Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nat Rev Mol Cell Biol ; 15(4): 257-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24651543

RESUMEN

Mitotic spindles are self-organizing protein machines that harness teams of multiple force generators to drive chromosome segregation. Kinesins are key members of these force-generating teams. Different kinesins walk directionally along dynamic microtubules, anchor, crosslink, align and sort microtubules into polarized bundles, and influence microtubule dynamics by interacting with microtubule tips. The mechanochemical mechanisms of these kinesins are specialized to enable each type to make a specific contribution to spindle self-organization and chromosome segregation.


Asunto(s)
Segregación Cromosómica/fisiología , Cinesinas/metabolismo , Microtúbulos/metabolismo , Mitosis/fisiología , Huso Acromático/fisiología , Animales , Humanos
2.
Biophys J ; 119(10): 1984-1994, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33091340

RESUMEN

Kinesin-1 is an ATP-driven molecular motor that transports cellular cargo along microtubules. At low loads, kinesin-1 almost always steps forward, toward microtubule plus ends, but at higher loads, it can also step backward. Backsteps are usually 8 nm but can be larger. These larger backward events of 16 nm, 24 nm, or more are thought to be slips rather than steps because they are too fast to consist of multiple, tightly coupled 8-nm steps. Here, we propose that not only these larger backsteps, but all kinesin-1 backsteps, are slips. We show first that kinesin waits before forward steps for less time than before backsteps and detachments; second, we show that kinesin waits for the same amount of time before backsteps and detachments; and third, we show that by varying the microtubule type, we can change the ratio of backsteps to detachments without affecting forward stepping. Our findings indicate that backsteps and detachments originate from the same state and that this state arises later in the mechanochemical cycle than the state that gives rise to forward steps. To explain our data, we propose that, in each cycle of ATP turnover, forward kinesin steps can only occur before Pi release, whereas backslips and detachments can only occur after Pi release. In the scheme we propose, Pi release gates access to a weak binding K⋅ADP-K⋅ADP state that can slip back along the microtubule, re-engage, release ADP, and try again to take an ATP-driven forward step. We predict that this rescued detachment pathway is key to maintaining kinesin processivity under load.


Asunto(s)
Adenosina Trifosfato , Cinesinas , Hidrólisis , Cinesinas/metabolismo , Cinética , Microtúbulos/metabolismo
3.
EMBO Rep ; 19(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206188

RESUMEN

In fission yeast, the lengths of interphase microtubule (iMT) arrays are adapted to cell length to maintain cell polarity and to help centre the nucleus and cell division ring. Here, we show that length regulation of iMTs is dictated by spatially regulated competition between MT-stabilising Tea2/Tip1/Mal3 (Kinesin-7) and MT-destabilising Klp5/Klp6/Mcp1 (Kinesin-8) complexes at iMT plus ends. During MT growth, the Tea2/Tip1/Mal3 complex remains bound to the plus ends of iMT bundles, thereby restricting access to the plus ends by Klp5/Klp6/Mcp1, which accumulate behind it. At cell ends, Klp5/Klp6/Mcp1 invades the space occupied by the Tea2/Tip1/Tea1 kinesin complex triggering its displacement from iMT plus ends and MT catastrophe. These data show that in vivo, whilst an iMT length-dependent model for catastrophe factor accumulation has validity, length control of iMTs is an emergent property reflecting spatially regulated competition between distinct kinesin complexes at the MT plus tip.


Asunto(s)
Cinesinas/metabolismo , Microtúbulos/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/citología , Polaridad Celular , Interfase/fisiología , Cinesinas/genética , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética
4.
Proc Natl Acad Sci U S A ; 113(47): E7483-E7489, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27834216

RESUMEN

Cut7, the sole kinesin-5 in Schizosaccharomyces pombe, is essential for mitosis. Like other yeast kinesin-5 motors, Cut7 can reverse its stepping direction, by mechanisms that are currently unclear. Here we show that for full-length Cut7, the key determinant of stepping direction is the degree of motor crowding on the microtubule lattice, with greater crowding converting the motor from minus end-directed to plus end-directed stepping. To explain how high Cut7 occupancy causes this reversal, we postulate a simple proximity sensing mechanism that operates via steric blocking. We propose that the minus end-directed stepping action of Cut7 is selectively inhibited by collisions with neighbors under crowded conditions, whereas its plus end-directed action, being less space-hungry, is not. In support of this idea, we show that the direction of Cut7-driven microtubule sliding can be reversed by crowding it with non-Cut7 proteins. Thus, crowding by either dynein microtubule binding domain or Klp2, a kinesin-14, converts Cut7 from net minus end-directed to net plus end-directed stepping. Biochemical assays confirm that the Cut7 N terminus increases Cut7 occupancy by binding directly to microtubules. Direct observation by cryoEM reveals that this occupancy-enhancing N-terminal domain is partially ordered. Overall, our data point to a steric blocking mechanism for directional reversal through which collisions of Cut7 motor domains with their neighbors inhibit their minus end-directed stepping action, but not their plus end-directed stepping action. Our model can potentially reconcile a number of previous, apparently conflicting, observations and proposals for the reversal mechanism of yeast kinesins-5.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/citología , Sitios de Unión , Segregación Cromosómica , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis , Dominios Proteicos , Schizosaccharomyces/química , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo
5.
Biophys J ; 110(1): 214-7, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26745424

RESUMEN

Current in vitro optical studies of microtubule dynamics tend to rely on fluorescent labeling of tubulin, with tracking accuracy thereby limited by the quantum yield of fluorophores and by photobleaching. Here, we demonstrate label-free tracking of microtubules with nanometer precision at kilohertz frame rates using interferometric scattering microscopy (iSCAT). With microtubules tethered to a glass substrate using low-density kinesin, we readily detect sequential 8 nm steps in the microtubule center of mass, characteristic of a single kinesin molecule moving a microtubule. iSCAT also permits dynamic changes in filament length to be measured with <5 nm precision. Using the arbitrarily long observation time enabled by label-free iSCAT imaging, we demonstrate continuous monitoring of microtubule disassembly over a 30 min period. The ability of iSCAT to track microtubules with nm precision together with its potential for label-free single protein detection and simultaneous single molecule fluorescence imaging represent a unique platform for novel approaches to studying microtubule dynamics.


Asunto(s)
Microscopía de Interferencia/métodos , Microtúbulos/metabolismo , Animales , Microtúbulos/efectos de los fármacos , Paclitaxel/farmacología , Porcinos , Factores de Tiempo
6.
Biophys J ; 110(5): 1202-1203, 2016 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-31265488

RESUMEN

[This corrects the article DOI: 10.1016/j.bpj.2015.10.055.].

7.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 123-33, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24419385

RESUMEN

Kinesins constitute a superfamily of microtubule-based motor proteins with important cellular functions ranging from intracellular transport to cell division. Some kinesin family members function during the mitotic phase of the eukaryotic cell cycle and are crucial for the successful progression of cell division. In the early stages of mitosis, during prometaphase, certain kinesins are required for the formation of the bipolar spindle, such as Eg5 and Kif15, which seem to possess partially overlapping functions. Because kinesins transform the chemical energy from ATP hydrolysis into mechanical work, inhibition of their function is a tractable approach for drug development. Drugs targeting Eg5 have shown promise as anticancer agents. Kif15 has recently come to the fore because it can substitute the functions of Eg5, and may itself have potential as a prospective drug target. Here, the initial biochemical, kinetic and structural characterization of Kif15 is reported and it is compared with the functionally related motor Eg5. Although Kif15 contains ADP in the catalytic site, its motor-domain structure was captured in the `ATP-like' configuration, with the neck linker docked to the catalytic core. The interaction of Kif15 with microtubules was also investigated and structural differences between these two motors were elucidated which indicate profound differences in their mode of action, in agreement with current models of microtubule cross-linking and sliding.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Neuronas/metabolismo , Huso Acromático/metabolismo , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cristalografía por Rayos X , Humanos , Magnesio/química , Magnesio/metabolismo , Mitosis , Datos de Secuencia Molecular , Neuronas/citología , Conformación Proteica , Ratas
8.
PLoS Biol ; 9(11): e1001207, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22140358

RESUMEN

The minimum motor domain of kinesin-1 is a single head. Recent evidence suggests that such minimal motor domains generate force by a biased binding mechanism, in which they preferentially select binding sites on the microtubule that lie ahead in the progress direction of the motor. A specific molecular mechanism for biased binding has, however, so far been lacking. Here we use atomistic Brownian dynamics simulations combined with experimental mutagenesis to show that incoming kinesin heads undergo electrostatically guided diffusion-to-capture by microtubules, and that this produces directionally biased binding. Kinesin-1 heads are initially rotated by the electrostatic field so that their tubulin-binding sites face inwards, and then steered towards a plus-endwards binding site. In tethered kinesin dimers, this bias is amplified. A 3-residue sequence (RAK) in kinesin helix alpha-6 is predicted to be important for electrostatic guidance. Real-world mutagenesis of this sequence powerfully influences kinesin-driven microtubule sliding, with one mutant producing a 5-fold acceleration over wild type. We conclude that electrostatic interactions play an important role in the kinesin stepping mechanism, by biasing the diffusional association of kinesin with microtubules.


Asunto(s)
Cinesinas/química , Microtúbulos/metabolismo , Sitios de Unión , Simulación por Computador , Cinesinas/genética , Cinesinas/metabolismo , Modelos Moleculares , Mutagénesis , Electricidad Estática , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
9.
Curr Opin Cell Biol ; 18(1): 61-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16361092

RESUMEN

Kinesin-1 is a single-molecule walking machine, driven by ATP turnover. Recent optical trapping experiments show that pulling backwards on a walking kinesin-1 molecule causes the mechanical walking action to reverse, while the coupled chemical cycle of ATP turnover continues, apparently, to run forwards -- kinesin can moonwalk. Individual forward- and back-steps are fast, and each appears to be a single event, complete in a few tens of microseconds, with no substeps. Between steps, kinesin pauses, waiting for the next ATP to arrive. Several lines of evidence indicate that during these between-step dwells, only one of the two heads is strongly attached to the microtubule. The position of the other head during the dwells is less certain, and more controversial.


Asunto(s)
Cinesinas/química , Proteínas Motoras Moleculares/metabolismo , Movimiento , Adenosina Trifosfato/química , Cinesinas/metabolismo , Microtúbulos/metabolismo , Modelos Biológicos , Conformación Proteica , Factores de Tiempo
10.
Commun Biol ; 6(1): 946, 2023 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-37717119

RESUMEN

Taxol is a small molecule effector that allosterically locks tubulin into the microtubule lattice. We show here that taxol has different effects on different single-isotype microtubule lattices. Using in vitro reconstitution, we demonstrate that single-isotype α1ß4 GDP-tubulin lattices are stabilised and expanded by 10 µM taxol, as reported by accelerated microtubule gliding in kinesin motility assays, whereas single-isotype α1ß3 GDP-tubulin lattices are stabilised but not expanded. This isotype-specific action of taxol drives gliding of segmented-isotype GDP-taxol microtubules along convoluted, sinusoidal paths, because their expanded α1ß4 segments try to glide faster than their compacted α1ß3 segments. In GMPCPP, single-isotype α1ß3 and α1ß4 lattices both show accelerated gliding, indicating that both can in principle be driven to expand. We therefore propose that taxol-induced lattice expansion requires a higher taxol occupancy than taxol-induced stabilisation, and that higher taxol occupancies are accessible to α1ß4 but not α1ß3 single-isotype lattices.


Asunto(s)
Paclitaxel , Tubulina (Proteína) , Paclitaxel/farmacología , Microtúbulos , Cinesinas
11.
Biochem Soc Trans ; 40(2): 400-3, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22435819

RESUMEN

Kinesin-1 is a walking machine that takes ~8 nm steps along microtubules. Some aspects of the molecular mechanism of walking are now clear, but many are not. In the present paper, we discuss currently controversial points, focusing on the pathways by which kinesin takes occasional backsteps. Backsteps represent failures of the forwards-biasing mechanism. By studying the mechanochemistry of backstepping, one can learn much about the underlying molecular mechanisms responsible for forwards directional bias in the walking action.


Asunto(s)
Cinesinas/metabolismo , Adenosina Trifosfato/metabolismo , Cinesinas/química , Conformación Proteica
12.
Commun Biol ; 5(1): 1368, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36539506

RESUMEN

Kinesin motor domains generate impulses of force and movement that have both translational and rotational (torque) components. Here, we ask how the torque component influences function in cargo-attached teams of weakly processive kinesins. Using an assay in which kinesin-coated gold nanorods (kinesin-GNRs) translocate on suspended microtubules, we show that for both single-headed KIF1A and dimeric ZEN-4, the intensities of polarized light scattered by the kinesin-GNRs in two orthogonal directions periodically oscillate as the GNRs crawl towards microtubule plus ends, indicating that translocating kinesin-GNRs unidirectionally rotate about their short (yaw) axes whilst following an overall left-handed helical orbit around the microtubule axis. For orientations of the GNR that generate a signal, the period of this short axis rotation corresponds to two periods of the overall helical trajectory. Torque force thus drives both rolling and yawing of near-spherical cargoes carrying rigidly-attached weakly processive kinesins, with possible relevance to intracellular transport.


Asunto(s)
Cinesinas , Nanotubos , Torque , Oro , Microtúbulos
13.
Curr Biol ; 32(17): 3862-3870.e6, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-35961316

RESUMEN

Intracellular transport is essential for neuronal function and survival. The most effective plus-end-directed neuronal transporter is the kinesin-3 KIF1C, which transports large secretory vesicles and endosomes.1-4 Mutations in KIF1C cause hereditary spastic paraplegia and cerebellar dysfunction in human patients.5-8 In contrast to other kinesin-3s, KIF1C is a stable dimer and a highly processive motor in its native state.9,10 Here, we establish a baseline for the single-molecule mechanics of Kif1C. We show that full-length KIF1C molecules can processively step against the load of an optical trap and reach average stall forces of 3.7 pN. Compared with kinesin-1, KIF1C has a higher propensity to slip backward under load, which results in a lower maximal single-molecule force. However, KIF1C remains attached to the microtubule while slipping backward and re-engages quickly, consistent with its super processivity. Two pathogenic mutations, P176L and R169W, that cause hereditary spastic paraplegia in humans7,8 maintain fast, processive single-molecule motility in vitro but with decreased run length and slightly increased unloaded velocity compared with the wild-type motor. Under load in an optical trap, force generation by these mutants is severely reduced. In cells, the same mutants are impaired in producing sufficient force to efficiently relocate organelles. Our results show how its mechanics supports KIF1C's role as an intracellular transporter and explain how pathogenic mutations at the microtubule-binding interface of KIF1C impair the cellular function of these long-distance transporters and result in neuronal disease.


Asunto(s)
Cinesinas , Paraplejía Espástica Hereditaria , Humanos , Cinesinas/genética , Microtúbulos/metabolismo , Mutación , Unión Proteica , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo
14.
BMC Biol ; 8: 107, 2010 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-20731883

RESUMEN

Kinesin-14 motor proteins step towards microtubule minus ends, in the opposite direction to other kinesins. Work on the still-enigmatic kinesin-14 mechanism published in BMC Structural Biology shows that the carboxyl terminus of the motor head undergoes a dock-undock cycle, like that of plus-end-directed kinesins.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Animales , Cristalografía por Rayos X , Drosophila/química , Drosophila/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Humanos , Unión Proteica , Conformación Proteica
15.
J Biol Chem ; 284(43): 29246-50, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19740752

RESUMEN

Schizosaccharomyces pombe Mal3 is a member of the EB family of proteins, which are proposed to be core elements in a tip-tracking network that regulates microtubule dynamics in cells. How Mal3 itself influences microtubule dynamics is unclear. We tested the effects of full-length recombinant Mal3 on dynamic microtubules assembled in vitro from purified S. pombe tubulin, using dark field video microscopy to avoid fluorescent tagging and data-averaging techniques to improve spatiotemporal resolution. We find that catastrophe occurs stochastically as a fast (<2.2 s) transition from constant speed growth to constant speed shrinkage with a constant probability that is independent of the Mal3 concentration. This implies that Mal3 neither stabilizes nor destabilizes microtubule tips. Mal3 does, however, stabilize the main part of the microtubule lattice, inhibiting shrinkage and increasing the frequency of rescues, consistent with recent models in which Mal3 on the lattice forms stabilizing lateral links between neighboring protofilaments. At high concentrations, Mal3 can entirely block shrinkage and induce very rapid rescue, making catastrophes impossible to detect, which may account for the apparent suppression of catastrophe by Mal3 and other EBs in vivo. Overall, we find that Mal3 stabilizes microtubules not by preventing catastrophe at the microtubule tip but by inhibiting lattice depolymerization and enhancing rescue. We argue that this implies that Mal3 binds microtubules in different modes at the tip and on the lattice.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Tubulina (Proteína)/metabolismo , Sistema Libre de Células/química , Sistema Libre de Células/metabolismo , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/química , Microtúbulos/genética , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Schizosaccharomyces/química , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética , Tubulina (Proteína)/química , Tubulina (Proteína)/genética
16.
EMBO Rep ; 9(8): 761-5, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18552767

RESUMEN

In most organisms, kinesin-5 motors are essential for mitosis and meiosis, where they crosslink and slide apart the antiparallel microtubule half-spindles. Recently, it was shown using single-molecule optical trapping that a truncated, double-headed human kinesin-5 dimer can step processively along microtubules. However, processivity is limited ( approximately 8 steps) with little coordination between the heads, raising the possibility that kinesin-5 motors might also be able to move by a nonprocessive mechanism. To investigate this, we engineered single-headed kinesin-5 dimers. We show that a set of these single-headed Eg5 dimers drive microtubule sliding at about 90% of wild-type velocity, indicating that Eg5 can slide microtubules by a mechanism in which one head of each Eg5 head-pair is effectively redundant. On the basis of this, we propose a muscle-like model for Eg5-driven microtubule sliding in spindles in which most force-generating events are single-headed interactions and alternate-heads processivity is rare.


Asunto(s)
Cinesinas/fisiología , Microtúbulos/fisiología , Proteínas de Xenopus/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Dimerización , Electroforesis en Gel de Poliacrilamida , Cinesinas/química , Cinesinas/metabolismo , Modelos Biológicos , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis
17.
Curr Biol ; 30(17): R988-R991, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32898497

RESUMEN

Modern life is replete with function-expanding dongles, and life at the molecular scale is, it turns out, no exception. Hanging out of the back of the Kif14 molecular motor is an intrinsically disordered domain that gives it superpowers.


Asunto(s)
Cinesinas , Cinesinas/genética
18.
Biochem Soc Trans ; 37(Pt 5): 1045-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19754449

RESUMEN

It is clear that the main cellular mission of the molecular motor kinesin-5 (known as Eg5 in vertebrates) is to cross-link antiparallel microtubules and to slide them apart, thus playing a critical role during bipolar spindle formation. Nonetheless, important questions about the cell biological and biophysical mechanisms of Eg5 remain unanswered. With the 20th 'birthday' of Eg5 approaching, we discuss recent insights into the in vitro and in vivo functions of Eg5, in the context of our own recent work.


Asunto(s)
Cinesinas/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Ciclo Celular/fisiología , Humanos , Microtúbulos/metabolismo , Huso Acromático/metabolismo
19.
J Cell Biol ; 156(2): 349-59, 2002 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-11807097

RESUMEN

Recent studies have shown that the targeting of substrate adhesions by microtubules promotes adhesion site disassembly (Kaverina, I., O. Krylyshkina, and J.V. Small. 1999. J. Cell Biol. 146:1033-1043). It was accordingly suggested that microtubules serve to convey a signal to adhesion sites to modulate their turnover. Because microtubule motors would be the most likely candidates for effecting signal transmission, we have investigated the consequence of blocking microtubule motor activity on adhesion site dynamics. Using a function-blocking antibody as well as dynamitin overexpression, we found that a block in dynein-cargo interaction induced no change in adhesion site dynamics in Xenopus fibroblasts. In comparison, a block of kinesin-1 activity, either via microinjection of the SUK-4 antibody or of a kinesin-1 heavy chain construct mutated in the motor domain, induced a dramatic increase in the size and reduction in number of substrate adhesions, mimicking the effect observed after microtubule disruption by nocodazole. Blockage of kinesin activity had no influence on either the ability of microtubules to target substrate adhesions or on microtubule polymerisation dynamics. We conclude that conventional kinesin is not required for the guidance of microtubules into substrate adhesions, but is required for the focal delivery of a component(s) that retards their growth or promotes their disassembly.


Asunto(s)
Adhesiones Focales , Cinesinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Microtúbulos/metabolismo , Xenopus/metabolismo , Animales , Línea Celular , Movimiento Celular , Dineínas/metabolismo , Fibroblastos , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/genética , Potenciales de la Membrana , Ratones , Microscopía por Video , Microtúbulos/efectos de los fármacos , Proteínas Motoras Moleculares/metabolismo , Mutagénesis , Nocodazol/farmacología , Reacción en Cadena de la Polimerasa , Unión Proteica , Ratas
20.
Trends Biochem Sci ; 29(6): 301-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15276184

RESUMEN

The chemical kinetic mechanism of kinesin (K) is considered by using a consensus scheme incorporating biochemically defined open, closed and trapped states. In the absence of microtubules, the dominant species is a trapped K*ADP state, which is defined by its ultra-slow release of ADP (off rate, k(off) approximately 0.002 s(-1)) and weak microtubule binding (dissociation constant, K(d) approximately 10-20 microM). Once bound, this trapped state equilibrates with a strongly binding open state that rapidly releases ADP (k(off) approximately 300 s(-1)). After ADP release, Mg*ATP binds (on rate, k(on) approximately 2 microM(-1)s(-1)) driving formation of a closed state that is defined by hydrolysis competence and by strong binding to microtubules. Hydrolysis (k(hyd) approximately 100-300 s(-1)) and phosphate release (k(off)>100 s(-1)) both occur in this microtubule-bound closed state. Phosphate release acts as a gate that controls reversion to the trapped K*ADP state, which detaches from the microtubule, completing the cycle.


Asunto(s)
Cinesinas/química , Adenosina Difosfato/química , Adenosina Trifosfato/química , Animales , Humanos , Hidrólisis , Cinética , Microtúbulos/química , Modelos Moleculares , Fosfatos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA