Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 297(5): 101063, 2021 11.
Article in English | MEDLINE | ID: mdl-34375637

ABSTRACT

Plasmodium parasites cause malaria and are responsible annually for hundreds of thousands of deaths. Kinesins are a superfamily of microtubule-dependent ATPases that play important roles in the parasite replicative machinery, which is a potential target for antiparasite drugs. Kinesin-5, a molecular motor that cross-links microtubules, is an established antimitotic target in other disease contexts, but its mechanism in Plasmodium falciparum is unclear. Here, we characterized P. falciparum kinesin-5 (PfK5) using cryo-EM to determine the motor's nucleotide-dependent microtubule-bound structure and introduced 3D classification of individual motors into our microtubule image processing pipeline to maximize our structural insights. Despite sequence divergence in PfK5, the motor exhibits classical kinesin mechanochemistry, including ATP-induced subdomain rearrangement and cover neck bundle formation, consistent with its plus-ended directed motility. We also observed that an insertion in loop5 of the PfK5 motor domain creates a different environment in the well-characterized human kinesin-5 drug-binding site. Our data reveal the possibility for selective inhibition of PfK5 and can be used to inform future exploration of Plasmodium kinesins as antiparasite targets.


Subject(s)
Kinesins , Plasmodium falciparum , Protozoan Proteins , Antimalarials/chemistry , Cryoelectron Microscopy , Humans , Kinesins/metabolism , Kinesins/ultrastructure , Plasmodium falciparum/chemistry , Plasmodium falciparum/metabolism , Plasmodium falciparum/ultrastructure , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Protozoan Proteins/ultrastructure
2.
Am J Med Genet A ; 185(4): 1113-1119, 2021 04.
Article in English | MEDLINE | ID: mdl-33506645

ABSTRACT

Cortical dysplasia, complex, with other brain malformations 3 (CDCBM3) is a rare autosomal dominant syndrome caused by Kinesin family Member 2A (KIF2A) gene mutation. Patients with CDCBM3 exhibit posterior dominant agyria/pachygyria with severe motor dysfunction. Here, we report an 8-year-old boy with CDCBM3 showing a typical, but relatively mild, clinical presentation of CDCBM3 features. Whole-exome sequencing identified a heterozygous mutation of NM_001098511.2:c.1298C>A [p.(Ser433Tyr)]. To our knowledge, the mutation has never been reported previously. The variant was located distal to the nucleotide binding domain (NBD), in which previously-reported variants in CDCBM3 patients have been located. The computational structural analysis showed the p.433 forms the pocket with NBD. Variants in KIF2A have been reported in the NBD for CDCBM3, in the kinesin motor 3 domain, but not in the NBD in epilepsy, and outside of the kinesin motor domain in autism spectrum syndrome, respectively. Our patient has a variant, that is not in the NBD but at the pocket with the NBD, resulting in a clinical features of CDCBM3 with mild symptoms. The clinical findings of patients with KIF2A variants appear restricted to the central nervous system and facial anomalies. We can call this spectrum "KIF2A syndrome" with variable severity.


Subject(s)
Epilepsy/genetics , Kinesins/genetics , Malformations of Cortical Development/genetics , Microtubule-Associated Proteins/genetics , Brain/diagnostic imaging , Brain/pathology , Child , Epilepsy/diagnosis , Epilepsy/diagnostic imaging , Epilepsy/pathology , Heterozygote , Humans , Kinesins/ultrastructure , Male , Malformations of Cortical Development/diagnosis , Malformations of Cortical Development/diagnostic imaging , Malformations of Cortical Development/pathology , Microtubule-Associated Proteins/ultrastructure , Mutation, Missense/genetics , Protein Conformation , Tubulin/genetics , Exome Sequencing
3.
Elife ; 92020 11 30.
Article in English | MEDLINE | ID: mdl-33252036

ABSTRACT

Subcellular compartmentalisation is necessary for eukaryotic cell function. Spatial and temporal regulation of kinesin activity is essential for building these local environments via control of intracellular cargo distribution. Kinesin-binding protein (KBP) interacts with a subset of kinesins via their motor domains, inhibits their microtubule (MT) attachment, and blocks their cellular function. However, its mechanisms of inhibition and selectivity have been unclear. Here we use cryo-electron microscopy to reveal the structure of KBP and of a KBP-kinesin motor domain complex. KBP is a tetratricopeptide repeat-containing, right-handed α-solenoid that sequesters the kinesin motor domain's tubulin-binding surface, structurally distorting the motor domain and sterically blocking its MT attachment. KBP uses its α-solenoid concave face and edge loops to bind the kinesin motor domain, and selected structure-guided mutations disrupt KBP inhibition of kinesin transport in cells. The KBP-interacting motor domain surface contains motifs exclusively conserved in KBP-interacting kinesins, suggesting a basis for kinesin selectivity.


Subject(s)
Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Cryoelectron Microscopy , Humans , Kinesins/chemistry , Kinesins/ultrastructure
4.
Elife ; 92020 01 20.
Article in English | MEDLINE | ID: mdl-31958056

ABSTRACT

Kinesin-5 motors organize mitotic spindles by sliding apart microtubules. They are homotetramers with dimeric motor and tail domains at both ends of a bipolar minifilament. Here, we describe a regulatory mechanism involving direct binding between tail and motor domains and its fundamental role in microtubule sliding. Kinesin-5 tails decrease microtubule-stimulated ATP-hydrolysis by specifically engaging motor domains in the nucleotide-free or ADP states. Cryo-EM reveals that tail binding stabilizes an open motor domain ATP-active site. Full-length motors undergo slow motility and cluster together along microtubules, while tail-deleted motors exhibit rapid motility without clustering. The tail is critical for motors to zipper together two microtubules by generating substantial sliding forces. The tail is essential for mitotic spindle localization, which becomes severely reduced in tail-deleted motors. Our studies suggest a revised microtubule-sliding model, in which kinesin-5 tails stabilize motor domains in the microtubule-bound state by slowing ATP-binding, resulting in high-force production at both homotetramer ends.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Humans , Hydrolysis , Kinesins/chemistry , Kinesins/ultrastructure , Kinetics , Protein Binding , Protein Domains , Spindle Apparatus/metabolism
5.
J Struct Biol ; 207(3): 312-316, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31288039

ABSTRACT

In many eukaryotes, kinesin-5 motors are essential for mitosis, and small molecules that inhibit human kinesin-5 disrupt cell division. To investigate whether fungal kinesin-5s could be targets for novel fungicides, we studied kinesin-5 from the pathogenic fungus Ustilago maydis. We used cryo-electron microscopy to determine the microtubule-bound structure of its motor domain with and without the N-terminal extension. The ATP-like conformations of the motor in the presence or absence of this N-terminus are very similar, suggesting this region is structurally disordered and does not directly influence the motor ATPase. The Ustilago maydis kinesin-5 motor domain adopts a canonical ATP-like conformation, thereby allowing the neck linker to bind along the motor domain towards the microtubule plus end. However, several insertions within this motor domain are structurally distinct. Loop2 forms a non-canonical interaction with α-tubulin, while loop8 may bridge between two adjacent protofilaments. Furthermore, loop5 - which in human kinesin-5 is involved in binding allosteric inhibitors - protrudes above the nucleotide binding site, revealing a distinct binding pocket for potential inhibitors. This work highlights fungal-specific elaborations of the kinesin-5 motor domain and provides the structural basis for future investigations of kinesins as targets for novel fungicides.


Subject(s)
Cryoelectron Microscopy/methods , Fungal Proteins/chemistry , Kinesins/chemistry , Microtubules/chemistry , Protein Domains , Ustilago/ultrastructure , Fungal Proteins/ultrastructure , Kinesins/metabolism , Kinesins/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Protein Binding , Ustilago/metabolism
6.
Article in English | MEDLINE | ID: mdl-29716949

ABSTRACT

Myosin motors power movements on actin filaments, whereas dynein and kinesin motors power movements on microtubules. The mechanisms of these motor proteins differ, but, in all cases, ATP hydrolysis and subsequent release of the hydrolysis products drives a cycle of interactions with the track (either an actin filament or a microtubule), resulting in force generation and directed movement.


Subject(s)
Dyneins/physiology , Kinesins/physiology , Myosins/physiology , Actin Cytoskeleton/metabolism , Adenosine Triphosphate/metabolism , Biological Transport/physiology , Dyneins/ultrastructure , Kinesins/ultrastructure , Models, Biological , Models, Molecular , Myosins/ultrastructure
7.
Nat Commun ; 9(1): 1662, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695795

ABSTRACT

Kinesin-13s constitute a distinct group within the kinesin superfamily of motor proteins that promote microtubule depolymerization and lack motile activity. The molecular mechanism by which kinesin-13s depolymerize microtubules and are adapted to perform a seemingly very different activity from other kinesins is still unclear. To address this issue, here we report the near atomic resolution cryo-electron microscopy (cryo-EM) structures of Drosophila melanogaster kinesin-13 KLP10A protein constructs bound to curved or straight tubulin in different nucleotide states. These structures show how nucleotide induced conformational changes near the catalytic site are coupled with movement of the kinesin-13-specific loop-2 to induce tubulin curvature leading to microtubule depolymerization. The data highlight a modular structure that allows similar kinesin core motor-domains to be used for different functions, such as motility or microtubule depolymerization.


Subject(s)
Drosophila Proteins/ultrastructure , Kinesins/ultrastructure , Microtubules/ultrastructure , Tubulin/ultrastructure , Adenosine Triphosphate/metabolism , Cell Movement , Cryoelectron Microscopy , Drosophila Proteins/chemistry , Drosophila Proteins/isolation & purification , Kinesins/chemistry , Kinesins/isolation & purification , Microtubules/metabolism , Molecular Docking Simulation , Polymerization , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/ultrastructure , Tubulin/chemistry
8.
Elife ; 62017 08 11.
Article in English | MEDLINE | ID: mdl-28826477

ABSTRACT

MKLP2, a kinesin-6, has critical roles during the metaphase-anaphase transition and cytokinesis. Its motor domain contains conserved nucleotide binding motifs, but is divergent in sequence (~35% identity) and size (~40% larger) compared to other kinesins. Using cryo-electron microscopy and biophysical assays, we have undertaken a mechanochemical dissection of the microtubule-bound MKLP2 motor domain during its ATPase cycle, and show that many facets of its mechanism are distinct from other kinesins. While the MKLP2 neck-linker is directed towards the microtubule plus-end in an ATP-like state, it does not fully dock along the motor domain. Furthermore, the footprint of the MKLP2 motor domain on the MT surface is altered compared to motile kinesins, and enhanced by kinesin-6-specific sequences. The conformation of the highly extended loop6 insertion characteristic of kinesin-6s is nucleotide-independent and does not contact the MT surface. Our results emphasize the role of family-specific insertions in modulating kinesin motor function.


Subject(s)
Kinesins/metabolism , Kinesins/ultrastructure , Mechanical Phenomena , Microtubules/metabolism , Microtubules/ultrastructure , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Protein Binding , Protein Conformation
9.
Sci Rep ; 7(1): 6166, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28733669

ABSTRACT

In vitro gliding assay of microtubules (MTs) on kinesins has provided us with valuable biophysical and chemo-mechanical insights of this biomolecular motor system. Visualization of MTs in an in vitro gliding assay has been mainly dependent on optical microscopes, limited resolution of which often render them insufficient sources of desired information. In this work, using high speed atomic force microscopy (HS-AFM), which allows imaging with higher resolution, we monitored MTs and protofilaments (PFs) of tubulins while gliding on kinesins. Moreover, under the HS-AFM, we also observed splitting of gliding MTs into single PFs at their leading ends. The split single PFs interacted with kinesins and exhibited translational motion, but with a slower velocity than the MTs. Our investigation at the molecular level, using the HS-AFM, would provide new insights to the mechanics of MTs in dynamic systems and their interaction with motor proteins.


Subject(s)
Kinesins/ultrastructure , Microscopy, Atomic Force/methods , Tubulin/ultrastructure , Microtubules/ultrastructure , Motion , Single Molecule Imaging
10.
Elife ; 62017 05 15.
Article in English | MEDLINE | ID: mdl-28504639

ABSTRACT

The detailed basis of walking by dimeric molecules of kinesin along microtubules has remained unclear, partly because available structural methods have been unable to capture microtubule-bound intermediates of this process. Utilizing novel electron cryomicroscopy methods, we solved structures of microtubule-attached, dimeric kinesin bound to an ATP analog. We find that under these conditions, the kinesin dimer can attach to the microtubule with either one or two motor domains, and we present sub-nanometer resolution reconstructions of both states. The former structure reveals a novel kinesin conformation that revises the current understanding of how ATP binding is coupled to forward stepping of the motor. The latter structure indicates how tension between the two motor domains keeps their cycles out of phase in order to stimulate directional motility. The methods presented here pave the way for future structural studies of a variety of challenging macromolecules that bind to microtubules and other filaments.


Subject(s)
Imaging, Three-Dimensional , Kinesins/metabolism , Kinesins/ultrastructure , Microtubules/metabolism , Microtubules/ultrastructure , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Protein Binding , Protein Conformation , Protein Multimerization
11.
Nat Struct Mol Biol ; 24(5): 461-468, 2017 May.
Article in English | MEDLINE | ID: mdl-28394326

ABSTRACT

Cilia are multifunctional organelles that are constructed using intraflagellar transport (IFT) of cargo to and from their tip. It is widely held that the retrograde IFT motor, dynein-2, must be controlled in order to reach the ciliary tip and then unleashed to power the return journey. However, the mechanism is unknown. Here, we systematically define the mechanochemistry of human dynein-2 motors as monomers, dimers, and multimotor assemblies with kinesin-II. Combining these data with insights from single-particle EM, we discover that dynein-2 dimers are intrinsically autoinhibited. Inhibition is mediated by trapping dynein-2's mechanical 'linker' and 'stalk' domains within a novel motor-motor interface. We find that linker-mediated inhibition enables efficient transport of dynein-2 by kinesin-II in vitro. These results suggest a conserved mechanism for autoregulation among dimeric dyneins, which is exploited as a switch for dynein-2's recycling activity during IFT.


Subject(s)
Dyneins/chemistry , Dyneins/metabolism , Dyneins/ultrastructure , Humans , Kinesins/chemistry , Kinesins/metabolism , Kinesins/ultrastructure , Microscopy, Electron , Models, Biological , Models, Molecular , Protein Conformation , Protein Multimerization
12.
J Eukaryot Microbiol ; 64(3): 293-307, 2017 05.
Article in English | MEDLINE | ID: mdl-27595611

ABSTRACT

Ciliates such as Tetrahymena thermophila have two distinct nuclei within one cell: the micronucleus that undergoes mitosis and meiosis and the macronucleus that undergoes amitosis, a type of nuclear division that does not involve a bipolar spindle, but still relies on intranuclear microtubules. Ciliates provide an opportunity for the discovery of factors that specifically contribute to chromosome segregation based on a bipolar spindle, by identification of factors that affect the micronuclear but not the macronuclear division. Kinesin-14 is a conserved minus-end directed microtubule motor that cross-links microtubules and contributes to the bipolar spindle sizing and organization. Here, we use homologous DNA recombination to knock out genes that encode kinesin-14 orthologues (KIN141, KIN142) in Tetrahymena. A loss of KIN141 led to severe defects in the chromosome segregation during both mitosis and meiosis but did not affect amitosis. A loss of KIN141 altered the shape of the meiotic spindle in a way consistent with the KIN141's contribution to the organization of the spindle poles. EGFP-tagged KIN141 preferentially accumulated at the spindle poles during the meiotic prophase and metaphase I. Thus, in ciliates, kinesin-14 is important for nuclear divisions that involve a bipolar spindle.


Subject(s)
Chromosome Segregation , Ciliophora/genetics , Kinesins/genetics , Kinesins/physiology , Meiosis , Mitosis , Tetrahymena thermophila/genetics , Animals , Cell Nucleus , Ciliophora/cytology , Gene Knockout Techniques , Kinesins/classification , Kinesins/ultrastructure , Macronucleus , Meiotic Prophase I , Metaphase , Microtubules , Mutation , Phylogeny , Recombinant Proteins , Spindle Apparatus , Spindle Poles , Tetrahymena/genetics , Tetrahymena thermophila/cytology , Tetrahymena thermophila/metabolism
13.
Proc Natl Acad Sci U S A ; 113(34): 9430-9, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27493215

ABSTRACT

Proteins that associate with microtubules (MTs) are crucial to generate MT arrays and establish different cellular architectures. One example is PRC1 (protein regulator of cytokinesis 1), which cross-links antiparallel MTs and is essential for the completion of mitosis and cytokinesis. Here we describe a 4-Å-resolution cryo-EM structure of monomeric PRC1 bound to MTs. Residues in the spectrin domain of PRC1 contacting the MT are highly conserved and interact with the same pocket recognized by kinesin. We additionally found that PRC1 promotes MT assembly even in the presence of the MT stabilizer taxol. Interestingly, the angle of the spectrin domain on the MT surface corresponds to the previously observed cross-bridge angle between MTs cross-linked by full-length, dimeric PRC1. This finding, together with molecular dynamic simulations describing the intrinsic flexibility of PRC1, suggests that the MT-spectrin domain interface determines the geometry of the MT arrays cross-linked by PRC1.


Subject(s)
Cell Cycle Proteins/ultrastructure , Kinesins/ultrastructure , Microtubules/ultrastructure , Protein Subunits/chemistry , Tubulin/ultrastructure , Amino Acid Motifs , Animals , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Gene Expression , Humans , Kinesins/genetics , Kinesins/metabolism , Microtubules/metabolism , Molecular Dynamics Simulation , Paclitaxel/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Swine , Tubulin/genetics , Tubulin/metabolism , Tubulin Modulators/chemistry
14.
Sci Rep ; 5: 11916, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26132639

ABSTRACT

Chromosome higher order structure has been an enigma for over a century. The most important structural finding has been the presence of a chromosome scaffold composed of non-histone proteins; so-called scaffold proteins. However, the organization and function of the scaffold are still controversial. Here, we use three dimensional-structured illumination microscopy (3D-SIM) and focused ion beam/scanning electron microscopy (FIB/SEM) to reveal the axial distributions of scaffold proteins in metaphase chromosomes comprising two strands. We also find that scaffold protein can adaptably recover its original localization after chromosome reversion in the presence of cations. This reversion to the original morphology underscores the role of the scaffold for intrinsic structural integrity of chromosomes. We therefore propose a new structural model of the chromosome scaffold that includes twisted double strands, consistent with the physical properties of chromosomal bending flexibility and rigidity. Our model provides new insights into chromosome higher order structure.


Subject(s)
Chromosomal Proteins, Non-Histone/ultrastructure , Chromosomes, Human/ultrastructure , Adenosine Triphosphatases/physiology , Adenosine Triphosphatases/ultrastructure , Antigens, Neoplasm/physiology , Antigens, Neoplasm/ultrastructure , Chromosomal Proteins, Non-Histone/physiology , Chromosomes, Human/physiology , DNA Topoisomerases, Type II/physiology , DNA Topoisomerases, Type II/ultrastructure , DNA-Binding Proteins/physiology , DNA-Binding Proteins/ultrastructure , HeLa Cells , Humans , Imaging, Three-Dimensional , Kinesins/physiology , Kinesins/ultrastructure , Metaphase , Multiprotein Complexes/physiology , Multiprotein Complexes/ultrastructure
15.
Biomech Model Mechanobiol ; 14(5): 1107-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25676575

ABSTRACT

Kinesin is a motor protein that delivers cargo inside a cell. Kinesin has many different families, but they perform basically same function and have same motions. The walking motion of kinesin enables the cargo delivery inside the cell. Autoinhibition of kinesin is important because it explains how function of kinesin inside a cell is stopped. Former researches showed that tail binding is related to autoinhibition of kinesin. In this work, we performed normal mode analysis with elastic network model using different conformation of kinesin to determine the effect of tail binding by considering four models such as functional form, autoinhibited form, autoinhibited form without tail, and autoinhibited form with carbon structure. Our calculation of the thermal fluctuation and cross-correlation shows the change of tail-binding region in structural motion. Also strain energy of kinesin showed that elimination of tail binding effect leads the structure to have energetically similar behavior with the functional form.


Subject(s)
Adenosine Diphosphate/chemistry , Kinesins/chemistry , Kinesins/ultrastructure , Models, Chemical , Models, Molecular , Molecular Motor Proteins/chemistry , Binding Sites , Computer Simulation , Elastic Modulus , Energy Transfer , Kinetics , Molecular Motor Proteins/ultrastructure , Motion , Protein Binding , Protein Conformation , Structure-Activity Relationship , Temperature
16.
Nanoscale ; 7(1): 82-5, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25254951

ABSTRACT

A smart self-powered cargo delivery system that is composed of creatine phosphate kinase (CPK) microspheres, kinesins and microtubules is demonstrated. The CPK microsphere not only acts as an ATP generation and buffering system, but also as a carrier for cargo transport, thus realizing the easy loading and self-powered delivery of cargos at the same time.


Subject(s)
Delayed-Action Preparations/chemistry , Kinesins/chemistry , Microtubules/chemistry , Molecular Motor Proteins/chemistry , Nanocapsules/chemistry , Robotics/methods , Kinesins/ultrastructure , Materials Testing , Microtubules/ultrastructure , Molecular Motor Proteins/ultrastructure , Motion , Nanocapsules/ultrastructure , Particle Size
17.
Biochemistry ; 54(3): 859-69, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25537000

ABSTRACT

We have employed molecular dynamics (MD) simulation to investigate, with atomic details, the structural dynamics and energetics of three major ATPase states (ADP, APO, and ATP state) of a human kinesin-1 monomer in complex with a tubulin dimer. Starting from a recently solved crystal structure of ATP-like kinesin-tubulin complex by the Knossow lab, we have used flexible fitting of cryo-electron-microscopy maps to construct new structural models of the kinesin-tubulin complex in APO and ATP state, and then conducted extensive MD simulations (total 400 ns for each state), followed by flexibility analysis, principal component analysis, hydrogen bond analysis, and binding free energy analysis. Our modeling and simulation have revealed key nucleotide-dependent changes in the structure and flexibility of the nucleotide-binding pocket (featuring a highly flexible and open switch I in APO state) and the tubulin-binding site, and allosterically coupled motions driving the APO to ATP transition. In addition, our binding free energy analysis has identified a set of key residues involved in kinesin-tubulin binding. On the basis of our simulation, we have attempted to address several outstanding issues in kinesin study, including the possible roles of ß-sheet twist and neck linker docking in regulating nucleotide release and binding, the structural mechanism of ADP release, and possible extension and shortening of α4 helix during the ATPase cycle. This study has provided a comprehensive structural and dynamic picture of kinesin's major ATPase states, and offered promising targets for future mutational and functional studies to investigate the molecular mechanism of kinesin motors.


Subject(s)
Adenosine Triphosphatases/metabolism , Kinesins/chemistry , Kinesins/metabolism , Molecular Dynamics Simulation , Protein Multimerization , Tubulin/chemistry , Tubulin/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Binding Sites , Cryoelectron Microscopy , Humans , Kinesins/ultrastructure , Models, Molecular , Myosins/metabolism , Principal Component Analysis , Protein Binding , Protein Structure, Secondary , Thermodynamics
18.
Sci Rep ; 4: 7255, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25434968

ABSTRACT

Within living cells, the transport of cargo is accomplished by groups of molecular motors. Such collective transport could utilize mechanisms which emerge from inter-motor interactions in ways that are yet to be fully understood. Here we combined experimental measurements of two-kinesin transport with a theoretical framework to investigate the functional ramifications of inter-motor interactions on individual motor function and collective cargo transport. In contrast to kinesin's low sidestepping frequency when present as a single motor, with exactly two kinesins per cargo, we observed substantial motion perpendicular to the microtubule. Our model captures a surface-associated mode of kinesin, which is only accessible via inter-motor interference in groups, in which kinesin diffuses along the microtubule surface and rapidly "hops" between protofilaments without dissociating from the microtubule. Critically, each kinesin transitions dynamically between the active stepping mode and this weak surface-associated mode enhancing local exploration of the microtubule surface, possibly enabling cellular cargos to overcome macromolecular crowding and to navigate obstacles along microtubule tracks without sacrificing overall travel distance.


Subject(s)
Kinesins/chemistry , Microtubules/chemistry , Models, Chemical , Molecular Motor Proteins/chemistry , Motion , Computer Simulation , Energy Transfer , Kinesins/ultrastructure , Microtubules/ultrastructure , Models, Molecular , Molecular Motor Proteins/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Protein Conformation
19.
Article in English | MEDLINE | ID: mdl-25353516

ABSTRACT

Intracellular transport based on molecular motors and its regulation are crucial to the functioning of cells. Filamentary tracks of the cells are abundantly decorated with nonmotile microtubule-associated proteins, such as tau. Motivated by experiments on kinesin-tau interactions [Dixit et al., Science 319, 1086 (2008)] we developed a stochastic model of interacting single-headed motor proteins KIF1A that also takes into account the interactions between motor proteins and tau molecules. Our model reproduces experimental observations and predicts significant effects of tau on bound time and run length which suggest an important role of tau in regulation of kinesin-based transport.


Subject(s)
Kinesins/chemistry , Models, Chemical , Models, Molecular , Molecular Motor Proteins/chemistry , tau Proteins/chemistry , Binding Sites , Computer Simulation , Kinesins/ultrastructure , Kinetics , Molecular Motor Proteins/ultrastructure , Motion , Protein Binding , Protein Conformation , tau Proteins/ultrastructure
20.
Elife ; 3: e03680, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25209998

ABSTRACT

Kinesins are a superfamily of microtubule-based ATP-powered motors, important for multiple, essential cellular functions. How microtubule binding stimulates their ATPase and controls force generation is not understood. To address this fundamental question, we visualized microtubule-bound kinesin-1 and kinesin-3 motor domains at multiple steps in their ATPase cycles--including their nucleotide-free states--at ∼ 7 Šresolution using cryo-electron microscopy. In both motors, microtubule binding promotes ordered conformations of conserved loops that stimulate ADP release, enhance microtubule affinity and prime the catalytic site for ATP binding. ATP binding causes only small shifts of these nucleotide-coordinating loops but induces large conformational changes elsewhere that allow force generation and neck linker docking towards the microtubule plus end. Family-specific differences across the kinesin-microtubule interface account for the distinctive properties of each motor. Our data thus provide evidence for a conserved ATP-driven mechanism for kinesins and reveal the critical mechanistic contribution of the microtubule interface.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Kinesins/metabolism , Microtubules/metabolism , Adenosine Triphosphatases/metabolism , Binding Sites , Biological Transport , Biomechanical Phenomena , Conserved Sequence , Cryoelectron Microscopy , Humans , Kinesins/chemistry , Kinesins/ultrastructure , Kinetics , Magnesium/metabolism , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...