Ventriculares/química , Actinas/química , Actomiosina/química , Difosfato de Adenosina/química , Animais , Relação Dose-Resposta a Droga , Humanos , Cinética , Espectrometria de Massas , Camundongos , Miosinas/química , Domínios Proteicos , Proteínas Recombinantes/química , Estresse Mecânico , Ureia/química
15.
Colloids Surf B Biointerfaces ; 151: 178-188, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28012406

RESUMO

The objective of this work was to develop a simple surface modification technique using keratin derived from human hair for efficient cardiomyogenic lineage commitment of human mesenchymal stem cells (hMSCs). Keratin was extracted from discarded human hair containing both the acidic and basic components along with the heterodimers. The extracted keratin was adsorbed to conventional tissue culture polystyrene surfaces at different concentration. Keratin solution of 500µg/ml yielded a well coated layer of 12±1nm thickness with minimal agglomeration. The keratin coated surfaces promoted cell attachment and proliferation. Large increases in the mRNA expression of known cardiomyocyte genes such as cardiac actinin, cardiac troponin and ß-myosin heavy chain were observed. Immunostaining revealed increased expression of sarcomeric α-actinin and tropomyosin whereas Western blots confirmed higher expression of tropomyosin and myocyte enhancer factor 2C in cells on the keratin coated surface than on the non-coated surface. Keratin promoted DNA demethylation of the Atp2a2 and Nkx2.5 genes thereby elucidating the importance of epigenetic changes as a possible molecular mechanism underlying the increased differentiation. A global gene expression analysis revealed a significant alteration in the expression of genes involved in pathways associated in cardiomyogenic commitment including cytokine and chemokine signaling, cell-cell and cell-matrix interactions, Wnt signaling, MAPK signaling, TGF-ß signaling and FGF signaling pathways among others. Thus, adsorption of keratin offers a facile and affordable yet potent route for inducing cardiomyogenic lineage commitment of stem cells with important implications in developing xeno-free strategies in cardiovascular regenerative medicine.


Assuntos
Cabelo/química , Queratinas/química , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Adsorção , Animais , Adesão Celular , Linhagem da Célula , Sobrevivência Celular , Ilhas de CpG , Metilação de DNA , Matriz Extracelular/metabolismo , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Fatores de Transcrição MEF2/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Poliestirenos/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Propriedades de Superfície , Fator de Crescimento Transformador beta/metabolismo , Miosinas Ventriculares/química
16.
Cell Rep ; 17(11): 2857-2864, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27974200

RESUMO

Hypertrophic cardiomyopathy (HCM) is a heritable cardiovascular disorder that affects 1 in 500 people. A significant percentage of HCM is attributed to mutations in ß-cardiac myosin, the motor protein that powers ventricular contraction. This study reports how two early-onset HCM mutations, D239N and H251N, affect the molecular biomechanics of human ß-cardiac myosin. We observed significant increases (20%-90%) in actin gliding velocity, intrinsic force, and ATPase activity in comparison to wild-type myosin. Moreover, for H251N, we found significantly lower binding affinity between the S1 and S2 domains of myosin, suggesting that this mutation may further increase hyper-contractility by releasing active motors. Unlike previous HCM mutations studied at the molecular level using human ß-cardiac myosin, early-onset HCM mutations lead to significantly larger changes in the fundamental biomechanical parameters and show clear hyper-contractility.


Assuntos
Actinas/genética , Cardiomiopatia Hipertrófica/genética , Proteínas Motores Moleculares/genética , Miosinas Ventriculares/genética , Actinas/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Genótipo , Humanos , Mutação , Contração Miocárdica/genética , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo
17.
Arch Biochem Biophys ; 580: 14-21, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26116789

RESUMO

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


Assuntos
Actinina/química , Actinas/química , Cadeias Leves de Miosina/química , Quinase de Cadeia Leve de Miosina/química , Miosinas Ventriculares/química , Actinina/genética , Actinas/genética , Animais , Galinhas , Expressão Gênica , Ventrículos do Coração/química , Humanos , Cinética , Movimento (Física) , Músculo Esquelético/química , Músculo Liso/química , Mutação , Cadeias Leves de Miosina/genética , Quinase de Cadeia Leve de Miosina/genética , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Suínos , Miosinas Ventriculares/genética
18.
Biophys J ; 107(12): L41-L44, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25517169

RESUMO

The heart adjusts its power output to meet specific physiological needs through the coordination of several mechanisms, including force-induced changes in contractility of the molecular motor, the ß-cardiac myosin (ßCM). Despite its importance in driving and regulating cardiac power output, the effect of force on the contractility of a single ßCM has not been measured. Using single molecule optical-trapping techniques, we found that ßCM has a two-step working stroke. Forces that resist the power stroke slow the myosin-driven contraction by slowing the rate of ADP release, which is the kinetic step that limits fiber shortening. The kinetic properties of ßCM are affected by load, suggesting that the properties of myosin contribute to the force-velocity relationship in intact muscle and play an important role in the regulation of cardiac power output.


Assuntos
Miócitos Cardíacos/metabolismo , Miosinas Ventriculares/metabolismo , Animais , Contração Miocárdica , Miócitos Cardíacos/fisiologia , Suínos , Miosinas Ventriculares/química
19.
Biochemistry ; 53(32): 5298-306, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25068717

RESUMO

Ventricular myosin (ßMys) is the motor protein in cardiac muscle generating force using ATP hydrolysis free energy to translate actin. In the cardiac muscle sarcomere, myosin and actin filaments interact cyclically and undergo rapid relative translation facilitated by the low duty cycle motor. It contrasts with high duty cycle processive myosins for which persistent actin association is the priority. The only pharmaceutical ßMys activator, omecamtive mecarbil (OM), upregulates cardiac contractility in vivo and is undergoing testing for heart failure therapy. In vitro ßMys step-size, motility velocity, and actin-activated myosin ATPase were measured to determine duty cycle in the absence and presence of OM. A new parameter, the relative step-frequency, was introduced and measured to characterize ßMys motility due to the involvement of its three unitary step-sizes. Step-size and relative step-frequency were measured using the Qdot assay. OM decreases motility velocity 10-fold without affecting step-size, indicating a large increase in duty cycle converting ßMys to a near processive myosin. The OM conversion dramatically increases force and modestly increases power over the native ßMys. Contrasting motility modification due to OM with that from the natural myosin activator, specific ßMys phosphorylation, provides insight into their respective activation mechanisms and indicates the boilerplate screening characteristics desired for pharmaceutical ßMys activators. New analytics introduced here for the fast and efficient Qdot motility assay create a promising method for high-throughput screening of motor proteins and their modulators.


Assuntos
Estreptavidina/farmacologia , Ureia/análogos & derivados , Miosinas Ventriculares/química , Miosinas Ventriculares/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Pontos Quânticos , Coelhos , Estreptavidina/química , Ureia/química , Ureia/farmacologia
20.
Biochem Biophys Res Commun ; 449(3): 284-8, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24857983

RESUMO

Binding of the utmost N-terminus of essential myosin light chains (ELC) to actin slows down myosin motor function. In this study, we investigated the binding constants of two different human cardiac ELC isoforms with actin. We employed circular dichroism (CD) and surface plasmon resonance (SPR) spectroscopy to determine structural properties and protein-protein interaction of recombinant human atrial and ventricular ELC (hALC-1 and hVLC-1, respectively) with α-actin as well as α-actin with alanin-mutated ELC binding site (α-actin(ala3)) as control. CD spectroscopy showed similar secondary structure of both hALC-1 and hVLC-1 with high degree of α-helicity. SPR spectroscopy revealed that the affinity of hALC-1 to α-actin (KD=575 nM) was significantly (p<0.01) lower compared with the affinity of hVLC-1 to α-actin (KD=186 nM). The reduced affinity of hALC-1 to α-actin was mainly due to a significantly (p<0.01) lower association rate (kon: 1,018 M(-1)s(-1)) compared with kon of the hVLC-1/α-actin complex interaction (2,908 M(-1)s(-1)). Hence, differential expression of ELC isoforms could modulate muscle contractile activity via distinct α-actin interactions.


Assuntos
Actinas/metabolismo , Miosinas Atriais/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosinas Ventriculares/metabolismo , Actinas/química , Actinas/genética , Miosinas Atriais/química , Miosinas Atriais/genética , Dicroísmo Circular , Humanos , Contração Miocárdica , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/genética , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Miosinas Ventriculares/química , Miosinas Ventriculares/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...