Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 564
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105565, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38103642

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato , Pruebas de Enzimas , Miosina Tipo IIA no Muscular , Porcinos , ortoaminobenzoatos , Animales , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Bencilaminas/farmacología , Pruebas de Enzimas/métodos , Pruebas de Enzimas/normas , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/metabolismo , Contracción Miocárdica , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Miosina Tipo IIA no Muscular/química , Miosina Tipo IIA no Muscular/metabolismo , ortoaminobenzoatos/metabolismo , Uracilo/análogos & derivados , Uracilo/farmacología
2.
J Biol Chem ; 296: 100114, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33234590

RESUMEN

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.


Asunto(s)
Bencilaminas/química , Bencilaminas/metabolismo , Miosinas/metabolismo , Uracilo/análogos & derivados , Adenosina Trifosfato/metabolismo , Animales , Humanos , Músculo Esquelético/metabolismo , Contracción Miocárdica/fisiología , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Miosinas/química , Fosforilación/fisiología , Uracilo/química , Uracilo/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(33): 16384-16393, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31358631

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/ultraestructura , Subfragmentos de Miosina/ultraestructura , Tropomiosina/ultraestructura , Troponina/ultraestructura , Citoesqueleto de Actina/química , Actinas/química , Animales , Calcio/metabolismo , Membrana Dobles de Lípidos/química , Modelos Moleculares , Imagen Molecular , Contracción Muscular/genética , Músculo Esquelético/química , Músculo Esquelético/ultraestructura , Miocardio/química , Miocardio/ultraestructura , Subfragmentos de Miosina/química , Miosinas/química , Unión Proteica , Conejos , Sarcómeros/química , Sarcómeros/ultraestructura , Tropomiosina/química , Troponina/química
4.
Proc Natl Acad Sci U S A ; 115(32): E7486-E7494, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30018063

RESUMEN

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


Asunto(s)
Actinas/metabolismo , Bencilaminas/farmacología , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica Familiar/tratamiento farmacológico , Subfragmentos de Miosina/metabolismo , Uracilo/análogos & derivados , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Regulación Alostérica/efectos de los fármacos , Bencilaminas/uso terapéutico , Miosinas Cardíacas/química , Cardiomiopatía Hipertrófica Familiar/etiología , Humanos , Cinética , Subfragmentos de Miosina/química , Estabilidad Proteica/efectos de los fármacos , Uracilo/farmacología , Uracilo/uso terapéutico
5.
Biochemistry ; 59(50): 4725-4734, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33290064

RESUMEN

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.


Asunto(s)
Tropomiosina/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Técnicas In Vitro , Cinética , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Miocardio/química , Miocardio/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Fosforilación , Conformación Proteica , Estabilidad Proteica , Proteolisis , Conejos , Ratas , Serina/química , Tropomiosina/metabolismo , Troponina/química , Troponina/metabolismo
6.
Arch Biochem Biophys ; 661: 168-177, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30465737

RESUMEN

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.


Asunto(s)
Músculo Esquelético/metabolismo , Subfragmentos de Miosina/metabolismo , Proteolisis , Actinas/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Modelos Moleculares , Movimiento , Músculo Esquelético/fisiología , Subfragmentos de Miosina/química , Conformación Proteica , Conejos , Tripsina/metabolismo , Soporte de Peso
7.
Nanotechnology ; 30(21): 214003, 2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-30699399

RESUMEN

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.


Asunto(s)
Técnicas Biosensibles , Mecanotransducción Celular/fisiología , Nanocables/química , Semiconductores , Biomarcadores/orina , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Interleucina-8/orina , Luz , Células MCF-7 , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Nanocables/ultraestructura , Espectrometría de Fluorescencia/métodos , Factor de Necrosis Tumoral alfa/orina , Xylella/citología , Xylella/fisiología , Óxido de Zinc/química
8.
J Biol Chem ; 292(40): 16571-16577, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28808052

RESUMEN

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.


Asunto(s)
Adenosina Difosfato/química , Adenosina Trifosfato/química , Bencilaminas/química , Miosinas Cardíacas/química , Subfragmentos de Miosina/química , Sarcómeros/química , Uracilo/análogos & derivados , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Miosinas Cardíacas/metabolismo , Cardiomegalia/metabolismo , Bovinos , Subfragmentos de Miosina/metabolismo , Sarcómeros/metabolismo , Uracilo/química
9.
Biochem Biophys Res Commun ; 495(1): 163-167, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29102634

RESUMEN

The molecular mechanism of muscle contraction is based on the ATP-dependent cyclic interaction of myosin heads with actin filaments. Myosin head (myosin subfragment-1, S1) consists of two major domains, the motor domain responsible for ATP hydrolysis and actin binding, and the regulatory domain stabilized by light chains. Essential light chain-1 (LC1) is of particular interest since it comprises a unique N-terminal extension (NTE) which can bind to actin thus forming an additional actin-binding site on the myosin head and modulating its motor activity. However, it remains unknown what happens to the NTE of LC1 when the head binds ATP during ATPase cycle and dissociates from actin. We assume that in this state of the head, when it undergoes global ATP-induced conformational changes, the NTE of LC1 can interact with the motor domain. To test this hypothesis, we applied fluorescence resonance energy transfer (FRET) to measure the distances from various sites on the NTE of LC1 to S1 active site in the motor domain and changes in these distances upon formation of S1-ADP-BeFx complex (stable analog of S1∗-AТP state). For this, we produced recombinant LC1 cysteine mutants, which were first fluorescently labeled with 1,5-IAEDANS (donor) at different positions in their NTE and then introduced into S1; the ADP analog (TNP-ADP) bound to the S1 active site was used as an acceptor. The results show that formation of S1-ADP-BeFx complex significantly decreases the distances from Cys residues in the NTE of LC1 to TNP-ADP in the S1 active site; this effect was the most pronounced for Cys residues located near the LC1 N-terminus. These results support the concept of the ATP-induced transient interaction of the LC1 N-terminus with the S1 motor domain.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Subfragmentos de Miosina/metabolismo , Adenosina Trifosfato/metabolismo , Dominio Catalítico , Transferencia Resonante de Energía de Fluorescencia , Humanos , Modelos Moleculares , Cadenas Ligeras de Miosina/química , Subfragmentos de Miosina/química , Miosinas/metabolismo , Conformación Proteica , Dominios Proteicos
10.
Arch Biochem Biophys ; 638: 41-51, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29229286

RESUMEN

The mechanical stability of the myosin subfragment-2 (S2) was tested with simulated force spectroscopy (SFS) and gravitational force spectroscopy (GFS). Experiments examined unzipping S2, since it required less force than stretching parallel to the coiled coil. Both GFS and SFS demonstrated that the force required to destabilize the light meromyosin (LMM) was greater than the force required to destabilize the coiled coil at each of three different locations along S2. GFS data also conveyed that the mechanical stability of the S2 region is independent from its association with the myosin thick filament using cofilaments of myosin tail and a single intact myosin. The C-terminal end of myosin binding protein C (MyBPC) binds to LMM and the N-terminal end can bind either S2 or actin. The force required to destabilize the myosin coiled coil molecule was 3 times greater in the presence of MyBPC than in its absence. Furthermore, the in vitro motility assay with full length slow skeletal MyBPC slowed down the actin filament sliding over myosin thick filaments. This study demonstrates that skeletal MyBPC both enhanced the mechanical stability of the S2 coiled coil and reduced the sliding velocity of actin filaments over polymerized myosin filaments.


Asunto(s)
Proteínas Portadoras/química , Subfragmentos de Miosina/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Animales , Proteínas Portadoras/metabolismo , Subfragmentos de Miosina/metabolismo , Dominios Proteicos , Estabilidad Proteica , Conejos , Análisis Espectral
11.
Proc Natl Acad Sci U S A ; 112(29): E3806-15, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26150528

RESUMEN

The rod of sarcomeric myosins directs thick filament assembly and is characterized by the insertion of four skip residues that introduce discontinuities in the coiled-coil heptad repeats. We report here that the regions surrounding the first three skip residues share high structural similarity despite their low sequence homology. Near each of these skip residues, the coiled-coil transitions to a nonclose-packed structure inducing local relaxation of the superhelical pitch. Moreover, molecular dynamics suggest that these distorted regions can assume different conformationally stable states. In contrast, the last skip residue region constitutes a true molecular hinge, providing C-terminal rod flexibility. Assembly of myosin with mutated skip residues in cardiomyocytes shows that the functional importance of each skip residue is associated with rod position and reveals the unique role of the molecular hinge in promoting myosin antiparallel packing. By defining the biophysical properties of the rod, the structures and molecular dynamic calculations presented here provide insight into thick filament formation, and highlight the structural differences occurring between the coiled-coils of myosin and the stereotypical tropomyosin. In addition to extending our knowledge into the conformational and biological properties of coiled-coil discontinuities, the molecular characterization of the four myosin skip residues also provides a guide to modeling the effects of rod mutations causing cardiac and skeletal myopathies.


Asunto(s)
Aminoácidos/química , Miosinas Cardíacas/química , Miosinas Cardíacas/metabolismo , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/metabolismo , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Secuencia de Aminoácidos , Humanos , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Docilidad , Estabilidad Proteica , Estructura Secundaria de Proteína , Secuencias Repetitivas de Aminoácido , Sarcómeros/metabolismo , Eliminación de Secuencia , Relación Estructura-Actividad
12.
Proc Natl Acad Sci U S A ; 112(15): 4660-5, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825773

RESUMEN

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.


Asunto(s)
Actomiosina/metabolismo , Miosinas Cardíacas/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Músculo Esquelético/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Actinas/química , Actinas/metabolismo , Actomiosina/química , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Algoritmos , Animales , Miosinas Cardíacas/química , Cinética , Modelos Moleculares , Contracción Muscular , Cadenas Ligeras de Miosina/química , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Conejos , Rotación , Factores de Tiempo
13.
J Struct Biol ; 200(3): 219-228, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28743637

RESUMEN

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.


Asunto(s)
Subfragmentos de Miosina/química , Miosinas Ventriculares/química , Cristalografía por Rayos X , Humanos , Leucina/genética , Modelos Moleculares , Subfragmentos de Miosina/genética , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Miosinas Ventriculares/genética
14.
Bioconjug Chem ; 28(12): 2976-2984, 2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29161498

RESUMEN

The bioconjugation of a polyoxometalate (POMs), i.e., dodecavanadate (V12O32), to DNA strands produces a functional labeled DNA primer, V12O32-DNA. The grafting of DNA primer onto streptavidin-coated magnetic nanoparticles (SVM) produces a novel composite, V12O32-DNA@SVM. The high binding-affinity of V12O32 with the ATP binding site in myosin subfragment-1 (S1) facilitates favorable adsorption of myosin, with an efficiency of 99.4% when processing 0.1 mL myosin solution (100 µg mL-1) using 0.1 mg composite. Myosin adsorption fits the Langmuir model, corresponding to a theoretical adsorption capacity of 613.5 mg g-1. The retained myosin is readily recovered by 1% SDS (m/m), giving rise to a recovery of 58.7%. No conformational change is observed for myosin after eliminating SDS by ultrafiltration. For practical use, high-purity myosin S1 is obtained by separation of myosin from the rough protein extract from porcine left ventricle, followed by digestion with α-chymotryptic and further isolation of S1 subfragment. The purified myosin S1 is identified with matrix-assisted laser desorption/ionization time-of-flight/mass spectrometry, giving rise to a sequence coverage of 38%.


Asunto(s)
ADN/química , Subfragmentos de Miosina/química , Subfragmentos de Miosina/aislamiento & purificación , Compuestos Organometálicos/química , Vanadio/química , Adsorción , Animales , Miocardio/química , Nanopartículas/química , Estreptavidina/química , Porcinos
15.
Eur Biophys J ; 46(2): 195-202, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28039513

RESUMEN

In vitro motility assay (IVMA) experiments were performed to analyze the movement of actin filaments sliding on a pavement of myosin molecules at different [ATP] and [ADP]. In standard experimental conditions at [ATP] = 2 mM, about 80% of the actin filaments move in unloaded conditions with a constant velocity. However, a fraction of at least 20% static actin filaments is always present. The accepted explanation is the occurrence of damaged "rigor"-like myosin heads that do not undergo the normal ATP-dependent cycling motion. However, in a series of IVMA experiments performed at different [ATP] we observed that the mobility of actin filaments increased with lowering [ATP]. We investigated the influence of [ATP] on the number of mobile actin filaments. IVMA experiments were performed at controlled nucleotide concentrations and the percentage of mobile filaments accurately determined by specific operator-guided software. The value of ΔG ATP involved was determined. Results showed that the number of mobile actin filaments sliding on type 2B heavy meromyosin isoform (2B HMM) increased at very low [ATP] accompanied by less negative ΔG ATP values. Similar results were obtained by increasing [ADP]. Performing experiments at the same [ATP] with different myosin types, we found a higher number of mobile actin filaments on slow type 1 HMM with respect to type 2B HMM while the highest number of mobile actin filaments was found on single-head myosin (S1 fraction). We also found that [ATP] did not influence the percentage of mobile actin filaments sliding on S1. Our results reveal novel aspects of actomyosin interaction.


Asunto(s)
Actomiosina/metabolismo , Adenosina Trifosfato/metabolismo , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actomiosina/química , Adenosina Trifosfato/química , Animales , Hidrólisis , Movimiento (Física) , Subfragmentos de Miosina/química , Subfragmentos de Miosina/metabolismo , Unión Proteica , Ratas , Ratas Wistar , Termodinámica
16.
J Chem Phys ; 147(21): 215101, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29221399

RESUMEN

A protein molecule is a dielectric substance, so the binding of a ligand is expected to induce dielectric response in the protein molecule, considering that ligands are charged or polar in general. We previously reported that binding of adenosine triphosphate (ATP) to molecular motor myosin actually induces such a dielectric response in myosin due to the net negative charge of ATP. By this dielectric response, referred to as "dielectric allostery," spatially separated two regions in myosin, the ATP-binding region and the actin-binding region, are allosterically coupled. In this study, from the statistically stringent analyses of the extensive molecular dynamics simulation data obtained in the ATP-free and the ATP-bound states, we show that there exists the dielectric allostery that transmits the signal of ATP binding toward the distant lever-arm region. The ATP-binding-induced electrostatic potential change observed on the surface of the main domain induced a movement of the converter subdomain from which the lever arm extends. The dielectric response was found to be caused by an underlying large-scale concerted rearrangement of the electrostatic bond network, in which highly conserved charged/polar residues are involved. Our study suggests the importance of the dielectric property for molecular machines in exerting their function.


Asunto(s)
Adenosina Trifosfato/metabolismo , Subfragmentos de Miosina/metabolismo , Miosina Tipo II/metabolismo , Pectinidae/metabolismo , Animales , Simulación de Dinámica Molecular , Subfragmentos de Miosina/química , Miosina Tipo II/química , Pectinidae/química , Unión Proteica , Dominios Proteicos , Electricidad Estática
17.
Proc Natl Acad Sci U S A ; 111(18): E1833-42, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753602

RESUMEN

Myosin-10 is an actin-based molecular motor that participates in essential intracellular processes such as filopodia formation/extension, phagocytosis, cell migration, and mitotic spindle maintenance. To study this motor protein's mechano-chemical properties, we used a recombinant, truncated form of myosin-10 consisting of the first 936 amino acids, followed by a GCN4 leucine zipper motif, to force dimerization. Negative-stain electron microscopy reveals that the majority of molecules are dimeric with a head-to-head contour distance of ∼50 nm. In vitro motility assays show that myosin-10 moves actin filaments smoothly with a velocity of ∼310 nm/s. Steady-state and transient kinetic analysis of the ATPase cycle shows that the ADP release rate (∼13 s(-1)) is similar to the maximum ATPase activity (∼12-14 s(-1)) and therefore contributes to rate limitation of the enzymatic cycle. Single molecule optical tweezers experiments show that under intermediate load (∼0.5 pN), myosin-10 interacts intermittently with actin and produces a power stroke of ∼17 nm, composed of an initial 15-nm and subsequent 2-nm movement. At low optical trap loads, we observed staircase-like processive movements of myosin-10 interacting with the actin filament, consisting of up to six ∼35-nm steps per binding interaction. We discuss the implications of this load-dependent processivity of myosin-10 as a filopodial transport motor.


Asunto(s)
Actinas/fisiología , Cadenas Pesadas de Miosina/fisiología , Actinas/química , Adenosina Difosfato/metabolismo , Secuencia de Aminoácidos , Animales , Fenómenos Biomecánicos , Bovinos , Técnicas In Vitro , Cinética , Microscopía Electrónica , Microscopía Fluorescente , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Subfragmentos de Miosina/química , Subfragmentos de Miosina/genética , Subfragmentos de Miosina/fisiología , Pinzas Ópticas , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Seudópodos/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
18.
Biophys J ; 111(7): 1465-1477, 2016 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-27705769

RESUMEN

Actin filaments have key roles in cell motility but are generally claimed to be passive interaction partners in actin-myosin-based motion generation. Here, we present evidence against this static view based on an altered myosin-induced actin filament gliding pattern in an in vitro motility assay at varied [MgATP]. The statistics that characterize the degree of meandering of the actin filament paths suggest that for [MgATP] ≥ 0.25 mM, the flexural rigidity of heavy meromyosin (HMM)-propelled actin filaments is similar (without phalloidin) or slightly lower (with phalloidin) than that of HMM-free filaments observed in solution without surface tethering. When [MgATP] was reduced to ≤0.1 mM, the actin filament paths in the in vitro motility assay became appreciably more winding in both the presence and absence of phalloidin. This effect of lowered [MgATP] was qualitatively different from that seen when HMM was mixed with ATP-insensitive, N-ethylmaleimide-treated HMM (NEM-HMM; 25-30%). In particular, the addition of NEM-HMM increased a non-Gaussian tail in the path curvature distribution as well as the number of events in which different parts of an actin filament followed different paths. These effects were the opposite of those observed with reduced [MgATP]. Theoretical modeling suggests a 30-40% lowered flexural rigidity of the actin filaments at [MgATP] ≤ 0.1 mM and local bending of the filament front upon each myosin head attachment. Overall, the results fit with appreciable structural changes in the actin filament during actomyosin-based motion generation, and modulation of the actin filament mechanical properties by the dominating chemomechanical actomyosin state.


Asunto(s)
Citoesqueleto de Actina/química , Adenosina Trifosfato/química , Subfragmentos de Miosina/química , Citoesqueleto de Actina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico , Elasticidad , Modelos Moleculares , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Subfragmentos de Miosina/metabolismo , Dinámicas no Lineales , Faloidina/química , Conformación Proteica , Soluciones/química
19.
Proteins ; 84(1): 172-189, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26573747

RESUMEN

Sarcomeric myosins have the remarkable ability to form regular bipolar thick filaments that, together with actin thin filaments, constitute the fundamental contractile unit of skeletal and cardiac muscle. This has been established for over 50 years and yet a molecular model for the thick filament has not been attained. In part this is due to the lack of a detailed molecular model for the coiled-coil that constitutes the myosin rod. The ability to self-assemble resides in the C-terminal section of myosin known as light meromyosin (LMM) which exhibits strong salt-dependent aggregation that has inhibited structural studies. Here we evaluate the feasibility of generating a complete model for the myosin rod by combining overlapping structures of five sections of coiled-coil covering 164 amino acid residues which constitute 20% of LMM. Each section contains ∼ 7-9 heptads of myosin. The problem of aggregation was overcome by incorporating the globular folding domains, Gp7 and Xrcc4 which enhance crystallization. The effect of these domains on the stability and conformation of the myosin rod was examined through biophysical studies and overlapping structures. In addition, a computational approach was developed to combine the sections into a contiguous model. The structures were aligned, trimmed to form a contiguous model, and simulated for >700 ns to remove the discontinuities and achieve an equilibrated conformation that represents the native state. This experimental and computational strategy lays the foundation for building a model for the entire myosin rod.


Asunto(s)
Subfragmentos de Miosina/química , Secuencia de Aminoácidos , Cardiomiopatías/genética , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutación , Subfragmentos de Miosina/genética , Conformación Proteica , Estabilidad Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Temperatura
20.
Analyst ; 141(3): 836-46, 2016 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-26617251

RESUMEN

Lab-on-a-chip systems with molecular motor driven transport of analytes attached to cytoskeletal filament shuttles (actin filaments, microtubules) circumvent challenges with nanoscale liquid transport. However, the filaments have limited cargo-carrying capacity and limitations either in transportation speed (microtubules) or control over motility direction (actin). To overcome these constraints we here report incorporation of covalently attached antibodies into self-propelled actin bundles (nanocarriers) formed by cross-linking antibody conjugated actin filaments via fascin, a natural actin-bundling protein. We demonstrate high maximum antigen binding activity and propulsion by surface adsorbed myosin motors. Analyte transport capacity is tested using both protein antigens and microvesicles, a novel class of diagnostic markers. Increased incubation concentration with protein antigen in the 0.1-100 nM range (1 min) reduces the fraction of motile bundles and their velocity but maximum transportation capacity of >1 antigen per nm of bundle length is feasible. At sub-nanomolar protein analyte concentration, motility is very well preserved opening for orders of magnitude improved limit of detection using motor driven concentration on nanoscale sensors. Microvesicle-complexing to monoclonal antibodies on the nanocarriers compromises motility but nanocarrier aggregation via microvesicles shows unique potential in label-free detection with the aggregates themselves as non-toxic reporter elements.


Asunto(s)
Actinas/química , Anticuerpos Monoclonales/química , Proteínas Portadoras/química , Micropartículas Derivadas de Células/química , Inmunoglobulina G/análisis , Proteínas de Microfilamentos/química , Subfragmentos de Miosina/química , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/química , Humanos , Inmunoglobulina G/inmunología , Antígenos Comunes de Leucocito/inmunología , Microscopía Fluorescente , Movimiento (Física) , Conejos , Rodaminas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA