Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(13): e2401625121, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38507449

RESUMO

Molecular motors employ chemical energy to generate unidirectional mechanical output against a track while navigating a chaotic cellular environment, potential disorder on the track, and against Brownian motion. Nevertheless, decades of nanometer-precise optical studies suggest that myosin-5a, one of the prototypical molecular motors, takes uniform steps spanning 13 subunits (36 nm) along its F-actin track. Here, we use high-resolution interferometric scattering microscopy to reveal that myosin takes strides spanning 22 to 34 actin subunits, despite walking straight along the helical actin filament. We show that cumulative angular disorder in F-actin accounts for the observed proportion of each stride length, akin to crossing a river on variably spaced stepping stones. Electron microscopy revealed the structure of the stepping molecule. Our results indicate that both motor and track are soft materials that can adapt to function in complex cellular conditions.


Assuntos
Actinas , Miosina Tipo V , Actinas/química , Miosinas/química , Citoesqueleto de Actina/química , Movimento (Física) , Miosina Tipo V/química
2.
Cell ; 141(5): 786-98, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20510926

RESUMO

Inner ear hair cells detect sound through deflection of mechanosensory stereocilia. Each stereocilium is supported by a paracrystalline array of parallel actin filaments that are packed more densely at the base, forming a rootlet extending into the cell body. The function of rootlets and the molecules responsible for their formation are unknown. We found that TRIOBP, a cytoskeleton-associated protein mutated in human hereditary deafness DFNB28, is localized to rootlets. In vitro, purified TRIOBP isoform 4 protein organizes actin filaments into uniquely dense bundles reminiscent of rootlets but distinct from bundles formed by espin, an actin crosslinker in stereocilia. We generated mutant Triobp mice (Triobp(Deltaex8/Deltaex8)) that are profoundly deaf. Stereocilia of Triobp(Deltaex8/Deltaex8) mice develop normally but fail to form rootlets and are easier to deflect and damage. Thus, F-actin bundling by TRIOBP provides durability and rigidity for normal mechanosensitivity of stereocilia and may contribute to resilient cytoskeletal structures elsewhere.


Assuntos
Citoesqueleto de Actina/metabolismo , Surdez/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Células Ciliadas Auditivas Internas/citologia , Humanos , Mecanotransdução Celular , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular
3.
J Biol Chem ; 299(9): 105143, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37562567

RESUMO

Recent genomic studies reported that 90 to 95% of human genes can undergo alternative splicing, by which multiple isoforms of proteins are synthesized. However, the functional consequences of most of the isoforms are largely unknown. Here, we report a novel alternatively spliced isoform of nonmuscle myosin IIA (NM IIA), called NM IIA2, which is generated by the inclusion of 21 amino acids near the actin-binding region (loop 2) of the head domain of heavy chains. Expression of NM IIA2 is found exclusively in the brain tissue, where it reaches a maximum level at 24 h during the circadian rhythm. The actin-dependent Mg2+-ATPase activity and in vitro motility assays reveal that NM IIA2 lacks its motor activities but localizes with actin filaments in cells. Interestingly, NM IIA2 can also make heterofilaments with NM IIA0 (noninserted isoform of NM IIA) and can retard the in vitro motility of NM IIA, when the two are mixed. Altogether, our findings provide the functional importance of a previously unknown alternatively spliced isoform, NM IIA2, and its potential physiological role in regulating NM IIA activity in the brain.


Assuntos
Processamento Alternativo , Encéfalo , Miosina não Muscular Tipo IIA , Humanos , Actinas/metabolismo , Encéfalo/metabolismo , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIA/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ritmo Circadiano , ATPase de Ca(2+) e Mg(2+)/metabolismo , Especificidade de Órgãos
4.
J Biol Chem ; 299(10): 105243, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37690683

RESUMO

Myosin-7a is an actin-based motor protein essential for vision and hearing. Mutations of myosin-7a cause type 1 Usher syndrome, the most common and severe form of deafblindness in humans. The molecular mechanisms that govern its mechanochemistry remain poorly understood, primarily because of the difficulty of purifying stable intact protein. Here, we recombinantly produce the complete human myosin-7a holoenzyme in insect cells and characterize its biochemical and motile properties. Unlike the Drosophila ortholog that primarily associates with calmodulin (CaM), we found that human myosin-7a utilizes a unique combination of light chains including regulatory light chain, CaM, and CaM-like protein 4. Our results further reveal that CaM-like protein 4 does not function as a Ca2+ sensor but plays a crucial role in maintaining the lever arm's structural-functional integrity. Using our recombinant protein system, we purified two myosin-7a splicing isoforms that have been shown to be differentially expressed along the cochlear tonotopic axis. We show that they possess distinct mechanoenzymatic properties despite differing by only 11 amino acids at their N termini. Using single-molecule in vitro motility assays, we demonstrate that human myosin-7a exists as an autoinhibited monomer and can move processively along actin when artificially dimerized or bound to cargo adaptor proteins. These results suggest that myosin-7a can serve multiple roles in sensory systems such as acting as a transporter or an anchor/force sensor. Furthermore, our research highlights that human myosin-7a has evolved unique regulatory elements that enable precise tuning of its mechanical properties suitable for mammalian auditory functions.


Assuntos
Actinas , Surdocegueira , Miosina VIIa , Humanos , Actinas/metabolismo , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Miosina VIIa/genética , Miosina VIIa/metabolismo , Calmodulina/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo
5.
J Cell Sci ; 134(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589498

RESUMO

The small molecular inhibitor of formin FH2 domains, SMIFH2, is widely used in cell biological studies. It inhibits formin-driven actin polymerization in vitro, but not polymerization of pure actin. It is active against several types of formin from different species. Here, we found that SMIFH2 inhibits retrograde flow of myosin 2 filaments and contraction of stress fibers. We further checked the effect of SMIFH2 on non-muscle myosin 2A and skeletal muscle myosin 2 in vitro, and found that SMIFH2 inhibits activity of myosin ATPase and the ability to translocate actin filaments in the gliding actin in vitro motility assay. Inhibition of non-muscle myosin 2A in vitro required a higher concentration of SMIFH2 compared with that needed to inhibit retrograde flow and stress fiber contraction in cells. We also found that SMIFH2 inhibits several other non-muscle myosin types, including bovine myosin 10, Drosophila myosin 7a and Drosophila myosin 5, more efficiently than it inhibits formins. These off-target inhibitions demand additional careful analysis in each case when solely SMIFH2 is used to probe formin functions. This article has an associated First Person interview with Yukako Nishimura, joint first author of the paper.


Assuntos
Citoesqueleto de Actina , Miosinas , Actinas/genética , Animais , Bovinos , Forminas , Miosinas/genética
6.
Nat Rev Mol Cell Biol ; 13(1): 13-26, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22146746

RESUMO

Cells use molecular motors, such as myosins, to move, position and segregate their organelles. Class V myosins possess biochemical and structural properties that should make them ideal actin-based cargo transporters. Indeed, studies show that class V myosins function as cargo transporters in yeast, moving a range of organelles, such as the vacuole, peroxisomes and secretory vesicles. There is also increasing evidence in vertebrate cells that class V myosins not only tether organelles to actin but also can serve as short-range, point-to-point organelle transporters, usually following long-range, microtubule-dependent organelle transport.


Assuntos
Proteínas de Transporte/metabolismo , Miosina Tipo V/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Actinas/metabolismo , Animais , Transporte Biológico , Dictyostelium/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Camundongos , Organelas/metabolismo , Saccharomyces cerevisiae/metabolismo
7.
J Biol Chem ; 296: 100243, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33372036

RESUMO

Cochlear hair cells each possess an exquisite bundle of actin-based stereocilia that detect sound. Unconventional myosin 15 (MYO15) traffics and delivers critical molecules required for stereocilia development and thus is essential for building the mechanosensory hair bundle. Mutations in the human MYO15A gene interfere with stereocilia trafficking and cause hereditary hearing loss, DFNB3, but the impact of these mutations is not known, as MYO15 itself is poorly characterized. To learn more, we performed a kinetic study of the ATPase motor domain to characterize its mechanochemical cycle. Using the baculovirus-Sf9 system, we purified a recombinant minimal motor domain (S1) by coexpressing the mouse MYO15 ATPase, essential and regulatory light chains that bind its IQ domains, and UNC45 and HSP90A chaperones required for correct folding of the ATPase. MYO15 purified with either UNC45A or UNC45B coexpression had similar ATPase activities (kcat = ∼ 6 s-1 at 20 °C). Using stopped-flow and quenched-flow transient kinetic analyses, we measured the major rate constants describing the ATPase cycle, including ATP, ADP, and actin binding; hydrolysis; and phosphate release. Actin-attached ADP release was the slowest measured transition (∼12 s-1 at 20 °C), although this did not rate-limit the ATPase cycle. The kinetic analysis shows the MYO15 motor domain has a moderate duty ratio (∼0.5) and weak thermodynamic coupling between ADP and actin binding. These findings are consistent with MYO15 being kinetically adapted for processive motility when oligomerized. Our kinetic characterization enables future studies into how deafness-causing mutations affect MYO15 and disrupt stereocilia trafficking necessary for hearing.


Assuntos
Surdez/genética , Chaperonas Moleculares/genética , Miosinas/genética , Estereocílios/genética , Adenosina Trifosfatases/genética , Animais , Surdez/patologia , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Audição/genética , Humanos , Cinética , Camundongos , Mutação/genética , Domínios Proteicos/genética , Estereocílios/patologia
8.
Blood ; 135(21): 1887-1898, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32315395

RESUMO

Megakaryocytes (MKs), the precursor cells for platelets, migrate from the endosteal niche of the bone marrow (BM) toward the vasculature, extending proplatelets into sinusoids, where circulating blood progressively fragments them into platelets. Nonmuscle myosin IIA (NMIIA) heavy chain gene (MYH9) mutations cause macrothrombocytopenia characterized by fewer platelets with larger sizes leading to clotting disorders termed myosin-9-related disorders (MYH9-RDs). MYH9-RD patient MKs have proplatelets with thicker and fewer branches that produce fewer and larger proplatelets, which is phenocopied in mouse Myh9-RD models. Defective proplatelet formation is considered to be the principal mechanism underlying the macrothrombocytopenia phenotype. However, MYH9-RD patient MKs may have other defects, as NMII interactions with actin filaments regulate physiological processes such as chemotaxis, cell migration, and adhesion. How MYH9-RD mutations affect MK migration and adhesion in BM or NMIIA activity and assembly prior to proplatelet production remain unanswered. NMIIA is the only NMII isoform expressed in mature MKs, permitting exploration of these questions without complicating effects of other NMII isoforms. Using mouse models of MYH9-RD (NMIIAR702C+/-GFP+/-, NMIIAD1424N+/-, and NMIIAE1841K+/-) and in vitro assays, we investigated MK distribution in BM, chemotaxis toward stromal-derived factor 1, NMIIA activity, and bipolar filament assembly. Results indicate that different MYH9-RD mutations suppressed MK migration in the BM without compromising bipolar filament formation but led to divergent adhesion phenotypes and NMIIA contractile activities depending on the mutation. We conclude that MYH9-RD mutations impair MK chemotaxis by multiple mechanisms to disrupt migration toward the vasculature, impairing proplatelet release and causing macrothrombocytopenia.


Assuntos
Movimento Celular , Perda Auditiva Neurossensorial/complicações , Megacariócitos/patologia , Mutação , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIA/genética , Trombocitopenia/congênito , Trombocitopenia/patologia , Animais , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Trombocitopenia/complicações , Trombocitopenia/etiologia , Trombocitopenia/metabolismo
9.
Nat Methods ; 14(9): 869-872, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28628128

RESUMO

We improve multiphoton structured illumination microscopy using a nonlinear guide star to determine optical aberrations and a deformable mirror to correct them. We demonstrate our method on bead phantoms, cells in collagen gels, nematode larvae and embryos, Drosophila brain, and zebrafish embryos. Peak intensity is increased (up to 40-fold) and resolution recovered (up to 176 ± 10 nm laterally, 729 ± 39 nm axially) at depths ∼250 µm from the coverslip surface.


Assuntos
Aumento da Imagem/instrumentação , Aumento da Imagem/métodos , Lentes , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Desenho de Equipamento , Análise de Falha de Equipamento , Retroalimentação , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
10.
Adv Exp Med Biol ; 1239: 153-181, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32451859

RESUMO

Myosin 5a is a two-headed myosin that functions as a cargo transporter in cells. To accomplish this task it has evolved several unique structural and kinetic features that allow it to move processively as a single molecule along actin filaments. A plethora of biophysical techniques have been used to elucidate the detailed mechanism of its movement along actin filaments in vitro. This chapter describes how this mechanism was deduced.


Assuntos
Movimento , Miosinas , Imagem Individual de Molécula , Citoesqueleto de Actina , Actinas , Biofísica , Humanos , Cinética , Miosinas/metabolismo
11.
Proc Natl Acad Sci U S A ; 114(32): E6516-E6525, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28739905

RESUMO

Addition of 1 mM ATP substantially reduces the light scattering of solutions of polymerized unphosphorylated nonmuscle myosin IIs (NM2s), and this is reversed by phosphorylation of the regulatory light chain (RLC). It has been proposed that these changes result from substantial depolymerization of unphosphorylated NM2 filaments to monomers upon addition of ATP, and filament repolymerization upon RLC-phosphorylation. We now show that the differences in myosin monomer concentration of RLC-unphosphorylated and -phosphorylated recombinant mammalian NM2A, NM2B, and NM2C polymerized in the presence of ATP are much too small to explain their substantial differences in light scattering. Rather, we find that the decrease in light scattering upon addition of ATP to polymerized unphosphorylated NM2s correlates with the formation of dimers, tetramers, and hexamers, in addition to monomers, an increase in length, and decrease in width of the bare zones of RLC-unphosphorylated filaments. Both effects of ATP addition are reversed by phosphorylation of the RLC. Our data also suggest that, contrary to previous models, assembly of RLC-phosphorylated NM2s at physiological ionic strength proceeds from folded monomers to folded antiparallel dimers, tetramers, and hexamers that unfold and polymerize into antiparallel filaments. This model could explain the dynamic relocalization of NM2 filaments in vivo by dephosphorylation of RLC-phosphorylated filaments, disassembly of the dephosphorylated filaments to folded monomers, dimers, and small oligomers, followed by diffusion of these species, and reassembly of filaments at the new location following rephosphorylation of the RLC.


Assuntos
Trifosfato de Adenosina/química , Modelos Moleculares , Cadeias Pesadas de Miosina/química , Miosina Tipo II/química , Multimerização Proteica , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Camundongos , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Fosforilação
12.
J Biol Chem ; 293(38): 14850-14867, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30087119

RESUMO

Nonmuscle myosin 2 (NM2) has three paralogs in mammals, NM2A, NM2B, and NM2C, which have both unique and overlapping functions in cell migration, formation of cell-cell adhesions, and cell polarity. Their assembly into homo- and heterotypic bipolar filaments in living cells is primarily regulated by phosphorylation of the N-terminally bound regulatory light chain. Here, we present evidence that the equilibrium between these filaments and single NM2A and NM2B molecules can be controlled via S100 calcium-binding protein interactions and phosphorylation at the C-terminal end of the heavy chains. Furthermore, we show that in addition to S100A4, other members of the S100 family can also mediate disassembly of homotypic NM2A filaments. Importantly, these proteins can selectively remove NM2A molecules from heterotypic filaments. We also found that tail phosphorylation (at Ser-1956 and Ser-1975) of NM2B by casein kinase 2, as well as phosphomimetic substitutions at sites targeted by protein kinase C (PKC) and transient receptor potential cation channel subfamily M member 7 (TRPM7), down-regulates filament assembly in an additive fashion. Tail phosphorylation of NM2A had a comparatively minor effect on filament stability. S100 binding and tail phosphorylation therefore preferentially disassemble NM2A and NM2B, respectively. These two distinct mechanisms are likely to contribute to the temporal and spatial sorting of the two NM2 paralogs within heterotypic filaments. The existence of multiple NM2A-depolymerizing S100 paralogs offers the potential for diverse regulatory inputs modulating NM2A filament disassembly in cells and provides functional redundancy under both physiological and pathological conditions.


Assuntos
Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas S100/metabolismo , Animais , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Citoesqueleto/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Mutação , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIB/química , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Células Sf9 , Canais de Cátion TRPM/metabolismo
13.
J Am Chem Soc ; 141(22): 9004-9017, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31117653

RESUMO

Although the α-helix has long been recognized as an all-important element of secondary structure, it generally requires stabilization by tertiary interactions with other parts of a protein's structure. Highly charged single α-helical (SAH) domains, consisting of a high percentage (>75%) of Arg, Lys, and Glu residues, are exceptions to this rule but have been difficult to characterize structurally. Our study focuses on the 68-residue medial tail domain of myosin-VI, which is found to contain a highly ordered α-helical structure extending from Glu-6 to Lys-63. High hydrogen exchange protection factors (15-150), small (ca. 4 Hz) 3 JHNHα couplings, and a near-perfect fit to an ideal model α-helix for its residual dipolar couplings (RDCs), measured in a filamentous phage medium, support the high regularity of this helix. Remarkably, the hydrogen exchange rates are far more homogeneous than the protection factors derived from them, suggesting that for these transiently broken helices the intrinsic exchange rates derived from the amino acid sequence are not appropriate reference values. 15N relaxation data indicate a very high degree of rotational diffusion anisotropy ( D∥/ D⊥ ≈ 7.6), consistent with the hydrodynamic behavior predicted for such a long, nearly straight α-helix. Alignment of the helix by a paramagnetic lanthanide ion attached to its N-terminal region shows a decrease in alignment as the distance from the tagging site increases. This decrease yields a precise measure for the persistence length of 224 ± 10 Å at 20 °C, supporting the idea that the role of the SAH helix is to act as an extension of the myosin-VI lever arm.


Assuntos
Cadeias Pesadas de Miosina/química , Domínios Proteicos , Sequência de Aminoácidos , Animais , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica em alfa-Hélice , Suínos
14.
Proc Natl Acad Sci U S A ; 113(52): E8387-E8395, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27956608

RESUMO

The organization of actomyosin networks lies at the center of many types of cellular motility, including cell polarization and collective cell migration during development and morphogenesis. Myosin-IXa is critically involved in these processes. Using total internal reflection fluorescence microscopy, we resolved actin bundles assembled by myosin-IXa. Electron microscopic data revealed that the bundles consisted of highly ordered lattices with parallel actin polarity. The myosin-IXa motor domains aligned across the network, forming cross-links at a repeat distance of precisely 36 nm, matching the helical repeat of actin. Single-particle image processing resolved three distinct conformations of myosin-IXa in the absence of nucleotide. Using cross-correlation of a modeled actomyosin crystal structure, we identified sites of additional mass, which can only be accounted for by the large insert in loop 2 exclusively found in the motor domain of class IX myosins. We show that the large insert in loop 2 binds calmodulin and creates two coordinated actin-binding sites that constrain the actomyosin interactions generating the actin lattices. The actin lattices introduce orientated tracks at specific sites in the cell, which might install platforms allowing Rho-GTPase-activating protein (RhoGAP) activity to be focused at a definite locus. In addition, the lattices might introduce a myosin-related, force-sensing mechanism into the cytoskeleton in cell polarization and collective cell migration.


Assuntos
Citoesqueleto de Actina/química , Actinas/química , Miosinas/química , Actomiosina/química , Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Calmodulina/química , Movimento Celular , Proteínas Ativadoras de GTPase/química , Humanos , Cinética , Microscopia Eletrônica , Microtúbulos/química , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Espectrometria de Fluorescência
15.
Traffic ; 17(8): 839-59, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26929436

RESUMO

Members of the myosin superfamily are involved in all aspects of eukaryotic life. Their function ranges from the transport of organelles and cargos to the generation of membrane tension, and the contraction of muscle. The diversity of physiological functions is remarkable, given that all enzymatically active myosins follow a conserved mechanoenzymatic cycle in which the hydrolysis of ATP to ADP and inorganic phosphate is coupled to either actin-based transport or tethering of actin to defined cellular compartments. Kinetic capacities and limitations of a myosin are determined by the extent to which actin can accelerate the hydrolysis of ATP and the release of the hydrolysis products and are indispensably linked to its physiological tasks. This review focuses on kinetic competencies that - together with structural adaptations - result in myosins with unique mechanoenzymatic properties targeted to their diverse cellular functions.


Assuntos
Actinas/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Miosinas/metabolismo , Organelas/metabolismo , Animais , Humanos , Cinética
16.
J Biol Chem ; 292(44): 18372-18385, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-28882893

RESUMO

Myosin-5B is a ubiquitous molecular motor that transports cargo vesicles of the endomembrane system in intracellular recycling pathways. Myosin-5B malfunction causes the congenital enteropathy microvillus inclusion disease, underlining its importance in cellular homeostasis. Here we describe the interaction of myosin-5B with F-actin, nucleotides, and the pyrazolopyrimidine compound myoVin-1. We show that single-headed myosin-5B is an intermediate duty ratio motor with a kinetic ATPase cycle that is rate-limited by the release of phosphate. The presence of a second head generates strain and gating in the myosin-5B dimer that alters the kinetic signature by reducing the actin-activated ADP release rate to become rate-limiting. This kinetic transition into a high-duty ratio motor is a prerequisite for the proposed transport function of myosin-5B in cellular recycling pathways. Moreover, we show that the small molecule compound myoVin-1 inhibits the enzymatic and functional activity of myosin-5B in vitro Partial inhibition of the actin-activated steady-state ATPase activity and sliding velocity suggests that caution should be used when probing the effect of myoVin-1 on myosin-5-dependent transport processes in cells.


Assuntos
Citoesqueleto de Actina/metabolismo , Síndromes de Malabsorção/metabolismo , Microvilosidades/patologia , Modelos Moleculares , Mucolipidoses/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Citoesqueleto de Actina/química , Substituição de Aminoácidos , Sítios de Ligação , Biologia Computacional , Dimerização , Inibidores Enzimáticos/farmacologia , Sistemas Inteligentes , Humanos , Cinética , Síndromes de Malabsorção/genética , Microvilosidades/genética , Microvilosidades/metabolismo , Simulação de Acoplamento Molecular , Mucolipidoses/genética , Mutação , Cadeias Pesadas de Miosina/antagonistas & inibidores , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/antagonistas & inibidores , Miosina Tipo V/química , Miosina Tipo V/genética , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Pirazóis/farmacologia , Pirimidinas/farmacologia , Homologia Estrutural de Proteína
17.
J Biol Chem ; 291(14): 7517-26, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26893376

RESUMO

ASAP1 regulates F-actin-based structures and functions, including focal adhesions (FAs) and circular dorsal ruffles (CDRs), cell spreading and migration. ASAP1 function requires its N-terminal BAR domain. We discovered that nonmuscle myosin 2A (NM2A) directly bound the BAR-PH tandem of ASAP1in vitro ASAP1 and NM2A co-immunoprecipitated and colocalized in cells. Knockdown of ASAP1 reduced colocalization of NM2A and F-actin in cells. Knockdown of ASAP1 or NM2A recapitulated each other's effects on FAs, cell migration, cell spreading, and CDRs. The NM2A-interacting BAR domain contributed to ASAP1 control of cell spreading and CDRs. Exogenous expression of NM2A rescued the effect of ASAP1 knockdown on CDRs but ASAP1 did not rescue NM2A knockdown defect in CDRs. Our results support the hypothesis that ASAP1 is a positive regulator of NM2A. Given other binding partners of ASAP1, ASAP1 may directly link signaling and the mechanical machinery of cell migration.


Assuntos
Actomiosina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular/fisiologia , Miosina não Muscular Tipo IIA/metabolismo , Transdução de Sinais/fisiologia , Actomiosina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Miosina não Muscular Tipo IIA/genética , Ligação Proteica , Estrutura Terciária de Proteína
18.
J Biol Chem ; 291(48): 24828-24837, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27697842

RESUMO

Mammalian cells express three Class II nonmuscle myosins (NM): NM2A, NM2B, and NM2C. The three NM2s have well established essential roles in cell motility, adhesion, and cytokinesis and less well defined roles in vesicle transport and other processes that would require association of NM2s with cell membranes. Previous evidence for the mechanism of NM2-membrane association includes direct interaction of NM2s with membrane lipids and indirect interaction by association of NM2s with membrane-bound F-actin or peripheral membrane proteins. Direct binding of NM2s to phosphatidylserine-liposomes, but not to phosphatidylcholine-liposomes, has been reported, but the molecular basis of the interaction between NM2s and acidic phospholipids has not been previously investigated. We now show that filamentous, full-length NM2A, NM2B, and NM2C and monomeric, non-filamentous heavy meromyosin bind to liposomes containing one or more acidic phospholipids (phosphatidylserine, phosphatidylinositol 4,5-diphosphate, and phosphatidylinositol 3,4,5-triphosphate) but do not bind to 100% phosphatidylcholine-liposomes. Binding of NM2s to acidic liposomes occurs predominantly through interaction of the liposomes with the regulatory light chain (RLC) binding site in the myosin heavy chain with concomitant dissociation of the RLC. Phosphorylation of myosin-bound RLC by myosin light chain kinase substantially inhibits binding to liposomes of both filamentous NM2 and non-filamentous heavy meromyosin; the addition of excess unbound RLC, but not excess unbound essential light chain, competes with liposome binding. Consistent with the in vitro data, we show that endogenous and expressed NM2A associates with the plasma membrane of HeLa cells and fibrosarcoma cells independently of F-actin.


Assuntos
Membrana Celular/metabolismo , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Fosfolipídeos/metabolismo , Actinas/química , Actinas/genética , Actinas/metabolismo , Membrana Celular/química , Membrana Celular/genética , Células HeLa , Humanos , Lipossomos/química , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/genética , Miosina Tipo II/química , Miosina Tipo II/genética , Fosfolipídeos/química
19.
Proc Natl Acad Sci U S A ; 111(34): 12390-5, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114250

RESUMO

Unconventional myosin 15 is a molecular motor expressed in inner ear hair cells that transports protein cargos within developing mechanosensory stereocilia. Mutations of myosin 15 cause profound hearing loss in humans and mice; however, the properties of this motor and its regulation within the stereocilia organelle are unknown. To address these questions, we expressed a subfragment 1-like (S1) truncation of mouse myosin 15, comprising the predicted motor domain plus three light-chain binding sites. Following unsuccessful attempts to express functional myosin 15-S1 using the Spodoptera frugiperda (Sf9)-baculovirus system, we discovered that coexpression of the muscle-myosin-specific chaperone UNC45B, in addition to the chaperone heat-shock protein 90 (HSP90) significantly increased the yield of functional protein. Surprisingly, myosin 15-S1 did not bind calmodulin with high affinity. Instead, the IQ domains bound essential and regulatory light chains that are normally associated with class II myosins. We show that myosin 15-S1 is a barbed-end-directed motor that moves actin filaments in a gliding assay (∼ 430 nm · s(-1) at 30 °C), using a power stroke of 7.9 nm. The maximum ATPase rate (k(cat) ∼ 6 s(-1)) was similar to the actin-detachment rate (k(det) = 6.2 s(-1)) determined in single molecule optical trapping experiments, indicating that myosin 15-S1 was rate limited by transit through strongly actin-bound states, similar to other processive myosin motors. Our data further indicate that in addition to folding muscle myosin, UNC45B facilitates maturation of an unconventional myosin. We speculate that chaperone coexpression may be a simple method to optimize the purification of other myosin motors from Sf9 insect cells.


Assuntos
Miosinas/isolamento & purificação , Miosinas/metabolismo , Estereocílios/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Calmodulina/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Chaperonas Moleculares , Dados de Sequência Molecular , Cadeias Leves de Miosina/metabolismo , Subfragmentos de Miosina/genética , Subfragmentos de Miosina/isolamento & purificação , Subfragmentos de Miosina/metabolismo , Miosinas/genética , Pinças Ópticas , Dobramento de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Células Sf9 , Spodoptera
20.
Proc Natl Acad Sci U S A ; 111(18): E1833-42, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753602

RESUMO

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.


Assuntos
Actinas/fisiologia , Cadeias Pesadas de Miosina/fisiologia , Actinas/química , Difosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Fenômenos Biomecânicos , Bovinos , Técnicas In Vitro , Cinética , Microscopia Eletrônica , Microscopia de Fluorescência , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Subfragmentos de Miosina/química , Subfragmentos de Miosina/genética , Subfragmentos de Miosina/fisiologia , Pinças Ópticas , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Pseudópodes/fisiologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA