Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Cell Sci ; 137(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39239883

ABSTRACT

KIF1A/UNC-104 proteins, which are members of the kinesin superfamily of motor proteins, play a pivotal role in the axonal transport of synaptic vesicles and their precursors. Drosophila melanogaster UNC-104 (DmUNC-104) is a relatively recently discovered Drosophila kinesin. Although some point mutations that disrupt synapse formation have been identified, the biochemical properties of the DmUNC-104 protein have not been investigated. Here, we prepared recombinant full-length DmUNC-104 protein and determined its biochemical features. We analyzed the effect of a previously identified missense mutation in the forkhead-associated (FHA) domain, called bristly (bris). The bris mutation strongly promoted the dimerization of DmUNC-104 protein, whereas wild-type DmUNC-104 was a mixture of monomers and dimers. We further tested the G618R mutation near the FHA domain, which was previously shown to disrupt the autoinhibition of Caenorhabditis elegans UNC-104. The biochemical properties of the G618R mutant recapitulated those of the bris mutant. Finally, we found that disease-associated mutations also promote the dimerization of DmUNC-104. Collectively, our results suggest that the FHA domain is essential for autoinhibition of KIF1A/UNC-104 proteins, and that abnormal dimerization of KIF1A might be linked to human diseases.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Kinesins , Animals , Kinesins/metabolism , Kinesins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Protein Domains , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Mutation, Missense , Protein Multimerization , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Humans
2.
J Cell Sci ; 137(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39279507

ABSTRACT

The axonal transport of synaptic vesicle precursors relies on KIF1A and UNC-104 ortholog motors. In mammals, KIF1Bß is also responsible for the axonal transport of synaptic vesicle precursors. Mutations in KIF1A and KIF1Bß lead to a wide range of neuropathies. Although previous studies have revealed the biochemical, biophysical and cell biological properties of KIF1A, and its defects in neurological disorders, the fundamental properties of KIF1Bß remain elusive. In this study, we determined the motile parameters of KIF1Bß through single-molecule motility assays. We found that the C-terminal region of KIF1Bß has an inhibitory role in motor activity. AlphaFold2 prediction suggests that the C-terminal region blocks the motor domain. Additionally, we established simple methods for testing the axonal transport activity of human KIF1Bß using Caenorhabditis elegans genetics. Taking advantage of these methods, we demonstrated that these assays enable the detection of reduced KIF1Bß activities, both in vitro and in vivo, caused by a Charcot-Marie-Tooth disease-associated Q98L mutation.


Subject(s)
Axonal Transport , Caenorhabditis elegans , Kinesins , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Kinesins/metabolism , Kinesins/genetics , Animals , Humans , Axonal Transport/genetics , Single Molecule Imaging/methods , Mutation/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Synaptic Vesicles/metabolism , Synaptic Vesicles/genetics
3.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36655764

ABSTRACT

Neuronal function depends on axonal transport by kinesin superfamily proteins (KIFs). KIF1A is the molecular motor that transports synaptic vesicle precursors, synaptic vesicles, dense core vesicles and active zone precursors. KIF1A is regulated by an autoinhibitory mechanism; many studies, as well as the crystal structure of KIF1A paralogs, support a model whereby autoinhibited KIF1A is monomeric in solution, whereas activated KIF1A is dimeric on microtubules. KIF1A-associated neurological disorder (KAND) is a broad-spectrum neuropathy that is caused by mutations in KIF1A. More than 100 point mutations have been identified in KAND. In vitro assays show that most mutations are loss-of-function mutations that disrupt the motor activity of KIF1A, whereas some mutations disrupt its autoinhibition and abnormally hyperactivate KIF1A. Studies on disease model worms suggests that both loss-of-function and gain-of-function mutations cause KAND by affecting the axonal transport and localization of synaptic vesicles. In this Review, we discuss how the analysis of these mutations by molecular genetics, single-molecule assays and force measurements have helped to reveal the physiological significance of KIF1A function and regulation, and what physical parameters of KIF1A are fundamental to axonal transport.


Subject(s)
Axonal Transport , Nervous System Diseases , Humans , Axonal Transport/genetics , Axonal Transport/physiology , Kinesins/genetics , Kinesins/metabolism , Microtubules/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neurons/metabolism , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
4.
Proc Natl Acad Sci U S A ; 119(32): e2113795119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35917346

ABSTRACT

KIF1A is a kinesin superfamily motor protein that transports synaptic vesicle precursors in axons. Cargo binding stimulates the dimerization of KIF1A molecules to induce processive movement along microtubules. Mutations in human Kif1a lead to a group of neurodegenerative diseases called KIF1A-associated neuronal disorder (KAND). KAND mutations are mostly de novo and autosomal dominant; however, it is unknown if the function of wild-type KIF1A motors is inhibited by heterodimerization with mutated KIF1A. Here, we have established Caenorhabditis elegans models for KAND using CRISPR-Cas9 technology and analyzed the effects of human KIF1A mutation on axonal transport. In our C. elegans models, both heterozygotes and homozygotes exhibited reduced axonal transport. Suppressor screening using the disease model identified a mutation that recovers the motor activity of mutated human KIF1A. In addition, we developed in vitro assays to analyze the motility of heterodimeric motors composed of wild-type and mutant KIF1A. We find that mutant KIF1A significantly impaired the motility of heterodimeric motors. Our data provide insight into the molecular mechanism underlying the dominant nature of de novo KAND mutations.


Subject(s)
Axonal Transport , Caenorhabditis elegans , Kinesins , Neurodegenerative Diseases , Synaptic Vesicles , Animals , Axonal Transport/genetics , Caenorhabditis elegans/genetics , Disease Models, Animal , Genes, Dominant , Humans , Kinesins/genetics , Motor Activity/genetics , Mutation , Neurodegenerative Diseases/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism
5.
Genes Cells ; 28(2): 97-110, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36461782

ABSTRACT

Dynein is a microtubule-dependent motor protein required for cell division, retrograde intracellular transport, and intraflagellar transport (IFT). Dynein 1 and dynein 2 serve as molecular motors in the cytoplasm and cilia, respectively. Each dynein consists of multiple subunits. Although the components of dynein 1 and dynein 2 are different and specific in most species, a previous study has suggested that dynein intermediate chain subunit DYCI-1 is shared by both dynein 1 and 2 in Caenorhabditis elegans (C. elegans). Here, we show that C. elegans has two dynein intermediate chains-DYCI-1 and WDR-60-and their functions are different. Mutational analysis showed that dyci-1 is essential for the retrograde axonal transport of synaptic vesicles. In the same mutant allele, IFT is not affected at all. Instead, wdr-60 is essential for IFT. Thus, we suggest that dynein 1 and dynein 2 have specific intermediate chains in C. elegans as in other organisms.


Subject(s)
Caenorhabditis elegans Proteins , Carrier Proteins , Cytoskeletal Proteins , Dyneins , Animals , Biological Transport , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cilia/metabolism , Dyneins/metabolism , Flagella/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism
6.
Cell ; 139(4): 802-13, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914172

ABSTRACT

The kinesin superfamily proteins (KIFs) are motor proteins that transport organelles and protein complexes in a microtubule- and ATP-dependent manner. We identified KIF26A as a new member of the murine KIFs. KIF26A is a rather atypical member as it lacks ATPase activity. Mice with a homozygous deletion of Kif26a developed a megacolon with enteric nerve hyperplasia. Kif26a-/- enteric neurons showed hypersensitivity for GDNF-Ret signaling, and we find that KIF26A suppressed GDNF-Ret signaling by direct binding and inhibition of Grb2, an essential component of GDNF/Akt/ERK signaling. We therefore propose that the unconventional kinesin KIF26A plays a key role in enteric nervous system development by repressing a cell growth signaling pathway.


Subject(s)
Enteric Nervous System/embryology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hirschsprung Disease/metabolism , Kinesins/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction , Animals , Cell Growth Processes , Cell Line , Colon/cytology , Colon/embryology , Colon/innervation , GRB2 Adaptor Protein/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/metabolism
7.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33542099

ABSTRACT

Caenorhabditis elegans is used as a model system to understand the neural basis of behavior, but application of caged compounds to manipulate and monitor the neural activity is hampered by the innate photophobic response of the nematode to short-wavelength light or by the low temporal resolution of photocontrol. Here, we develop boron dipyrromethene (BODIPY)-derived caged compounds that release bioactive phenol derivatives upon illumination in the yellow wavelength range. We show that activation of the transient receptor potential vanilloid 1 (TRPV1) cation channel by spatially targeted optical uncaging of the TRPV1 agonist N-vanillylnonanamide at 580 nm modulates neural activity. Further, neuronal activation by illumination-induced uncaging enables optical control of the behavior of freely moving C. elegans without inducing a photophobic response and without crosstalk between uncaging and simultaneous fluorescence monitoring of neural activity.


Subject(s)
Behavior Control , Caenorhabditis elegans/physiology , Caenorhabditis elegans/radiation effects , Light , Neurons/physiology , Neurons/radiation effects , Animals , Fluorescence , Interneurons/physiology , Promoter Regions, Genetic/genetics , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism
8.
Biophys J ; 122(22): 4348-4359, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37853694

ABSTRACT

KIF1A is a member of the kinesin-3 motor protein family that transports synaptic vesicle precursors in axons. Mutations in the Kif1a gene cause neuronal diseases. Most patients are heterozygous and have both mutated and intact KIF1A alleles, suggesting that heterodimers composed of wild-type KIF1A and mutant KIF1A are likely involved in pathogenesis. In this study, we propose mathematical models to describe the motility of KIF1A heterodimers composed of wild-type KIF1A and mutant KIF1A. Our models precisely describe run length, run time, and velocity of KIF1A heterodimers using a few parameters obtained from two homodimers. The first model is a simple hand-over-hand model in which stepping and detachment rates from a microtubule of each head are identical to those in the respective homodimers. Although the velocities of heterodimers expected from this model were in good agreement with the experimental results, this model underestimated the run lengths and run times of some heterodimeric motors. To address this discrepancy, we propose the tethered-head affinity model, in which we hypothesize a tethered head, in addition to a microtubule-binding head, contributes to microtubule binding in a vulnerable one-head-bound state. The run lengths and run times of the KIF1A heterodimers predicted by the tethered-head affinity model matched well with experimental results, suggesting a possibility that the tethered head affects the microtubule binding of KIF1A. Our models provide insights into how each head contributes to the processive movement of KIF1A and can be used to estimate motile parameters of KIF1A heterodimers.


Subject(s)
Axons , Kinesins , Humans , Kinesins/genetics , Kinesins/metabolism , Axons/metabolism , Neurons/metabolism , Microtubules/metabolism , Synaptic Vesicles/metabolism
9.
Genes Cells ; 27(6): 421-435, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35430760

ABSTRACT

KIF5A is a kinesin superfamily motor protein that transports various cargos in neurons. Mutations in Kif5a cause familial amyotrophic lateral sclerosis (ALS). These ALS mutations are in the intron of Kif5a and induce mis-splicing of KIF5A mRNA, leading to splicing out of exon 27, which in human KIF5A encodes the cargo-binding tail domain of KIF5A. Therefore, it has been suggested that ALS is caused by loss of function of KIF5A. However, the precise mechanisms regarding how mutations in KIF5A cause ALS remain unclear. Here, we show that an ALS-associated mutant of KIF5A, KIF5A(Δexon27), is predisposed to form oligomers and aggregates in cultured mouse cell lines. Interestingly, purified KIF5A(Δexon27) oligomers showed more active movement on microtubules than wild-type KIF5A in vitro. Purified KIF5A(∆exon27) was prone to form aggregates in vitro. Moreover, KIF5A(Δexon27)-expressing Caenorhabditis elegans neurons showed morphological defects. These data collectively suggest that ALS-associated mutations of KIF5A are toxic gain-of-function mutations rather than simple loss-of-function mutations.


Subject(s)
Amyotrophic Lateral Sclerosis , Kinesins , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Dyneins/genetics , Dyneins/metabolism , Kinesins/genetics , Kinesins/metabolism , Mice , Mutation , Neurons/metabolism , Neurons/pathology , Protein Aggregation, Pathological
10.
Proc Natl Acad Sci U S A ; 116(37): 18429-18434, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31455732

ABSTRACT

KIF1A is a kinesin family motor involved in the axonal transport of synaptic vesicle precursors (SVPs) along microtubules (MTs). In humans, more than 10 point mutations in KIF1A are associated with the motor neuron disease hereditary spastic paraplegia (SPG). However, not all of these mutations appear to inhibit the motility of the KIF1A motor, and thus a cogent molecular explanation for how KIF1A mutations lead to neuropathy is not available. In this study, we established in vitro motility assays with purified full-length human KIF1A and found that KIF1A mutations associated with the hereditary SPG lead to hyperactivation of KIF1A motility. Introduction of the corresponding mutations into the Caenorhabditis elegans KIF1A homolog unc-104 revealed abnormal accumulation of SVPs at the tips of axons and increased anterograde axonal transport of SVPs. Our data reveal that hyperactivation of kinesin motor activity, rather than its loss of function, is a cause of motor neuron disease in humans.


Subject(s)
Axonal Transport/genetics , Genetic Predisposition to Disease/genetics , Kinesins/genetics , Kinesins/metabolism , Mutation , Synaptic Vesicles/metabolism , Animals , Axons/metabolism , Caenorhabditis elegans/genetics , Humans , Motor Neuron Disease/genetics , Spastic Paraplegia, Hereditary/genetics
11.
Biophys J ; 120(9): 1605-1614, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33617835

ABSTRACT

Synaptic cargo transport by kinesin and dynein in hippocampal neurons was investigated by noninvasively measuring the transport force based on nonequilibrium statistical mechanics. Although direct physical measurements such as force measurement using optical tweezers are difficult in an intracellular environment, the noninvasive estimations enabled enumerating force-producing units (FPUs) carrying a cargo comprising the motor proteins generating force. The number of FPUs served as a barometer for stable and long-distance transport by multiple motors, which was then used to quantify the extent of damage to axonal transport by dynarrestin, a dynein inhibitor. We found that dynarrestin decreased the FPU for retrograde transport more than for anterograde transport. This result indicates the applicability of the noninvasive force measurements. In the future, these measurements may be used to quantify damage to axonal transport resulting from neuronal diseases, including Alzheimer's, Parkinson's, and Huntington's diseases.


Subject(s)
Dyneins , Kinesins , Axonal Transport , Dyneins/metabolism , Kinesins/metabolism , Myosins , Optical Tweezers
12.
Nat Rev Mol Cell Biol ; 10(10): 682-96, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19773780

ABSTRACT

Intracellular transport is fundamental for cellular function, survival and morphogenesis. Kinesin superfamily proteins (also known as KIFs) are important molecular motors that directionally transport various cargos, including membranous organelles, protein complexes and mRNAs. The mechanisms by which different kinesins recognize and bind to specific cargos, as well as how kinesins unload cargo and determine the direction of transport, have now been identified. Furthermore, recent molecular genetic experiments have uncovered important and unexpected roles for kinesins in the regulation of such physiological processes as higher brain function, tumour suppression and developmental patterning. These findings open exciting new areas of kinesin research.


Subject(s)
Kinesins/metabolism , Kinesins/physiology , Molecular Motor Proteins/metabolism , Animals , Biological Transport/genetics , Dyneins/genetics , Dyneins/metabolism , Humans , Kinesins/chemistry , Kinesins/classification , Kinesins/genetics , Models, Biological , Molecular Motor Proteins/genetics , Organelles/genetics , Organelles/metabolism , Phylogeny , Proteins/metabolism , RNA, Messenger/metabolism
13.
Org Biomol Chem ; 17(28): 6771-6776, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31259993

ABSTRACT

Design, synthesis and functional analysis of fluorescent derivatives of neopeltolide, an antiproliferative marine macrolide, are reported herein. Live cell imaging using the fluorescent derivatives showed rapid cellular uptake and localization within the endoplasmic reticulum as well as the mitochondria.


Subject(s)
Fluorescence , Fluorescent Dyes/chemistry , Macrolides/chemistry , Optical Imaging , A549 Cells , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/drug effects , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacology , Humans , Macrolides/chemical synthesis , Macrolides/pharmacology , Mitochondria/chemistry , Mitochondria/drug effects , Molecular Conformation , Tissue Distribution
14.
Phys Chem Chem Phys ; 20(5): 3403-3410, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-29349444

ABSTRACT

Kinesin superfamily protein UNC-104, a member of the kinesin-3 family, transports synaptic vesicle precursors (SVPs). In this study, the number of active UNC-104 molecules hauling a single SVP in axons in the worm Caenorhabditis elegans was counted by applying a newly developed non-invasive force measurement technique. The distribution of the force acting on a SVP transported by UNC-104 was spread out over several clusters, implying the presence of several force-producing units (FPUs). We then compared the number of FPUs in the wild-type worms with that in arl-8 gene-deletion mutant worms. ARL-8 is a SVP-bound arf-like small guanosine triphosphatase, and is known to promote unlocking of the autoinhibition of the motor, which is critical for avoiding unnecessary consumption of adenosine triphosphate when the motor does not bind to a SVP. There were fewer FPUs in the arl-8 mutant worms. This finding indicates that a lack of ARL-8 decreased the number of active UNC-104 motors, which then led to a decrease in the number of motors responsible for SVP transport.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , GTP Phosphohydrolases/metabolism , Kinesins/metabolism , Synaptic Vesicles/metabolism , Animals , Axonal Transport , Axons/metabolism , Caenorhabditis elegans , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Kinesins/chemistry , Microscopy, Fluorescence , Mutagenesis , Synaptic Vesicles/chemistry
15.
EMBO J ; 32(10): 1352-64, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23503589

ABSTRACT

Microtubules are fundamental to neuronal morphogenesis and function. Mutations in tubulin, the major constituent of microtubules, result in neuronal diseases. Here, we have analysed ß-tubulin mutations that cause neuronal diseases and we have identified mutations that strongly inhibit axonal transport of vesicles and mitochondria. These mutations are in the H12 helix of ß-tubulin and change the negative charge on the surface of the microtubule. This surface is the interface between microtubules and kinesin superfamily motor proteins (KIF). The binding of axonal transport KIFs to microtubules is dominant negatively disrupted by these mutations, which alters the localization of KIFs in neurons and inhibits axon elongation in vivo. In humans, these mutations induce broad neurological symptoms, such as loss of axons in the central nervous system and peripheral neuropathy. Thus, our data identified the critical region of ß-tubulin required for axonal transport and suggest a molecular mechanism for human neuronal diseases caused by tubulin mutations.


Subject(s)
Axonal Transport/genetics , Mutation , Peripheral Nervous System Diseases/genetics , Tubulin/genetics , Tubulin/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Kinesins/metabolism , Microtubules/genetics , Microtubules/metabolism , Mitochondria/metabolism , Motor Neuron Disease/genetics , Neurons/metabolism , Protein Structure, Tertiary , Tubulin/chemistry , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
16.
J Neurosci ; 35(12): 5067-86, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25810535

ABSTRACT

An organelle's subcellular localization is closely related to its function. Early endosomes require localization to somatodendritic regions in neurons to enable neuronal morphogenesis, polarized sorting, and signal transduction. However, it is not known how the somatodendritic localization of early endosomes is achieved. Here, we show that the kinesin superfamily protein 16B (KIF16B) is essential for the correct localization of early endosomes in mouse hippocampal neurons. Loss of KIF16B induced the aggregation of early endosomes and perturbed the trafficking and functioning of receptors, including the AMPA and NGF receptors. This defect was rescued by KIF16B, emphasizing the critical functional role of the protein in early endosome and receptor transport. Interestingly, in neurons expressing a KIF16B deletion mutant lacking the second and third coiled-coils of the stalk domain, the early endosomes were mistransported to the axons. Additionally, the binding of the motor domain of KIF16B to microtubules was inhibited by the second and third coiled-coils (inhibitory domain) in an ATP-dependent manner. This suggests that the intramolecular binding we find between the inhibitory domain and motor domain of KIF16B may serve as a switch to control the binding of the motor to microtubules, thereby regulating KIF16B activity. We propose that this novel autoregulatory "stalk inhibition" mechanism underlies the ability of KIF16B to potentiate the selective somatodendritic localization of early endosomes.


Subject(s)
Dendrites/metabolism , Endosomes/metabolism , Kinesins/physiology , Neurons/cytology , Neurons/metabolism , Animals , Cells, Cultured , Gene Knockdown Techniques , Hippocampus/metabolism , Humans , Kinesins/genetics , Mice , Microtubules/metabolism , Protein Binding/genetics , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Transport/genetics , Protein Transport/physiology , Receptor, Nerve Growth Factor/metabolism , Receptors, AMPA/metabolism , Sequence Deletion
17.
Proc Natl Acad Sci U S A ; 109(5): 1725-30, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307638

ABSTRACT

Neuronal morphology is regulated by cytoskeletons. Kinesin superfamily protein 2A (KIF2A) depolymerizes microtubules (MTs) at growth cones and regulates axon pathfinding. The factors controlling KIF2A in neurite development remain totally elusive. Here, using immunoprecipitation with an antibody specific to KIF2A, we identified phosphatidylinositol 4-phosphate 5-kinase alpha (PIPKα) as a candidate membrane protein that regulates the activity of KIF2A. Yeast two-hybrid and biochemical assays demonstrated direct binding between KIF2A and PIPKα. Partial colocalization of the clusters of punctate signals for these two molecules was detected by confocal microscopy and photoactivated localization microscopy. Additionally, the MT-depolymerizing activity of KIF2A was enhanced in the presence of PIPKα in vitro and in vivo. PIPKα suppressed the elongation of axon branches in a KIF2A-dependent manner, suggesting a unique PIPK-mediated mechanism controlling MT dynamics in neuronal development.


Subject(s)
Axons , Kinesins/metabolism , Microtubules/metabolism , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Repressor Proteins/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Gene Knockdown Techniques , Kinesins/genetics , Mice , Microscopy, Fluorescence , Phosphotransferases (Alcohol Group Acceptor)/genetics , Repressor Proteins/genetics
18.
Curr Opin Cell Biol ; 86: 102301, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096601

ABSTRACT

Kinesin-1, composed of kinesin heavy chain and kinesin light chain, is a founding member of kinesin superfamily and transports various neuronal cargos. Kinesin-1 is one of the most abundant ATPases in the cell and thus need to be tightly regulated to avoid wastage of energy. It has been well established that kinesin-1 is regulated by the autoinhibition mechanism. This review focuses on the recent researches that have contributed to the understanding of mechanisms for the autoinhibition of kinesin-1 and its unlocking. Recent electron microscopic studies have shown an unanticipated structure of autoinhibited kinesin-1. Biochemical reconstitution have revealed detailed molecular mechanisms how the autoinhibition is unlocked. Importantly, misregulation of kinesin-1 is emerging as one of the major causes of amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis , Kinesins , Humans , Kinesins/metabolism , Neurons/metabolism , Biological Transport
19.
Elife ; 122024 Jan 11.
Article in English | MEDLINE | ID: mdl-38206323

ABSTRACT

Kinesin-3 is a family of microtubule-dependent motor proteins that transport various cargos within the cell. However, the mechanism underlying kinesin-3 activations remains largely elusive. In this study, we compared the biochemical properties of two Caenorhabditis elegans kinesin-3 family proteins, KLP-6 and UNC-104. Both KLP-6 and UNC-104 are predominantly monomeric in solution. As previously shown for UNC-104, non-processive KLP-6 monomer is converted to a processive motor when artificially dimerized. We present evidence that releasing the autoinhibition is sufficient to trigger dimerization of monomeric UNC-104 at nanomolar concentrations, which results in processive movement of UNC-104 on microtubules, although it has long been thought that enrichment in the phospholipid microdomain on cargo vesicles is required for the dimerization and processive movement of UNC-104. In contrast, KLP-6 remains to be a non-processive monomer even when its autoinhibition is unlocked, suggesting a requirement of other factors for full activation. By examining the differences between KLP-6 and UNC-104, we identified a coiled-coil domain called coiled-coil 2 (CC2) that is required for the efficient dimerization and processive movement of UNC-104. Our results suggest a common activation mechanism for kinesin-3 family members, while also highlighting their diversification.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Kinesins , Nerve Tissue Proteins , Animals , Caenorhabditis elegans Proteins/genetics , Kinesins/genetics , Microtubule Proteins , Nerve Tissue Proteins/genetics , Protein Multimerization
20.
Curr Biol ; 34(16): 3747-3762.e6, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39163829

ABSTRACT

The acentrosomal spindle apparatus has kinetochore fibers organized and converged toward opposite poles; however, mechanisms underlying the organization of these microtubule fibers into an orchestrated bipolar array were largely unknown. Kinesin-14D is one of the four classes of Kinesin-14 motors that are conserved from green algae to flowering plants. In Arabidopsis thaliana, three Kinesin-14D members displayed distinct cell cycle-dependent localization patterns on spindle microtubules in mitosis. Notably, Kinesin-14D1 was enriched on the midzone microtubules of prophase and mitotic spindles and later persisted in the spindle and phragmoplast midzones. The kinesin-14d1 mutant had kinetochore fibers disengaged from each other during mitosis and exhibited hypersensitivity to the microtubule-depolymerizing herbicide oryzalin. Oryzalin-treated kinesin-14d1 mutant cells had kinetochore fibers tangled together in collapsed spindle microtubule arrays. Kinesin-14D1, unlike other Kinesin-14 motors, showed slow microtubule plus end-directed motility, and its localization and function were dependent on its motor activity and the novel malectin-like domain. Our findings revealed a Kinesin-14D1-dependent mechanism that employs interpolar microtubules to regulate the organization of kinetochore fibers for acentrosomal spindle morphogenesis.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Kinesins , Microtubules , Spindle Apparatus , Arabidopsis/metabolism , Arabidopsis/genetics , Kinesins/metabolism , Kinesins/genetics , Microtubules/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Spindle Apparatus/metabolism , Mitosis , Morphogenesis , Kinetochores/metabolism , Dinitrobenzenes/pharmacology , Sulfanilamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL