Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 5833, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37730751

ABSTRACT

Processive transport by the microtubule motor cytoplasmic dynein requires the regulated assembly of a dynein-dynactin-adapter complex. Interactions between dynein and dynactin were initially ascribed to the dynein intermediate chain N-terminus and the dynactin subunit p150Glued. However, recent cryo-EM structures have not resolved this interaction, questioning its importance. The intermediate chain also interacts with Nde1/Ndel1, which compete with p150Glued for binding. We reveal that the intermediate chain N-terminus is a critical evolutionarily conserved hub that interacts with dynactin and Ndel1, the latter of which recruits LIS1 to drive complex assembly. In additon to revealing that the intermediate chain N-terminus is likely bound to p150Glued in active transport complexes, our data support a model whereby Ndel1-LIS1 must dissociate prior to LIS1 being handed off to dynein in temporally discrete steps. Our work reveals previously unknown steps in the dynein activation pathway, and provide insight into the integrated activities of LIS1/Ndel1 and dynactin/cargo-adapters.


Subject(s)
Cytoplasmic Dyneins , Dyneins , Dynactin Complex , Actin Cytoskeleton , Cytoskeleton
3.
Nat Commun ; 14(1): 4715, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37543636

ABSTRACT

Microtubules are major components of the eukaryotic cytoskeleton. Posttranslational modifications (PTMs) of tubulin regulates interactions with microtubule-associated proteins (MAPs). One unique PTM is the cyclical removal and re-addition of the C-terminal tyrosine of α-tubulin and MAPs containing CAP-Gly domains specifically recognize tyrosinated microtubules. KIF13B, a long-distance transport kinesin, contains a conserved CAP-Gly domain, but the role of the CAP-Gly domain in KIF13B's motility along microtubules remains unknown. To address this, we investigate the interaction between KIF13B's CAP-Gly domain, and tyrosinated microtubules. We find that KIF13B's CAP-Gly domain influences the initial motor-microtubule interaction, as well as processive motility along microtubules. The effect of the CAP-Gly domain is enhanced when the motor domain is in the ADP state, suggesting an interplay between the N-terminal motor domain and C-terminal CAP-Gly domain. These results reveal that specialized kinesin tail domains play active roles in the initiation and continuation of motor movement.


Subject(s)
Kinesins , Microtubule-Associated Proteins , Microtubule-Associated Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism , Tubulin/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational
4.
J Cell Biol ; 222(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37265445

ABSTRACT

It is known that microtubule-binding proteins including the Ska1 complex and the DNA replication licensing factor, Cdt1, enable the kinetochore-localized Ndc80 complex to form robust kinetochore-microtubule attachments. However, it is not clear how the Ndc80 complex is stably coupled to dynamic spindle microtubule plus-ends. Here, we have developed a conditional auxin-inducible degron approach to reveal a function for Cdt1 in chromosome segregation and kinetochore-microtubule interactions that is separable from its role in DNA replication licensing. Further, we demonstrate that a direct interaction between Cdt1 and Ska1 is required for recruiting Cdt1 to kinetochores and spindle microtubules. Cdt1 phosphorylation by Cdk1 kinase is critical for Ska1 binding, kinetochore-microtubule attachments, and mitotic progression. Furthermore, we show that Cdt1 synergizes with Ndc80 and Ska1 for microtubule binding, including forming a diffusive, tripartite Ndc80-Cdt1-Ska1 complex that can processively track dynamic microtubule plus-ends in vitro. Taken together, our data identify the Ndc80-Cdt1-Ska1 complex as a central molecular unit that can promote processive bidirectional tip-tracking of microtubules by kinetochores.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Kinetochores , Microtubule-Associated Proteins , Nuclear Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Kinetochores/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Mitosis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism
5.
bioRxiv ; 2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36711700

ABSTRACT

Cytoplasmic dynein, the primary retrograde microtubule transport motor within cells, must be activated for processive motility through the regulated assembly of a dynein-dynactin-adapter (DDA) complex. The interaction between dynein and dynactin was initially ascribed to the N-terminus of the dynein intermediate chain (IC) and a coiled-coil of the dynactin subunit p150 Glued . However, cryo-EM structures of DDA complexes have not resolve these regions of the IC and p150 Glued , raising questions about the importance of this interaction. The IC N-terminus (ICN) also interacts with the dynein regulators Nde1/Ndel1, which compete with p150 Glued for binding to ICN. Using a combination of approaches, we reveal that the ICN plays critical, evolutionarily conserved roles in DDA assembly by interacting with dynactin and Ndel1, the latter of which recruits the DDA assembly factor LIS1 to the dynein complex. In contrast to prior models, we find that LIS1 cannot simultaneously bind to Ndel1 and dynein, indicating that LIS1 must be handed off from Ndel1 to dynein in temporally discrete steps. Whereas exogenous Ndel1 or p150 Glued disrupts DDA complex assembly in vitro , neither perturbs preassembled DDA complexes, indicating that the IC is stably bound to p150 Glued within activated DDA complexes. Our study reveals previously unknown regulatory steps in the dynein activation pathway, and provides a more complete model for how the activities of LIS1/Ndel1 and dynactin/cargo-adapters are integrated to regulate dynein motor activity.

6.
PLoS Genet ; 19(1): e1010457, 2023 01.
Article in English | MEDLINE | ID: mdl-36716349

ABSTRACT

The tumor suppressor BRCA1-BARD1 complex regulates many cellular processes; of critical importance to its tumor suppressor function is its role in genome integrity. Although RING E3 ubiquitin ligase activity is the only known enzymatic activity of the complex, the in vivo requirement for BRCA1-BARD1 E3 ubiquitin ligase activity has been controversial. Here we probe the role of BRCA1-BARD1 E3 ubiquitin ligase activity in vivo using C. elegans. Genetic, cell biological, and biochemical analyses of mutants defective for E3 ligase activity suggest there is both E3 ligase-dependent and independent functions of the complex in the context of DNA damage repair and meiosis. We show that E3 ligase activity is important for nuclear accumulation of the complex and specifically to concentrate at meiotic recombination sites but not at DNA damage sites in proliferating germ cells. While BRCA1 alone is capable of monoubiquitylation, BARD1 is required with BRCA1 to promote polyubiquitylation. We find that the requirement for E3 ligase activity and BARD1 in DNA damage signaling and repair can be partially alleviated by driving the nuclear accumulation and self-association of BRCA1. Our data suggest that in addition to E3 ligase activity, BRCA1 may serve a structural role for DNA damage signaling and repair while BARD1 plays an accessory role to enhance BRCA1 function.


Subject(s)
Caenorhabditis elegans , Tumor Suppressor Proteins , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Tumor Suppressor Proteins/genetics , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , DNA Repair/genetics , DNA Damage/genetics , Meiosis/genetics , Germ Cells/metabolism
7.
Nat Chem Biol ; 18(11): 1224-1235, 2022 11.
Article in English | MEDLINE | ID: mdl-35996000

ABSTRACT

Tau is an intrinsically disordered microtubule-associated protein (MAP) implicated in neurodegenerative disease. On microtubules, tau molecules segregate into two kinetically distinct phases, consisting of either independently diffusing molecules or interacting molecules that form cohesive 'envelopes' around microtubules. Envelopes differentially regulate lattice accessibility for other MAPs, but the mechanism of envelope formation remains unclear. Here we find that tau envelopes form cooperatively, locally altering the spacing of tubulin dimers within the microtubule lattice. Envelope formation compacted the underlying lattice, whereas lattice extension induced tau envelope disassembly. Investigating other members of the tau family, we find that MAP2 similarly forms envelopes governed by lattice spacing, whereas MAP4 cannot. Envelopes differentially biased motor protein movement, suggesting that tau family members could spatially divide the microtubule surface into functionally distinct regions. We conclude that the interdependent allostery between lattice spacing and cooperative envelope formation provides the molecular basis for spatial regulation of microtubule-based processes by tau and MAP2.


Subject(s)
Neurodegenerative Diseases , tau Proteins , Humans , tau Proteins/metabolism , Tubulin/metabolism , Neurodegenerative Diseases/metabolism , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Proteins/metabolism
8.
Cell Rep ; 39(9): 110900, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35649356

ABSTRACT

Kinesin-1 activity is regulated by autoinhibition. Intramolecular interactions within the kinesin heavy chain (KHC) are proposed to be one facet of motor regulation. The KHC also binds to the kinesin light chain (KLC), which has been implicated in both autoinhibition and activation of the motor. We show that the KLC inhibits the kinesin-microtubule interaction independently from the proposed intramolecular interaction within KHC. Cargo-adaptor proteins that bind the KLC stimulated processive movement, but the landing rate of activated kinesin complexes remained low. Mitogen-activated protein 7 (MAP7) enhanced motility by increasing the landing rate and run length of the activated kinesin motors. Our results support a model whereby the motor activity of the kinesin is regulated by synergistic inhibition mechanisms and that cargo-adaptor binding to the KLC releases both mechanisms. However, a non-motor MAP is required for robust microtubule association of the activated motor. Thus, human kinesin is regulated by synergistic autoinhibition and activation mechanisms.


Subject(s)
Kinesins , Microtubules , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Humans , Kinesins/metabolism , Microtubules/metabolism , Motor Activity
10.
Methods Mol Biol ; 2415: 123-138, 2022.
Article in English | MEDLINE | ID: mdl-34972950

ABSTRACT

The separation of duplicated chromosomes during mitosis is a pivotal step in the process of cellular division. Therefore, the orchestrated events that take place to ensure proper attachment and stabilization of kMTs are keen areas of interest in the mitosis field. Here we describe the methods used to study kMT attachments via in vitro biochemical methods and in vivo cell biological approaches.


Subject(s)
Kinetochores , Microtubules , Chromosome Segregation , Mitosis , Spindle Apparatus
11.
Sci Adv ; 7(18)2021 04.
Article in English | MEDLINE | ID: mdl-33931448

ABSTRACT

KIF1A is a critical cargo transport motor within neurons. More than 100 known mutations result in KIF1A-associated neurological disorder (KAND), a degenerative condition for which there is no cure. A missense mutation, P305L, was identified in children diagnosed with KAND, but the molecular basis for the disease is unknown. We find that this conserved residue is part of an unusual 310 helix immediately adjacent to the family-specific K-loop, which facilitates a high microtubule-association rate. We find that the mutation negatively affects several biophysical parameters of the motor. However, the microtubule-association rate of the motor is most markedly affected, revealing that the presence of an intact K-loop is not sufficient for its function. We hypothesize that the 310 helix facilitates a specific K-loop conformation that is critical for its function. We find that the function of this proline is conserved in kinesin-1, revealing a fundamental principle of the kinesin motor mechanism.


Subject(s)
Kinesins , Microtubules , Child , Humans , Kinesins/genetics , Mutation , Mutation, Missense , Neurons
12.
J Med Genet ; 58(4): 254-263, 2021 04.
Article in English | MEDLINE | ID: mdl-32527956

ABSTRACT

BACKGROUND: Mutation in S-phase cyclin A-associated protein rin the endoplasmic reticulum (SCAPER) have been found across ethnicities and have been shown to cause variable penetrance of an array of pathological traits, including intellectual disability, retinitis pigmentosa and ciliopathies. METHODS: Human clinical phenotyping, surgical testicular sperm extraction and testicular tissue staining. Generation and analysis of short spindle 3 (ssp3) (SCAPER orthologue) Drosophila CAS9-knockout lines. In vitro microtubule (MT) binding assayed by total internal reflection fluorescence microscopy. RESULTS: We show that patients homozygous for a SCAPER mutation lack SCAPER expression in spermatogonia (SPG) and are azoospermic due to early defects in spermatogenesis, leading to the complete absence of meiotic cells. Interestingly, Drosophila null mutants for the ubiquitously expressed ssp3 gene are viable and female fertile but male sterile. We further show that male sterility in ssp3 null mutants is due to failure in both chromosome segregation and cytokinesis. In cells undergoing male meiosis, the MTs emanating from the centrosomes do not appear to interact properly with the chromosomes, which remain dispersed within dividing spermatocytes (SPCs). In addition, mutant SPCs are unable to assemble a normal central spindle and undergo cytokinesis. Consistent with these results, an in vitro assay demonstrated that both SCAPER and Ssp3 directly bind MTs. CONCLUSIONS: Our results show that SCAPER null mutations block the entry into meiosis of SPG, causing azoospermia. Null mutations in ssp3 specifically disrupt MT dynamics during male meiosis, leading to sterility. Moreover, both SCAPER and Ssp3 bind MTs in vitro. These results raise the intriguing possibility of a common feature between human and Drosophila meiosis.


Subject(s)
Carrier Proteins/genetics , Infertility, Male/genetics , Microtubules/genetics , Serine Endopeptidases/genetics , Animals , Chromosome Segregation/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Genetic Predisposition to Disease , Humans , Infertility, Male/pathology , Male , Meiosis/genetics , Mutation/genetics , Spermatocytes/growth & development , Spermatocytes/pathology , Spindle Apparatus/genetics , Spindle Apparatus/pathology , Testis/growth & development , Testis/pathology
13.
Elife ; 92020 07 21.
Article in English | MEDLINE | ID: mdl-32692650

ABSTRACT

Lissencephaly ('smooth brain') is a severe brain disease associated with numerous symptoms, including cognitive impairment, and shortened lifespan. The main causative gene of this disease - lissencephaly-1 (LIS1) - has been a focus of intense scrutiny since its first identification almost 30 years ago. LIS1 is a critical regulator of the microtubule motor cytoplasmic dynein, which transports numerous cargoes throughout the cell, and is a key effector of nuclear and neuronal transport during brain development. Here, we review the role of LIS1 in cellular dynein function and discuss recent key findings that have revealed a new mechanism by which this molecule influences dynein-mediated transport. In addition to reconciling prior observations with this new model for LIS1 function, we also discuss phylogenetic data that suggest that LIS1 may have coevolved with an autoinhibitory mode of cytoplasmic dynein regulation.


Subject(s)
Brain/growth & development , Cell Movement/physiology , Dyneins/metabolism , Lissencephaly/complications , Lissencephaly/genetics , Lissencephaly/physiopathology , Microtubule-Associated Proteins/metabolism , Animals , Cell Movement/genetics , Humans , Microtubule-Associated Proteins/genetics , Models, Animal
14.
Nat Cell Biol ; 22(5): 515-517, 2020 05.
Article in English | MEDLINE | ID: mdl-32341546
15.
Dev Cell ; 53(1): 60-72.e4, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32109385

ABSTRACT

Many eukaryotic cells distribute their intracellular components asymmetrically through regulated active transport driven by molecular motors along microtubule tracks. While intrinsic and extrinsic regulation of motor activity exists, what governs the overall distribution of activated motor-cargo complexes within cells remains unclear. Here, we utilize in vitro reconstitution of purified motor proteins and non-enzymatic microtubule-associated proteins (MAPs) to demonstrate that MAPs exhibit distinct influences on the motility of the three main classes of transport motors: kinesin-1, kinesin-3, and cytoplasmic dynein. Further, we dissect how combinations of MAPs affect motors and unveil MAP9 as a positive modulator of kinesin-3 motility. From these data, we propose a general "MAP code" that has the capacity to strongly bias directed movement along microtubules and helps elucidate the intricate intracellular sorting observed in highly polarized cells such as neurons.


Subject(s)
Dyneins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Transport/physiology , Animals , Biological Transport/physiology , Cell Movement/physiology , Cytoplasm/metabolism , Kinesins/metabolism
16.
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
17.
Nat Cell Biol ; 21(9): 1078-1085, 2019 09.
Article in English | MEDLINE | ID: mdl-31481790

ABSTRACT

Tau is an abundant microtubule-associated protein in neurons. Tau aggregation into insoluble fibrils is a hallmark of Alzheimer's disease and other types of dementia1, yet the physiological state of tau molecules within cells remains unclear. Using single-molecule imaging, we directly observe that the microtubule lattice regulates reversible tau self-association, leading to localized, dynamic condensation of tau molecules on the microtubule surface. Tau condensates form selectively permissible barriers, spatially regulating the activity of microtubule-severing enzymes and the movement of molecular motors through their boundaries. We propose that reversible self-association of tau molecules, gated by the microtubule lattice, is an important mechanism of the biological functions of tau, and that oligomerization of tau is a common property shared between the physiological and disease-associated forms of the molecule.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spastin/metabolism , tau Proteins/metabolism , Animals , Mice , Neuroimaging/methods , Neurons/metabolism , Swine
18.
Nat Chem Biol ; 15(11): 1033-1034, 2019 11.
Article in English | MEDLINE | ID: mdl-31501591
19.
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
20.
J Biol Chem ; 294(22): 8779-8790, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30992364

ABSTRACT

Tau, a member of the MAP2/tau family of microtubule-associated proteins, stabilizes and organizes axonal microtubules in healthy neurons. In neurodegenerative tauopathies, tau dissociates from microtubules and forms neurotoxic extracellular aggregates. MAP2/tau family proteins are characterized by three to five conserved, intrinsically disordered repeat regions that mediate electrostatic interactions with the microtubule surface. Here, we used molecular dynamics, microtubule-binding experiments, and live-cell microscopy, revealing that highly-conserved histidine residues near the C terminus of each microtubule-binding repeat are pH sensors that can modulate tau-microtubule interaction strength within the physiological intracellular pH range. We observed that at low pH (<7.5), these histidines are positively charged and interact with phenylalanine residues in a hydrophobic cleft between adjacent tubulin dimers. At higher pH (>7.5), tau deprotonation decreased binding to microtubules both in vitro and in cells. Electrostatic and hydrophobic characteristics of histidine were both required for tau-microtubule binding, as substitutions with constitutively and positively charged nonaromatic lysine or uncharged alanine greatly reduced or abolished tau-microtubule binding. Consistent with these findings, tau-microtubule binding was reduced in a cancer cell model with increased intracellular pH but was rapidly restored by decreasing the pH to normal levels. These results add detailed insights into the intracellular regulation of tau activity that may be relevant in both normal and pathological conditions.


Subject(s)
Histidine/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...