Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.354
Filter
Add more filters

Publication year range
1.
Cell ; 186(12): 2531-2543.e11, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37295401

ABSTRACT

RNA editing is a widespread epigenetic process that can alter the amino acid sequence of proteins, termed "recoding." In cephalopods, most transcripts are recoded, and recoding is hypothesized to be an adaptive strategy to generate phenotypic plasticity. However, how animals use RNA recoding dynamically is largely unexplored. We investigated the function of cephalopod RNA recoding in the microtubule motor proteins kinesin and dynein. We found that squid rapidly employ RNA recoding in response to changes in ocean temperature, and kinesin variants generated in cold seawater displayed enhanced motile properties in single-molecule experiments conducted in the cold. We also identified tissue-specific recoded squid kinesin variants that displayed distinct motile properties. Finally, we showed that cephalopod recoding sites can guide the discovery of functional substitutions in non-cephalopod kinesin and dynein. Thus, RNA recoding is a dynamic mechanism that generates phenotypic plasticity in cephalopods and can inform the characterization of conserved non-cephalopod proteins.


Subject(s)
Cephalopoda , Dyneins , Animals , Dyneins/genetics , Dyneins/metabolism , Kinesins/genetics , Kinesins/metabolism , RNA/metabolism , Cephalopoda/genetics , Cephalopoda/metabolism , Proteins/metabolism , Microtubules/metabolism , Microtubule Proteins , Myosins/metabolism
2.
Cell ; 185(26): 4971-4985.e16, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36462505

ABSTRACT

Intraflagellar transport (IFT) trains are massive molecular machines that traffic proteins between cilia and the cell body. Each IFT train is a dynamic polymer of two large complexes (IFT-A and -B) and motor proteins, posing a formidable challenge to mechanistic understanding. Here, we reconstituted the complete human IFT-A complex and obtained its structure using cryo-EM. Combined with AlphaFold prediction and genome-editing studies, our results illuminate how IFT-A polymerizes, interacts with IFT-B, and uses an array of ß-propeller and TPR domains to create "carriages" of the IFT train that engage TULP adaptor proteins. We show that IFT-A⋅TULP carriages are essential for cilia localization of diverse membrane proteins, as well as ICK-the key kinase regulating IFT train turnaround. These data establish a structural link between IFT-A's distinct functions, provide a blueprint for IFT-A in the train, and shed light on how IFT evolved from a proto-coatomer ancestor.


Subject(s)
Cilia , Kinesins , Humans , Cilia/metabolism , Biological Transport , Kinesins/metabolism , Dyneins/metabolism , Membrane Proteins/metabolism , Protein Transport , Flagella/metabolism
3.
Cell ; 175(3): 796-808.e14, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340043

ABSTRACT

During cell division, mitotic motors organize microtubules in the bipolar spindle into either polar arrays at the spindle poles or a "nematic" network of aligned microtubules at the spindle center. The reasons for the distinct self-organizing capacities of dynamic microtubules and different motors are not understood. Using in vitro reconstitution experiments and computer simulations, we show that the human mitotic motors kinesin-5 KIF11 and kinesin-14 HSET, despite opposite directionalities, can both organize dynamic microtubules into either polar or nematic networks. We show that in addition to the motor properties the natural asymmetry between microtubule plus- and minus-end growth critically contributes to the organizational potential of the motors. We identify two control parameters that capture system composition and kinetic properties and predict the outcome of microtubule network organization. These results elucidate a fundamental design principle of spindle bipolarity and establish general rules for active filament network organization.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Molecular Dynamics Simulation , Spindle Apparatus/metabolism , Animals , Humans , Kinesins/chemistry , Microtubules/chemistry , Sf9 Cells , Spindle Apparatus/chemistry , Spodoptera
4.
Cell ; 173(4): 839-850.e18, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628142

ABSTRACT

Maize abnormal chromosome 10 (Ab10) encodes a classic example of true meiotic drive that converts heterochromatic regions called knobs into motile neocentromeres that are preferentially transmitted to egg cells. Here, we identify a cluster of eight genes on Ab10, called the Kinesin driver (Kindr) complex, that are required for both neocentromere motility and preferential transmission. Two meiotic drive mutants that lack neocentromere activity proved to be kindr epimutants with increased DNA methylation across the entire gene cluster. RNAi of Kindr induced a third epimutant and corresponding loss of meiotic drive. Kinesin gliding assays and immunolocalization revealed that KINDR is a functional minus-end-directed kinesin that localizes specifically to knobs containing 180 bp repeats. Sequence comparisons suggest that Kindr diverged from a Kinesin-14A ancestor ∼12 mya and has driven the accumulation of > 500 Mb of knob repeats and affected the segregation of thousands of genes linked to knobs on all 10 chromosomes.


Subject(s)
Centromere/metabolism , Kinesins/metabolism , Meiosis , Plant Proteins/metabolism , Zea mays/metabolism , Centromere/genetics , Chromosomes, Plant , Evolution, Molecular , Haplotypes , In Situ Hybridization, Fluorescence , Kinesins/antagonists & inhibitors , Kinesins/classification , Kinesins/genetics , Models, Genetic , Mutagenesis , Phylogeny , Plant Proteins/antagonists & inhibitors , Plant Proteins/classification , Plant Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Whole Genome Sequencing , Zea mays/genetics
5.
Annu Rev Biochem ; 85: 659-83, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145846

ABSTRACT

Life depends on cell proliferation and the accurate segregation of chromosomes, which are mediated by the microtubule (MT)-based mitotic spindle and ∼200 essential MT-associated proteins. Yet, a mechanistic understanding of how the mitotic spindle is assembled and achieves chromosome segregation is still missing. This is mostly due to the density of MTs in the spindle, which presumably precludes their direct observation. Recent insight has been gained into the molecular building plan of the metaphase spindle using bulk and single-molecule measurements combined with computational modeling. MT nucleation was uncovered as a key principle of spindle assembly, and mechanistic details about MT nucleation pathways and their coordination are starting to be revealed. Lastly, advances in studying spindle assembly can be applied to address the molecular mechanisms of how the spindle segregates chromosomes.


Subject(s)
Centrosome/metabolism , Kinetochores/metabolism , Metaphase , Microtubules/metabolism , Spindle Apparatus/metabolism , Animals , Centrosome/ultrastructure , Chromosome Segregation , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Gene Expression Regulation , Humans , Kinesins/genetics , Kinesins/metabolism , Kinetochores/ultrastructure , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/ultrastructure , Signal Transduction , Spindle Apparatus/ultrastructure , Tubulin/genetics , Tubulin/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zygote/cytology , Zygote/metabolism
6.
Cell ; 165(2): 396-409, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27020753

ABSTRACT

Multiple division cycles without growth are a characteristic feature of early embryogenesis. The female germline loads proteins and RNAs into oocytes to support these divisions, which lack many quality control mechanisms operating in somatic cells undergoing growth. Here, we describe a small RNA-Argonaute pathway that ensures early embryonic divisions in C. elegans by employing catalytic slicing activity to broadly tune, instead of silence, germline gene expression. Misregulation of one target, a kinesin-13 microtubule depolymerase, underlies a major phenotype associated with pathway loss. Tuning of target transcript levels is guided by the density of homologous small RNAs, whose generation must ultimately be related to target sequence. Thus, the tuning action of a small RNA-catalytic Argonaute pathway generates oocytes capable of supporting embryogenesis. We speculate that the specialized nature of germline chromatin led to the emergence of small RNA-catalytic Argonaute pathways in the female germline as a post-transcriptional control layer to optimize oocyte composition.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Embryo, Nonmammalian/metabolism , Metabolic Networks and Pathways , Oocytes/metabolism , Animals , Argonaute Proteins/metabolism , Base Sequence , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/metabolism , Cell Division , Embryo, Nonmammalian/cytology , Embryonic Development , Female , Kinesins/metabolism , Microtubules/metabolism , Molecular Sequence Data , RNA Processing, Post-Transcriptional
7.
Cell ; 167(2): 539-552.e14, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27716509

ABSTRACT

Microtubule-organizing centers (MTOCs) nucleate microtubules that can grow autonomously in any direction. To generate bundles of parallel microtubules originating from a single MTOC, the growth of multiple microtubules needs to coordinated, but the underlying mechanism is unknown. Here, we show that a conserved two-component system consisting of the plus-end tracker EB1 and the minus-end-directed molecular motor Kinesin-14 is sufficient to promote parallel microtubule growth. The underlying mechanism relies on the ability of Kinesin-14 to guide growing plus ends along existing microtubules. The generality of this finding is supported by yeast, Drosophila, and human EB1/Kinesin-14 pairs. We demonstrate that plus-end guiding involves a directional switch of the motor due to a force applied via a growing microtubule end. The described mechanism can account for the generation of parallel microtubule networks required for a broad range of cellular functions such as spindle assembly or cell polarization.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , Kinesins/metabolism , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Motor Proteins/metabolism , Oncogene Proteins/metabolism , RNA Helicases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Drosophila melanogaster , Humans , Mechanical Phenomena
8.
Genes Dev ; 37(5-6): 191-203, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859340

ABSTRACT

Subcellular localization of messenger RNA (mRNA) is a widespread phenomenon that can impact the regulation and function of the encoded protein. In nonneuronal cells, specific mRNAs localize to cell protrusions, and proper mRNA localization is required for cell migration. However, the mechanisms by which mRNA localization regulates protein function in this setting remain unclear. Here, we examined the functional consequences of localization of the mRNA encoding KIF1C. KIF1C is a kinesin motor protein required for cell migration and mRNA trafficking, including trafficking of its own mRNA. We show that Kif1c mRNA localization does not regulate KIF1C's protein abundance, distribution, or ability to traffic other mRNAs. Conversely, Kif1c mRNA localization to protrusions is required for directed cell migration. We used mass spectrometry to identify binding partners of endogenous KIF1C, which revealed dramatic dysregulation of the number and identity of KIF1C interactors in response to Kif1c mRNA mislocalization. These results therefore uncovered a mechanistic connection between mRNA localization to cell protrusions and the specificity of protein-protein interactions. We anticipate that this mechanism is not limited to Kif1c and is likely to be a general principle that impacts the functions of proteins encoded by protrusion-enriched mRNAs in nonneuronal cells.


Subject(s)
Kinesins , Proteins , RNA, Messenger/metabolism , Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism , Dyneins/metabolism , Cell Movement/genetics
9.
Genes Dev ; 37(5-6): 137-139, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36889919

ABSTRACT

Distinct subcellular localizations of mRNAs have been described across a wide variety of cell types. While common themes emerge for neuronal cells, functional roles of mRNA localization in space and time are much less understood in nonneuronal cells. Emerging areas of interest are cell models with protrusions, often linked with cell mobility in cancer systems. In this issue of Genes & Development, Norris and Mendell (pp. 191-203) systematically investigate a link between mRNA localization to cell protrusions in a mouse melanoma cell system and a mechanistic link to downstream consequences for cell mobility. The study first identifies a model mRNA of interest in an unbiased way that exhibits a set of phenotypes associated with cell mobility. The candidate mRNA that fulfills all requirements is Kif1c mRNA. Further systematic investigation links Kif1c mRNA localization to assembly of a protein-protein network on the KIF1C protein itself. What's clear is that this work will inspire a further mechanistic dissection of the Kif1c mRNA/KIF1C protein interplay in this important nonneuronal model cell system. More broadly, this work suggests that a broad set of model mRNAs should be investigated to understand mRNA dynamics and downstream functional consequences across a variety of cell models.


Subject(s)
Kinesins , Proteins , Mice , Animals , Kinesins/genetics , Kinesins/metabolism , Protein Binding , RNA, Messenger/metabolism , Proteins/metabolism , Cell Movement/genetics
10.
Cell ; 160(6): 1159-68, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25748652

ABSTRACT

Cytoskeletal remodeling is essential to eukaryotic cell division and morphogenesis. The mechanical forces driving the restructuring are attributed to the action of molecular motors and the dynamics of cytoskeletal filaments, which both consume chemical energy. By contrast, non-enzymatic filament crosslinkers are regarded as mere friction-generating entities. Here, we experimentally demonstrate that diffusible microtubule crosslinkers of the Ase1/PRC1/Map65 family generate directed microtubule sliding when confined between partially overlapping microtubules. The Ase1-generated forces, directly measured by optical tweezers to be in the piconewton-range, were sufficient to antagonize motor-protein driven microtubule sliding. Force generation is quantitatively explained by the entropic expansion of confined Ase1 molecules diffusing within the microtubule overlaps. The thermal motion of crosslinkers is thus harnessed to generate mechanical work analogous to compressed gas propelling a piston in a cylinder. As confinement of diffusible proteins is ubiquitous in cells, the associated entropic forces are likely of importance for cellular mechanics beyond cytoskeletal networks.


Subject(s)
Microtubules/metabolism , Animals , Biomechanical Phenomena , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Friction , Green Fluorescent Proteins/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Optical Tweezers , Schizosaccharomyces pombe Proteins/metabolism
11.
EMBO J ; 43(7): 1244-1256, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38424239

ABSTRACT

During mitosis, motor proteins and microtubule-associated protein organize the spindle apparatus by cross-linking and sliding microtubules. Kinesin-5 plays a vital role in spindle formation and maintenance, potentially inducing twist in the spindle fibers. The off-axis power stroke of kinesin-5 could generate this twist, but its implications in microtubule organization remain unclear. Here, we investigate 3D microtubule-microtubule sliding mediated by the human kinesin-5, KIF11, and found that the motor caused right-handed helical motion of anti-parallel microtubules around each other. The sidestepping ratio increased with reduced ATP concentration, indicating that forward and sideways stepping of the motor are not strictly coupled. Further, the microtubule-microtubule distance (motor extension) during sliding decreased with increasing sliding velocity. Intriguingly, parallel microtubules cross-linked by KIF11 orbited without forward motion, with nearly full motor extension. Altering the length of the neck linker increased the forward velocity and pitch of microtubules in anti-parallel overlaps. Taken together, we suggest that helical motion and orbiting of microtubules, driven by KIF11, contributes to flexible and context-dependent filament organization, as well as torque regulation within the mitotic spindle.


Subject(s)
Kinesins , Microtubules , Humans , Kinesins/metabolism , Microtubules/metabolism , Spindle Apparatus/physiology , Microtubule-Associated Proteins/metabolism , Mitosis
12.
EMBO J ; 43(13): 2715-2732, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769437

ABSTRACT

Microtubules regulate cell polarity and migration via local activation of focal adhesion turnover, but the mechanism of this process is insufficiently understood. Molecular complexes containing KANK family proteins connect microtubules with talin, the major component of focal adhesions. Here, local optogenetic activation of KANK1-mediated microtubule/talin linkage promoted microtubule targeting to an individual focal adhesion and subsequent withdrawal, resulting in focal adhesion centripetal sliding and rapid disassembly. This sliding is preceded by a local increase of traction force due to accumulation of myosin-II and actin in the proximity of the focal adhesion. Knockdown of the Rho activator GEF-H1 prevented development of traction force and abolished sliding and disassembly of focal adhesions upon KANK1 activation. Other players participating in microtubule-driven, KANK-dependent focal adhesion disassembly include kinases ROCK, PAK, and FAK, as well as microtubules/focal adhesion-associated proteins kinesin-1, APC, and αTAT. Based on these data, we develop a mathematical model for a microtubule-driven focal adhesion disruption involving local GEF-H1/RhoA/ROCK-dependent activation of contractility, which is consistent with experimental data.


Subject(s)
Focal Adhesions , Kinesins , Microtubules , Rho Guanine Nucleotide Exchange Factors , Focal Adhesions/metabolism , Microtubules/metabolism , Humans , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Kinesins/metabolism , Kinesins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Myosin Type II/metabolism , Talin/metabolism , Talin/genetics , Animals
13.
EMBO J ; 43(13): 2606-2635, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806659

ABSTRACT

Microtubule-based kinesin motor proteins are crucial for intracellular transport, but their hyperactivation can be detrimental for cellular functions. This study investigated the impact of a constitutively active ciliary kinesin mutant, OSM-3CA, on sensory cilia in C. elegans. Surprisingly, we found that OSM-3CA was absent from cilia but underwent disposal through membrane abscission at the tips of aberrant neurites. Neighboring glial cells engulf and eliminate the released OSM-3CA, a process that depends on the engulfment receptor CED-1. Through genetic suppressor screens, we identified intragenic mutations in the OSM-3CA motor domain and mutations inhibiting the ciliary kinase DYF-5, both of which restored normal cilia in OSM-3CA-expressing animals. We showed that conformational changes in OSM-3CA prevent its entry into cilia, and OSM-3CA disposal requires its hyperactivity. Finally, we provide evidence that neurons also dispose of hyperactive kinesin-1 resulting from a clinic variant associated with amyotrophic lateral sclerosis, suggesting a widespread mechanism for regulating hyperactive kinesins.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cilia , Kinesins , Neuroglia , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Kinesins/metabolism , Kinesins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Neuroglia/metabolism , Cilia/metabolism , Neurons/metabolism , Mutation , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology
14.
Cell ; 154(2): 377-90, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870126

ABSTRACT

Error-free cell division depends on the assembly of the spindle midzone, a specialized array of overlapping microtubules that emerges between segregating chromosomes during anaphase. The molecular mechanisms by which a subset of dynamic microtubules from the metaphase spindle are selected and organized into a stable midzone array are poorly understood. Here, we show using in vitro reconstitution assays that PRC1 and kinesin-4, two microtubule-associated proteins required for midzone assembly, can tag microtubule plus ends. Remarkably, the size of these tags is proportional to filament length. We determine the crystal structure of the PRC1 homodimer and map the protein-protein interactions needed for tagging microtubule ends. Importantly, length-dependent microtubule plus-end-tagging by PRC1 is also observed in dividing cells. Our findings suggest how biochemically similar microtubules can be differentially marked, based on length, for selective regulation during the formation of specialized arrays, such as those required for cytokinesis.


Subject(s)
Cell Cycle Proteins/metabolism , Microtubules/metabolism , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Cytokinesis , Dimerization , Humans , Kinesins/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment
15.
Genes Dev ; 34(17-18): 1110-1112, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873577

ABSTRACT

Maize heterochromatic knobs cheat female meiosis by forming neocentromeres that bias their segregation into the future egg cell. In this issue of Genes & Development, Swentowsky and colleagues (pp. 1239-1251) show that two types of knobs, those composed of 180-bp and TR1 sequences, recruit their own novel and divergent kinesin-14 family members to form neocentromeres.


Subject(s)
Genome, Plant , Zea mays/genetics , Centromere/genetics , Genome, Plant/genetics , Kinesins/genetics , Kinesins/metabolism , Meiosis/genetics
16.
Genes Dev ; 34(17-18): 1239-1251, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32820038

ABSTRACT

A maize chromosome variant called abnormal chromosome 10 (Ab10) converts knobs on chromosome arms into neocentromeres, causing their preferential segregation to egg cells in a process known as meiotic drive. We previously demonstrated that the gene Kinesin driver (Kindr) on Ab10 encodes a kinesin-14 required to mobilize neocentromeres made up of the major tandem repeat knob180. Here we describe a second kinesin-14 gene, TR-1 kinesin (Trkin), that is required to mobilize neocentromeres made up of the minor tandem repeat TR-1. Trkin lies in a 4-Mb region of Ab10 that is not syntenic with any other region of the maize genome and shows extraordinary sequence divergence from Kindr and other kinesins in plants. Despite its unusual structure, Trkin encodes a functional minus end-directed kinesin that specifically colocalizes with TR-1 in meiosis, forming long drawn out neocentromeres. TRKIN contains a nuclear localization signal and localizes to knobs earlier in prophase than KINDR. The fact that TR-1 repeats often co-occur with knob180 repeats suggests that the current role of the TRKIN/TR-1 system is to facilitate the meiotic drive of the KINDR/knob180 system.


Subject(s)
Centromere/genetics , Centromere/metabolism , Kinesins/genetics , Kinesins/metabolism , Zea mays/genetics , Zea mays/metabolism , Chromosomes, Plant/genetics , Genes, Plant/genetics , Meiosis , Models, Genetic , Protein Transport/genetics
17.
EMBO J ; 42(21): e113647, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37592895

ABSTRACT

During mitosis, spindle architecture alters as chromosomes segregate into daughter cells. The microtubule crosslinker protein regulator of cytokinesis 1 (PRC1) is essential for spindle stability, chromosome segregation and completion of cytokinesis, but how it recruits motors to the central spindle to coordinate the segregation of chromosomes is unknown. Here, we combine structural and cell biology approaches to show that the human CENP-E motor, which is essential for chromosome capture and alignment by microtubules, binds to PRC1 through a conserved hydrophobic motif. This binding mechanism is also used by Kinesin-4 Kif4A:PRC1. Using in vitro reconstitution, we demonstrate that CENP-E slides antiparallel PRC1-crosslinked microtubules. We find that the regulation of CENP-E -PRC1 interaction is spatially and temporally coupled with relocalization to overlapping microtubules in anaphase. Finally, we demonstrate that the PRC1-microtubule motor interaction is essential in anaphase to control chromosome partitioning, retain central spindle integrity and ensure cytokinesis. Taken together our findings reveal the molecular basis for the cell cycle regulation of motor-PRC1 complexes to couple chromosome segregation and cytokinesis.


Subject(s)
Cytokinesis , Kinesins , Humans , Cytokinesis/physiology , Kinesins/genetics , Kinesins/metabolism , Phosphorylation , Spindle Apparatus/metabolism , Mitosis , Cell Cycle Proteins/metabolism , Microtubules/metabolism
18.
EMBO J ; 42(16): e112812, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37403793

ABSTRACT

Intracellular organelle organization is conserved in eukaryotic cells and is primarily achieved through active transport by motor proteins along the microtubule cytoskeleton. Microtubule post-translational modifications (PTMs) can contribute to microtubule diversity and differentially regulate motor-mediated transport. Here, we show that centrosome amplification, commonly observed in cancer and shown to promote aneuploidy and invasion, induces a global change in organelle positioning towards the cell periphery and facilitates nuclear migration through confined spaces. This reorganization requires kinesin-1 and is analogous to the loss of dynein. Cells with amplified centrosomes display increased levels of acetylated tubulin, a PTM that could enhance kinesin-1-mediated transport. Depletion of α-tubulin acetyltransferase 1 (αTAT1) to block tubulin acetylation rescues the displacement of centrosomes, mitochondria, and vimentin but not Golgi or endosomes. Analyses of the distribution of total and acetylated microtubules indicate that the polarized distribution of modified microtubules, rather than levels alone, plays an important role in the positioning of specific organelles, such as the centrosome. We propose that increased tubulin acetylation differentially impacts kinesin-1-mediated organelle displacement to regulate intracellular organization.


Subject(s)
Kinesins , Tubulin , Tubulin/metabolism , Kinesins/genetics , Kinesins/metabolism , Acetylation , Microtubules/metabolism , Centrosome/metabolism , Dyneins/metabolism , Protein Processing, Post-Translational
19.
EMBO J ; 42(5): e112101, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36636822

ABSTRACT

Tubulin posttranslational modifications have been predicted to control cytoskeletal functions by coordinating the molecular interactions between microtubules and their associating proteins. A prominent tubulin modification in neurons is polyglutamylation, the deregulation of which causes neurodegeneration. Yet, the underlying molecular mechanisms have remained elusive. Here, using in-vitro reconstitution, we determine how polyglutamylation generated by the two predominant neuronal polyglutamylases, TTLL1 and TTLL7, specifically modulates the activities of three major microtubule interactors: the microtubule-associated protein Tau, the microtubule-severing enzyme katanin and the molecular motor kinesin-1. We demonstrate that the unique modification patterns generated by TTLL1 and TTLL7 differentially impact those three effector proteins, thus allowing for their selective regulation. Given that our experiments were performed with brain tubulin from mouse models in which physiological levels and patterns of polyglutamylation were altered by the genetic knockout of the main modifying enzymes, our quantitative measurements provide direct mechanistic insight into how polyglutamylation could selectively control microtubule interactions in neurons.


Subject(s)
Tubulin , Animals , Mice , Cytoskeleton/metabolism , Kinesins/metabolism , Microtubules/metabolism , Tubulin/metabolism , Peptide Synthases , Microtubule-Associated Proteins
20.
EMBO J ; 42(11): e112953, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37014312

ABSTRACT

Microtubule (MT) minus ends are stabilized by CAMSAP family proteins at noncentrosomal MT-organizing centers. Despite progress in identifying diverse positive regulators, knowledge on the negative regulation of the MT minus-end distribution is lacking. Here, we identify CEP170B as a MT minus-end-binding protein that colocalizes with the microtubule-stabilizing complex at the cortical patches. CEP170B depends on the scaffold protein liprin-α1 for its cortical targeting and requires liprin-α1-bound PP2A phosphatase for its MT localization. CEP170B excludes CAMSAPs-stabilized MT minus ends from the cell periphery in HeLa cells and the basal cortex in human epithelial cells and is required for directional vesicle trafficking and cyst formation in 3D culture. Reconstitution experiments demonstrate that CEP170B autonomously tracks growing MT minus ends and blocks minus-end growth. Furthermore, CEP170B in a complex with the kinesin KIF2A acts as a potent MT minus-end depolymerase capable of antagonizing the stabilizing effect of CAMSAPs. Our study uncovers an antagonistic mechanism for controlling the spatial distribution of MT minus ends, which contributes to the establishment of polarized MT network and cell polarity.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , HeLa Cells , Microtubules/metabolism , Cytoskeletal Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL