Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.599
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(1): 105521, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38042484

RESUMO

Myosin essential light chains A1 and A2 are identical isoforms except for an extension of ∼40 amino acids at the N terminus of A1 that binds F-actin. The extension has no bearing on the burst hydrolysis rate (M-ATP → M-ADP-Pi) as determined by chemical quench flow (100 µM isoenzyme). Whereas actomyosin-S1A2 steady state MgATPase (low ionic strength, 20 °C) is hyperbolically dependent on concentration: Vmax 7.6 s-1, Kapp 6.4 µM (F-actin) and Vmax 10.1 s-1, Kapp 5.5 µM (native thin filaments, pCa 4), the relationship for myosin-S1A1 is bimodal; an initial rise at low concentration followed by a decline to one-third the Vmax of S1A2, indicative of more than one rate-limiting step and A1-enforced flux through the slower actomyosin-limited hydrolysis pathway. In double-mixing stopped-flow with an indicator, Ca(II)-mediated activation of Pi dissociation (regulatedAM-ADP-Pi → regulatedAM-ADP + Pi) is attenuated by A1 attachment to thin filaments (pCa 4). The maximum accelerated rates of Pi dissociation are: 81 s-1 (S1A1, Kapp 8.9 µM) versus 129 s-1 (S1A2, Kapp 58 µM). To investigate apomyosin-S1-mediated activation, thin filaments (EGTA) are premixed with a given isomyosin-S1 and double-mixing is repeated with myosin-S1A1 in the first mix. Similar maximum rates of Pi dissociation are observed, 44.5 s-1 (S1A1) and 47.1 s-1 (S1A2), which are lower than for Ca(II) activation. Overall, these results biochemically demonstrate how the longer light chain A1 can contribute to slower contraction and higher force and the shorter version A2 to faster contraction and lower force, consistent with their distribution in different types of striated muscle.


Assuntos
Actomiosina , Cadeias Leves de Miosina , Actinas/metabolismo , Actomiosina/metabolismo , Trifosfato de Adenosina/metabolismo , Hidrólise , Isoenzimas/metabolismo , Cinética , Cadeias Leves de Miosina/química , Subfragmentos de Miosina/metabolismo , Humanos , Animais
2.
J Biol Chem ; 300(1): 105565, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38103642

RESUMO

The biochemical SRX (super-relaxed) state of myosin has been defined as a low ATPase activity state. This state can conserve energy when the myosin is not recruited for muscle contraction. The SRX state has been correlated with a structurally defined ordered (versus disordered) state of muscle thick filaments. The two states may be linked via a common interacting head motif (IHM) where the two heads of heavy meromyosin (HMM), or myosin, fold back onto each other and form additional contacts with S2 and the thick filament. Experimental observations of the SRX, IHM, and the ordered form of thick filaments, however, do not always agree, and result in a series of unresolved paradoxes. To address these paradoxes, we have reexamined the biochemical measurements of the SRX state for porcine cardiac HMM. In our hands, the commonly employed mantATP displacement assay was unable to quantify the population of the SRX state with all data fitting very well by a single exponential. We further show that mavacamten inhibits the basal ATPases of both porcine ventricle HMM and S1 (Ki, 0.32 and 1.76 µM respectively) while dATP activates HMM cooperatively without any evidence of an SRX state. A combination of our experimental observations and theories suggests that the displacement of mantATP in purified proteins is not a reliable assay to quantify the SRX population. This means that while the structurally defined IHM and ordered thick filaments clearly exist, great care must be employed when using the mantATP displacement assay.


Assuntos
Trifosfato de Adenosina , Ensaios Enzimáticos , Miosina não Muscular Tipo IIA , Suínos , ortoaminobenzoatos , Animais , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Benzilaminas/farmacologia , Ensaios Enzimáticos/métodos , Ensaios Enzimáticos/normas , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Ventrículos do Coração/metabolismo , Contração Miocárdica , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/metabolismo , ortoaminobenzoatos/metabolismo , Uracila/análogos & derivados , Uracila/farmacologia
3.
Arch Biochem Biophys ; 747: 109753, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37714251

RESUMO

The MF30 monoclonal antibody, which binds to the myosin subfragment-2 (S2), was found to increase the extent of myofibril shortening. Yet, previous observations found no effect of this antibody on actin sliding over myosin during in vitro motility assays with purified proteins in which myosin binding protein C (MyBPC) was absent. MF30 is hypothesized to enhance the availability of myosin heads (subfragment-1 or S1) to bind actin by destabilizing the myosin S2 coiled-coil and sterically blocking S2 from binding S1. The mechanism of action likely includes MF30's substantial size, thereby inhibiting S1 heads and MyBPC from binding S2. Hypothetically, MF30 should enhance the ON state of myosin, thereby increasing muscle contraction. Our findings indicate that MF30 binds preferentially to the unfolded heavy chains of S2, displaying positive cooperativity. However, the dose-response curve of MF30's enhancement of myofibril shortening did not suggest complex interactions with S2. Single, double, and triple-stained myofibrils with increasing amounts of antibodies against myosin rods indicate a possible competition with MyBPC. Additional assays revealed decreased fluorescence intensity at the C-zone (central zone in the sarcomere, where MyBPC is located), where MyBPC may inhibit MF30 binding. Another monoclonal antibody named MF20, which binds to the light meromyosin (LMM) without affecting myofibril contraction, showed less reduction in fluorescence intensity at the C-zone in expansion microscopy than MF30. Expansion microscopy images of myofibrils labeled with MF20 revealed labeling of the A-band (anisotropic band) and a slight reduction in the labeling at the C-zone. The staining pattern obtained from the expansion microscopy image was consistent with images from photolocalization microscopy which required the synthesis of unique photoactivatable quantum dots, and Zeiss Airyscan imaging as well as alternative expansion microscopy digestion methods. Consistent with the hypothesis that MF30 competes with MyBPC binding to S2, cardiac tissue from MyBPC knockout mice was stained more intensely, especially in the C-zone, by MF30 compared to the wild type.


Assuntos
Actinas , Microscopia , Animais , Camundongos , Actinas/metabolismo , Ligação Competitiva , Miosinas/metabolismo , Subfragmentos de Miosina/metabolismo , Anticorpos Monoclonais
4.
J Biol Chem ; 296: 100114, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33234590

RESUMO

A hallmark feature of myosin-II is that it can spontaneously self-assemble into bipolar synthetic thick filaments (STFs) in low-ionic-strength buffers, thereby serving as a reconstituted in vitro model for muscle thick filaments. Although these STFs have been extensively used for structural characterization, their functional evaluation has been limited. In this report, we show that myosins in STFs mirror the more electrostatic and cooperative interactions that underlie the energy-sparing super-relaxed (SRX) state, which are not seen using shorter myosin subfragments, heavy meromyosin (HMM) and myosin subfragment 1 (S1). Using these STFs, we show several pathophysiological insults in hypertrophic cardiomyopathy, including the R403Q myosin mutation, phosphorylation of myosin light chains, and an increased ADP:ATP ratio, destabilize the SRX population. Furthermore, WT myosin containing STFs, but not S1, HMM, or STFs-containing R403Q myosin, recapitulated the ADP-induced destabilization of the SRX state. Studies involving a clinical-stage small-molecule inhibitor, mavacamten, showed that it is more effective in not only increasing myosin SRX population in STFs than in S1 or HMM but also in increasing myosin SRX population equally well in STFs made of healthy and disease-causing R403Q myosin. Importantly, we also found that pathophysiological perturbations such as elevated ADP concentration weakens mavacamten's ability to increase the myosin SRX population, suggesting that mavacamten-bound myosin heads are not permanently protected in the SRX state but can be recruited into action. These findings collectively emphasize that STFs serve as a valuable tool to provide novel insights into the myosin SRX state in healthy, diseased, and therapeutic conditions.


Assuntos
Benzilaminas/química , Benzilaminas/metabolismo , Miosinas/metabolismo , Uracila/análogos & derivados , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Músculo Esquelético/metabolismo , Contração Miocárdica/fisiologia , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Miosinas/química , Fosforilação/fisiologia , Uracila/química , Uracila/metabolismo
5.
J Biol Chem ; 296: 100471, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33639160

RESUMO

Actin-myosin mediated contractile forces are crucial for many cellular functions, including cell motility, cytokinesis, and muscle contraction. We determined the effects of ten actin-binding compounds on the interaction of cardiac myosin subfragment 1 (S1) with pyrene-labeled F-actin (PFA). These compounds, previously identified from a small-molecule high-throughput screen (HTS), perturb the structural dynamics of actin and the steady-state actin-activated myosin ATPase activity. However, the mechanisms underpinning these perturbations remain unclear. Here we further characterize them by measuring their effects on PFA fluorescence, which is decreased specifically by the strong binding of myosin to actin. We measured these effects under equilibrium and steady-state conditions, and under transient conditions, in stopped-flow experiments following addition of ATP to S1-bound PFA. We observed that these compounds affect early steps of the myosin ATPase cycle to different extents. They increased the association equilibrium constant K1 for the formation of the strongly bound collision complex, indicating increased ATP affinity for actin-bound myosin, and decreased the rate constant k+2 for subsequent isomerization to the weakly bound ternary complex, thus slowing the strong-to-weak transition that actin-myosin interaction undergoes early in the ATPase cycle. The compounds' effects on actin structure allosterically inhibit the kinetics of the actin-myosin interaction in ways that may be desirable for treatment of hypercontractile forms of cardiomyopathy. This work helps to elucidate the mechanisms of action for these compounds, several of which are currently used therapeutically, and sets the stage for future HTS campaigns that aim to discover new drugs for treatment of heart failure.


Assuntos
Actinas/química , Actinas/metabolismo , Miosinas Cardíacas/metabolismo , Actinas/efeitos dos fármacos , Adenosina Trifosfatases/efeitos dos fármacos , Adenosina Trifosfatases/metabolismo , Animais , Miosinas Cardíacas/efeitos dos fármacos , Miosinas Cardíacas/fisiologia , Bovinos , Fluorescência , Ensaios de Triagem em Larga Escala/métodos , Cinética , Contração Muscular/fisiologia , Subfragmentos de Miosina/efeitos dos fármacos , Subfragmentos de Miosina/metabolismo , Miosinas/efeitos dos fármacos , Miosinas/metabolismo , Física , Ligação Proteica , Pirenos/química , Coelhos , Bibliotecas de Moléculas Pequenas/farmacologia
6.
Arch Biochem Biophys ; 726: 109240, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35667907

RESUMO

Rabbit cardiac myosin contains fewer cysteine residues than the skeletal myosin (7 and 8.8 moles/105 gm. of myosin, respectively). A similar difference is found between the cysteine content of rabbit cardiac and skeletal heavy meromyosins; the cardiac heavy meromyosin contains 8.9 moles/105 gm. of protein as compared to 11 moles in the skeletal heavy meromyosin. In skeletal myosin, actomyosin, and myofibrils the Ca++-ATPase, Ca++-ITPase, and EDTA-ATPase activities are about three times higher than those of cardiac myosin, actomyosin, and myofibrils; whereas the skeletal to cardiac actomyosin-ATPase activity ratio is higher. The ATPase activities of both cardiac and skeletal myosins, actomyosins, and myofibrils, however, are close to each other when determined in the presence of Mg++ at high ionic strength. The abilities of cardiac and skeletal myosins to combine with actin at either high or low ionic strength are also essentially the same. The Ca++-ATPase, Ca++-ITPase, EDTA-ATPase, and actomyosin-ATPase activities of cardiac myosin, heavy meromyosin, and myofibrils, unlike those of skeletal myosin, heavy meromyosin, and myofibrils, do not increase over pH 8.0. The ATPase activities of cardiac and skeletal myosins in the presence of Mg++ at high ionic strength, on the other hand, are affected similarly by changes of pH. In cardiac myosin, heavy meromyosin, and myofibrils, the Ca++activated ATPase is less sensitive to high KC1 concentrations than is that of skeletal myosin, heavy meromyosin, and myofibrils.


Assuntos
Actomiosina , Subfragmentos de Miosina , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Miosinas Cardíacas , Cisteína , Miofibrilas/metabolismo , Subfragmentos de Miosina/metabolismo , Miosinas/metabolismo , Coelhos
7.
Proc Natl Acad Sci U S A ; 116(33): 16384-16393, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31358631

RESUMO

High-speed atomic force microscopy (HS-AFM) can be used to study dynamic processes with real-time imaging of molecules within 1- to 5-nm spatial resolution. In the current study, we evaluated the 3-state model of activation of cardiac thin filaments (cTFs) isolated as a complex and deposited on a mica-supported lipid bilayer. We studied this complex for dynamic conformational changes 1) at low and high [Ca2+] (pCa 9.0 and 4.5), and 2) upon myosin binding to the cTF in the nucleotide-free state or in the presence of ATP. HS-AFM was used to directly visualize the tropomyosin-troponin complex and Ca2+-induced tropomyosin movements accompanied by structural transitions of actin monomers within cTFs. Our data show that cTFs at relaxing or activating conditions are not ultimately in a blocked or activated state, respectively, but rather the combination of states with a prevalence that is dependent on the [Ca2+] and the presence of weakly or strongly bound myosin. The weakly and strongly bound myosin induce similar changes in the structure of cTFs as confirmed by the local dynamical displacement of individual tropomyosin strands in the center of a regulatory unit of cTF at the relaxed and activation conditions. The displacement of tropomyosin at the relaxed conditions had never been visualized directly and explains the ability of myosin binding to TF at the relaxed conditions. Based on the ratios of nonactivated and activated segments within cTFs, we proposed a mechanism of tropomyosin switching from different states that includes both weakly and strongly bound myosin.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Actinas/ultraestrutura , Subfragmentos de Miosina/ultraestrutura , Tropomiosina/ultraestrutura , Troponina/ultraestrutura , Citoesqueleto de Actina/química , Actinas/química , Animais , Cálcio/metabolismo , Bicamadas Lipídicas/química , Modelos Moleculares , Imagem Molecular , Contração Muscular/genética , Músculo Esquelético/química , Músculo Esquelético/ultraestrutura , Miocárdio/química , Miocárdio/ultraestrutura , Subfragmentos de Miosina/química , Miosinas/química , Ligação Proteica , Coelhos , Sarcômeros/química , Sarcômeros/ultraestrutura , Tropomiosina/química , Troponina/química
8.
Proc Natl Acad Sci U S A ; 116(24): 11731-11736, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31142654

RESUMO

Phosphorylation of cardiac myosin binding protein-C (cMyBP-C) accelerates cardiac contractility. However, the mechanisms by which cMyBP-C phosphorylation increases contractile kinetics have not been fully elucidated. In this study, we tested the hypothesis that phosphorylation of cMyBP-C releases myosin heads from the inhibited super-relaxed state (SRX), thereby determining the fraction of myosin available for contraction. Mice with various alanine (A) or aspartic acid (D) substitutions of the three main phosphorylatable serines of cMyBP-C (serines 273, 282, and 302) were used to address the association between cMyBP-C phosphorylation and SRX. Single-nucleotide turnover in skinned ventricular preparations demonstrated that phosphomimetic cMyBP-C destabilized SRX, whereas phospho-ablated cMyBP-C had a stabilizing effect on SRX. Strikingly, phosphorylation at serine 282 site was found to play a critical role in regulating the SRX. Treatment of WT preparations with protein kinase A (PKA) reduced the SRX, whereas, in nonphosphorylatable cMyBP-C preparations, PKA had no detectable effect. Mice with stable SRX exhibited reduced force production. Phosphomimetic cMyBP-C with reduced SRX exhibited increased rates of tension redevelopment and reduced binding to myosin. We also used recombinant myosin subfragment-2 to disrupt the endogenous interaction between cMyBP-C and myosin and observed a significant reduction in the population of SRX myosin. This peptide also increased force generation and rate of tension redevelopment in skinned fibers. Taken together, this study demonstrates that the phosphorylation-dependent interaction between cMyBP-C and myosin is a determinant of the fraction of myosin available for contraction. Furthermore, the binding between cMyBP-C and myosin may be targeted to improve contractile function.


Assuntos
Miosinas Cardíacas/metabolismo , Proteínas de Transporte/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fosforilação/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas do Citoesqueleto/metabolismo , Cinética , Camundongos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Subfragmentos de Miosina/metabolismo , Sarcômeros/metabolismo
9.
Biophys J ; 120(11): 2222-2236, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33864791

RESUMO

Cardiac muscle contraction is driven by the molecular motor myosin, which uses the energy from ATP hydrolysis to generate a power stroke when interacting with actin filaments, although it is unclear how this mechanism is impaired by mutations in myosin that can lead to heart failure. We have applied a fluorescence resonance energy transfer (FRET) strategy to investigate structural changes in the lever arm domain of human ß-cardiac myosin subfragment 1 (M2ß-S1). We exchanged the human ventricular regulatory light chain labeled at a single cysteine (V105C) with Alexa 488 onto M2ß-S1, which served as a donor for Cy3ATP bound to the active site. We monitored the FRET signal during the actin-activated product release steps using transient kinetic measurements. We propose that the fast phase measured with our FRET probes represents the macroscopic rate constant associated with actin-activated rotation of the lever arm during the power stroke in M2ß-S1. Our results demonstrated M2ß-S1 has a slower actin-activated power stroke compared with fast skeletal muscle myosin and myosin V. Measurements at different temperatures comparing the rate constants of the actin-activated power stroke and phosphate release are consistent with a model in which the power stroke occurs before phosphate release and the two steps are tightly coupled. We suggest that the actin-activated power stroke is highly reversible but followed by a highly irreversible phosphate release step in the absence of load and free phosphate. We demonstrated that hypertrophic cardiomyopathy (R723G)- and dilated cardiomyopathy (F764L)-associated mutations both reduced actin activation of the power stroke in M2ß-S1. We also demonstrate that both mutations alter in vitro actin gliding in the presence and absence of load. Thus, examining the structural kinetics of the power stroke in M2ß-S1 has revealed critical mutation-associated defects in the myosin ATPase pathway, suggesting these measurements will be extremely important for establishing structure-based mechanisms of contractile dysfunction.


Assuntos
Actinas , Cardiomiopatias , Actinas/genética , Trifosfato de Adenosina , Miosinas Cardíacas , Humanos , Mutação , Subfragmentos de Miosina
10.
J Muscle Res Cell Motil ; 42(2): 137-147, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32929610

RESUMO

Two single mutations, R694N and E45Q, were introduced in the beta isoform of human cardiac myosin to remove permanent salt bridges E45:R694 and E98:R694 in the SH1-SH2 helix of the myosin head. Beta isoform-specific bridges E45:R694 and E98:R694 were discovered in the molecular dynamics simulations of the alpha and beta myosin isoforms. Alpha and beta isoforms exhibit different kinetics, ADP dissociates slower from actomyosin containing beta myosin isoform, therefore, beta myosin stays strongly bound to actin longer. We hypothesize that the electrostatic interactions in the SH1-SH2 helix modulate the affinity of ADP to actomyosin, and therefore, the time of the strong actomyosin binding. Wild type and the mutants of the myosin head construct (1-843 amino acid residues) were expressed in differentiated C2C12 cells, and the duration of the strongly bound state of actomyosin was characterized using transient kinetics spectrophotometry. All myosin constructs exhibited a fast rate of ATP binding to actomyosin and a slow rate of ADP dissociation, showing that ADP release limits the time of the strongly bound state of actomyosin. The mutant R694N showed a faster rate of ADP release from actomyosin, compared to the wild type and the E45Q mutant, thus indicating that electrostatic interactions within the SH1-SH2 helix region of human cardiac myosin modulate ADP release and thus, the duration of the strongly bound state of actomyosin.


Assuntos
Actomiosina , Miosinas Cardíacas , Actinas/metabolismo , Actomiosina/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina , Miosinas Cardíacas/genética , Humanos , Cinética , Subfragmentos de Miosina/metabolismo , Ligação Proteica , Eletricidade Estática
11.
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
12.
Biochemistry ; 59(50): 4725-4734, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33290064

RESUMO

The phosphorylated and unphosphorylated forms of tropomyosin Tpm1.1(α) are prepared from adult rabbit heart and compared biochemically. Electrophoresis confirms the high level of enrichment of the chromatography fractions and is consistent with a single site of phosphorylation. Covalently bound phosphate groups at position 283 of Tpm1.1(α) increase the rate of digestion at Leu-169, suggestive of a conformational rearrangement that extends to the midregion. Such a rearrangement, which is supported by ellipticity measurements between 25 and 42 °C, is consistent with a phosphorylation-mediated tightening of the interaction between various myofilament components. In a nonradioactive, co-sedimentation assay [30 mM KCl, 1 mM Mg(II), and 4 °C], phosphorylated Tpm1.1(α) displays a higher affinity for F-actin compared to that of the unphosphorylated control (Kd, 0.16 µM vs 0.26 µM). Phosphorylation decreases the concentration of thin filaments (pCa 4 plus ATP) required to attain a half-maximal rate of release of product from a pre-power stroke complex [myosin-S1-2-deoxy-3-O-(N-methylanthraniloyl)ADP-Pi], as investigated by double-mixing stopped-flow fluorescence, suggestive of a change in the proportion of active (turned on) and inactive (turned off) conformers, but similar maximum rates of product release are observed with either type of reconstituted thin filament. Phosphorylated thin filaments (pCa 4 and 8) display a higher affinity for myosin-S1(ADP) versus the control scenario without affecting isotherm steepness. Specific activities of ATP and Tpm1.1(α) are determined during an in vitro incubation of rat cardiac tissue [12 day-old, 50% phosphorylated Tpm1.1(α)] with [32P]orthophosphate. The incorporation of an isotope into tropomyosin lags behind that of ATP by a factor of approximately 10, indicating that transfer is a comparatively slow process.


Assuntos
Tropomiosina/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Técnicas In Vitro , Cinética , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Miocárdio/química , Miocárdio/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Fosforilação , Conformação Proteica , Estabilidade Proteica , Proteólise , Coelhos , Ratos , Serina/química , Tropomiosina/metabolismo , Troponina/química , Troponina/metabolismo
13.
Biophys J ; 116(12): 2246-2252, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31126584

RESUMO

Striated muscle contraction occurs when myosin thick filaments bind to thin filaments in the sarcomere and generate pulling forces. This process is regulated by calcium, and it can be perturbed by pathological conditions (e.g., myopathies), physiological adaptations (e.g., ß-adrenergic stimulation), and pharmacological interventions. Therefore, it is important to have a methodology to robustly determine the impact of these perturbations and statistically evaluate their effects. Here, we present an approach to measure the equilibrium constants that govern muscle activation, estimate uncertainty in these parameters, and statistically test the effects of perturbations. We provide a MATLAB-based computational tool for these analyses, along with easy-to-follow tutorials that make this approach accessible. The hypothesis testing and error estimation approaches described here are broadly applicable, and the provided tools work with other types of data, including cellular measurements. To demonstrate the utility of the approach, we apply it to elucidate the biophysical mechanism of a mutation that causes familial hypertrophic cardiomyopathy. This approach is generally useful for studying muscle diseases and therapeutic interventions that target muscle contraction.


Assuntos
Biologia Computacional , Cardiopatias/fisiopatologia , Músculos/fisiopatologia , Cardiomiopatia Hipertrófica/genética , Cardiopatias/genética , Cardiopatias/patologia , Modelos Cardiovasculares , Músculos/patologia , Mutação , Subfragmentos de Miosina/metabolismo , Incerteza
14.
Biophys J ; 123(5): 525-526, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38297835
15.
Biochem Biophys Res Commun ; 515(2): 372-377, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31155291

RESUMO

Substitution of Ala for Thr residue in 155th position in γ-tropomyosin (Tpm3.12) is associated with muscle weakness. To understand the mechanisms of this defect, we studied the Ca2+-sensitivity of thin filaments in solution and multistep changes in mobility and spatial arrangement of actin, Tpm, and myosin heads during the ATPase cycle in reconstituted muscle fibres, using the polarized fluorescence microscopy. It was shown that the Ala155Thr (A155T) mutation increased the Ca2+-sensitivity of the thin filaments in solution. In the absence of the myosin heads in the muscle fibres, the mutation did not alter the ability of troponin to switch the thin filaments on and off at high and low Ca2+, respectively. However, upon the binding of myosin heads to the thin filaments at low Ca2+, the mutant Tpm was found to be markedly closer to the open position, than the wild-type Tpm. In the presence of the mutant Tpm, switching on of actin monomers and formation of the strong-binding state of the myosin heads were observed at low Ca2+, which indicated a higher myofilament Ca2+-sensitivity. The mutation decreased the amount of myosin heads bound strongly to actin at high Ca2+ and increased the number of these heads at relaxation. It is suggested that direct binding of myosin to Tpm may be one оf the reasons for muscle weakness associated with the A155T mutation. The use of reagents that decrease the Ca2+-sensitivity of the troponin complex may not be adequate to restore muscle function in patients with the A155T mutation.


Assuntos
Cálcio/metabolismo , Debilidade Muscular/genética , Debilidade Muscular/fisiopatologia , Tropomiosina/genética , Tropomiosina/fisiologia , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Substituição de Aminoácidos , Animais , Polarização de Fluorescência , Humanos , Técnicas In Vitro , Masculino , Debilidade Muscular/etiologia , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Mutação de Sentido Incorreto , Miofibrilas/metabolismo , Subfragmentos de Miosina/metabolismo , Coelhos , Tropomiosina/química , Troponina/metabolismo
16.
Arch Biochem Biophys ; 661: 168-177, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30465737

RESUMO

BACKGROUND: The mechanical work and the actin-activated ATP kinetics in skeletal muscles are closely associated with two surface loops that are present in the myosin molecule: loop 1 and loop 2. They are located close to the ATP-loop (loop 1), and the actin binding domain (loop 2). In this study we investigated the roles of loops 1 and 2 in the regulation of the load-dependent velocity of actin sliding and ATPase activity. METHODS: Heavy meromyosin (HMM) from rabbit skeletal muscle was subjected to limited tryptic proteolysis to obtain fragments containing different amounts of loops 1 and 2. The amino-acid sequences of these fragments were confirmed with quantitative mass-spectrometry. The velocity of actin motility propelled by the HMM fragments was measured using in-vitro motility assays, with varying loads induced by the addition of different concentrations of α-actinin. RESULTS: The load-dependent velocity of the myosin-propelled actin motility, and the fraction of actin filaments motility, were decreased in close association with the depletion of loop 1 in the HMM. The ATPase activity was decreased in close association with depletion of loops 1 and 2. CONCLUSIONS: Loop 1 is responsible for regulating the load-dependent velocity of actin motility. GENERAL SIGNIFICANCE: Myosin-actin interaction is closely regulated by two flexible loops in the structure of myosin. The results of this study are important for the understanding of the molecular mechanisms of contraction, and therefore the most basic functions of life, such as locomotion, heart beating, and breathing.


Assuntos
Músculo Esquelético/metabolismo , Subfragmentos de Miosina/metabolismo , Proteólise , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Modelos Moleculares , Movimento , Músculo Esquelético/fisiologia , Subfragmentos de Miosina/química , Conformação Proteica , Coelhos , Tripsina/metabolismo , Suporte de Carga
17.
Nanotechnology ; 30(21): 214003, 2019 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-30699399

RESUMO

Due to their high aspect ratio and increased surface-to-foot-print area, arrays of vertical semiconductor nanowires are used in numerous biological applications, such as cell transfection and biosensing. Here we focus on two specific valuable biosensing approaches that, so far, have received relatively limited attention in terms of their potential capabilities: cellular mechanosensing and lightguiding-induced enhanced fluorescence detection. Although proposed a decade ago, these two applications for using vertical nanowire arrays have only very recently achieved significant breakthroughs, both in terms of understanding their fundamental phenomena, and in the ease of their implementation. We review the status of the field in these areas and describe significant findings and potential future directions.


Assuntos
Técnicas Biossensoriais , Mecanotransdução Celular/fisiologia , Nanofios/química , Semicondutores , Biomarcadores/urina , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Interleucina-8/urina , Luz , Células MCF-7 , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Nanofios/ultraestrutura , Espectrometria de Fluorescência/métodos , Fator de Necrose Tumoral alfa/urina , Xylella/citologia , Xylella/fisiologia , Óxido de Zinco/química
18.
PLoS Genet ; 12(7): e1006195, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27447488

RESUMO

Cytokinesis requires the spatio-temporal coordination of membrane deposition and primary septum (PS) formation at the division site to drive acto-myosin ring (AMR) constriction. It has been demonstrated that AMR constriction invariably occurs only after the mitotic spindle disassembly. It has also been established that Chitin Synthase II (Chs2p) neck localization precedes mitotic spindle disassembly during mitotic exit. As AMR constriction depends upon PS formation, the question arises as to how chitin deposition is regulated so as to prevent premature AMR constriction and mitotic spindle breakage. In this study, we propose that cells regulate the coordination between spindle disassembly and AMR constriction via timely endocytosis of cytokinetic enzymes, Chs2p, Chs3p, and Fks1p. Inhibition of endocytosis leads to over accumulation of cytokinetic enzymes during mitotic exit, which accelerates the constriction of the AMR, and causes spindle breakage that eventually could contribute to monopolar spindle formation in the subsequent round of cell division. Intriguingly, the mitotic spindle breakage observed in endocytosis mutants can be rescued either by deleting or inhibiting the activities of, CHS2, CHS3 and FKS1, which are involved in septum formation. The findings from our study highlight the importance of timely endocytosis of cytokinetic enzymes at the division site in safeguarding mitotic spindle integrity during mitotic exit.


Assuntos
Quitina Sintase/genética , Equinocandinas/genética , Endocitose/genética , Glucosiltransferases/genética , Proteínas de Membrana/genética , Mitose/genética , Proteínas de Saccharomyces cerevisiae/genética , Ciclo Celular/genética , Membrana Celular/genética , Quitina/genética , Citocinese/genética , Microscopia de Fluorescência , Subfragmentos de Miosina/genética , Saccharomyces cerevisiae/genética , Fuso Acromático/genética
19.
Proc Natl Acad Sci U S A ; 113(6): 1558-63, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26831109

RESUMO

Mutations in genes encoding myosin, the molecular motor that powers cardiac muscle contraction, and its accessory protein, cardiac myosin binding protein C (cMyBP-C), are the two most common causes of hypertrophic cardiomyopathy (HCM). Recent studies established that the N-terminal domains (NTDs) of cMyBP-C (e.g., C0, C1, M, and C2) can bind to and activate or inhibit the thin filament (TF). However, the molecular mechanism(s) by which NTDs modulate interaction of myosin with the TF remains unknown and the contribution of each individual NTD to TF activation/inhibition is unclear. Here we used an integrated structure-function approach using cryoelectron microscopy, biochemical kinetics, and force measurements to reveal how the first two Ig-like domains of cMyPB-C (C0 and C1) interact with the TF. Results demonstrate that despite being structural homologs, C0 and C1 exhibit different patterns of binding on the surface of F-actin. Importantly, C1 but not C0 binds in a position to activate the TF by shifting tropomyosin (Tm) to the "open" structural state. We further show that C1 directly interacts with Tm and traps Tm in the open position on the surface of F-actin. Both C0 and C1 compete with myosin subfragment 1 for binding to F-actin and effectively inhibit actomyosin interactions when present at high ratios of NTDs to F-actin. Finally, we show that in contracting sarcomeres, the activating effect of C1 is apparent only once low levels of Ca(2+) have been achieved. We suggest that Ca(2+) modulates the interaction of cMyBP-C with the TF in the sarcomere.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Hidrólise , Modelos Moleculares , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Contração Muscular/efeitos dos fármacos , Subfragmentos de Miosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Coelhos , Sarcômeros/efeitos dos fármacos , Sarcômeros/metabolismo , Relação Estrutura-Atividade , Sus scrofa
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA