Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.300
Filter
Add more filters

Publication year range
1.
Cell ; 186(21): 4694-4709.e16, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37832525

ABSTRACT

Cytoplasmic divisions are thought to rely on nuclear divisions and mitotic signals. We demonstrate in Drosophila embryos that cytoplasm can divide repeatedly without nuclei and mitotic CDK/cyclin complexes. Cdk1 normally slows an otherwise faster cytoplasmic division cycle, coupling it with nuclear divisions, and when uncoupled, cytoplasm starts dividing before mitosis. In developing embryos where CDK/cyclin activity can license mitotic microtubule (MT) organizers like the spindle, cytoplasmic divisions can occur without the centrosome, a principal organizer of interphase MTs. However, centrosomes become essential in the absence of CDK/cyclin activity, implying that the cytoplasm can employ either the centrosome-based interphase or CDK/cyclin-dependent mitotic MTs to facilitate its divisions. Finally, we present evidence that autonomous cytoplasmic divisions occur during unperturbed fly embryogenesis and that they may help extrude mitotically stalled nuclei during blastoderm formation. We postulate that cytoplasmic divisions occur in cycles governed by a yet-to-be-uncovered clock mechanism autonomous from CDK/cyclin complexes.


Subject(s)
Cytokinesis , Embryo, Nonmammalian , Animals , Cell Nucleus , Centrosome , Cyclins/metabolism , Drosophila , Mitosis , Spindle Apparatus/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism
2.
Cell ; 184(23): 5791-5806.e19, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34715025

ABSTRACT

Dynein-decorated doublet microtubules (DMTs) are critical components of the oscillatory molecular machine of cilia, the axoneme, and have luminal surfaces patterned periodically by microtubule inner proteins (MIPs). Here we present an atomic model of the 48-nm repeat of a mammalian DMT, derived from a cryoelectron microscopy (cryo-EM) map of the complex isolated from bovine respiratory cilia. The structure uncovers principles of doublet microtubule organization and features specific to vertebrate cilia, including previously unknown MIPs, a luminal bundle of tektin filaments, and a pentameric dynein-docking complex. We identify a mechanism for bridging 48- to 24-nm periodicity across the microtubule wall and show that loss of the proteins involved causes defective ciliary motility and laterality abnormalities in zebrafish and mice. Our structure identifies candidate genes for diagnosis of ciliopathies and provides a framework to understand their functions in driving ciliary motility.


Subject(s)
Cilia/ultrastructure , Cryoelectron Microscopy , Mammals/metabolism , Proteins/metabolism , Proteins/ultrastructure , Amino Acid Sequence , Animals , Cattle , Cilia/metabolism , Dyneins/metabolism , Embryo, Mammalian/metabolism , Female , Male , Mice, Inbred C57BL , Microtubule Proteins/chemistry , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Mutation/genetics , Trachea/anatomy & histology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Cell ; 184(11): 2860-2877.e22, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33964210

ABSTRACT

Most human embryos are aneuploid. Aneuploidy frequently arises during the early mitotic divisions of the embryo, but its origin remains elusive. Human zygotes that cluster their nucleoli at the pronuclear interface are thought to be more likely to develop into healthy euploid embryos. Here, we show that the parental genomes cluster with nucleoli in each pronucleus within human and bovine zygotes, and clustering is required for the reliable unification of the parental genomes after fertilization. During migration of intact pronuclei, the parental genomes polarize toward each other in a process driven by centrosomes, dynein, microtubules, and nuclear pore complexes. The maternal and paternal chromosomes eventually cluster at the pronuclear interface, in direct proximity to each other, yet separated. Parental genome clustering ensures the rapid unification of the parental genomes on nuclear envelope breakdown. However, clustering often fails, leading to chromosome segregation errors and micronuclei, incompatible with healthy embryo development.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Development/genetics , Aneuploidy , Animals , Cattle , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Centrosome/metabolism , Chromosome Segregation/physiology , Chromosomes/metabolism , Fertilization/genetics , Humans , Male , Microtubules/metabolism , Mitosis , Oocytes/metabolism , Spermatozoa/metabolism , Zygote/metabolism
4.
Cell ; 180(1): 165-175.e16, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31862189

ABSTRACT

The γ-tubulin ring complex (γ-TuRC) is an essential regulator of centrosomal and acentrosomal microtubule formation, yet its structure is not known. Here, we present a cryo-EM reconstruction of the native human γ-TuRC at ∼3.8 Å resolution, revealing an asymmetric, cone-shaped structure. Pseudo-atomic models indicate that GCP4, GCP5, and GCP6 form distinct Y-shaped assemblies that structurally mimic GCP2/GCP3 subcomplexes distal to the γ-TuRC "seam." We also identify an unanticipated structural bridge that includes an actin-like protein and spans the γ-TuRC lumen. Despite its asymmetric architecture, the γ-TuRC arranges γ-tubulins into a helical geometry poised to nucleate microtubules. Diversity in the γ-TuRC subunits introduces large (>100,000 Å2) surfaces in the complex that allow for interactions with different regulatory factors. The observed compositional complexity of the γ-TuRC could self-regulate its assembly into a cone-shaped structure to control microtubule formation across diverse contexts, e.g., within biological condensates or alongside existing filaments.


Subject(s)
Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/ultrastructure , Tubulin/ultrastructure , Actins/metabolism , Cryoelectron Microscopy/methods , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/ultrastructure , Microtubules/metabolism , Tubulin/metabolism
5.
Annu Rev Cell Dev Biol ; 37: 23-41, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34186005

ABSTRACT

The purpose of this review is to explore self-organizing mechanisms that pattern microtubules (MTs) and spatially organize animal cell cytoplasm, inspired by recent experiments in frog egg extract. We start by reviewing conceptual distinctions between self-organizing and templating mechanisms for subcellular organization. We then discuss self-organizing mechanisms that generate radial MT arrays and cell centers in the absence of centrosomes. These include autocatalytic MT nucleation, transport of minus ends, and nucleation from organelles such as melanosomes and Golgi vesicles that are also dynein cargoes. We then discuss mechanisms that partition the cytoplasm in syncytia, in which multiple nuclei share a common cytoplasm, starting with cytokinesis, when all metazoan cells are transiently syncytial. The cytoplasm of frog eggs is partitioned prior to cytokinesis by two self-organizing modules, protein regulator of cytokinesis 1 (PRC1)-kinesin family member 4A (KIF4A) and chromosome passenger complex (CPC)-KIF20A. Similar modules may partition longer-lasting syncytia, such as early Drosophila embryos. We end by discussing shared mechanisms and principles for the MT-based self-organization of cellular units.


Subject(s)
Centrosome , Microtubules , Animals , Centrosome/metabolism , Cytokinesis , Cytoskeleton , Golgi Apparatus , Microtubules/metabolism
6.
Cell ; 173(3): 776-791.e17, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29576449

ABSTRACT

Transformation from morula to blastocyst is a defining event of preimplantation embryo development. During this transition, the embryo must establish a paracellular permeability barrier to enable expansion of the blastocyst cavity. Here, using live imaging of mouse embryos, we reveal an actin-zippering mechanism driving this embryo sealing. Preceding blastocyst stage, a cortical F-actin ring assembles at the apical pole of the embryo's outer cells. The ring structure forms when cortical actin flows encounter a network of polar microtubules that exclude F-actin. Unlike stereotypical actin rings, the actin rings of the mouse embryo are not contractile, but instead, they expand to the cell-cell junctions. Here, they couple to the junctions by recruiting and stabilizing adherens and tight junction components. Coupling of the actin rings triggers localized myosin II accumulation, and it initiates a tension-dependent zippering mechanism along the junctions that is required to seal the embryo for blastocyst formation.


Subject(s)
Actins/chemistry , Blastocyst/metabolism , Microtubules/metabolism , Myosin Type II/chemistry , Animals , Cell Communication , Cytoskeletal Proteins/chemistry , Embryo, Mammalian , Embryonic Development , Female , Green Fluorescent Proteins , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Morula , RNA, Small Interfering/metabolism , Tight Junctions
7.
Annu Rev Cell Dev Biol ; 35: 29-54, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31394046

ABSTRACT

Microtubules are core components of the cytoskeleton and serve as tracks for motor protein-based intracellular transport. Microtubule networks are highly diverse across different cell types and are believed to adapt to cell type-specific transport demands. Here we review how the spatial organization of different subsets of microtubules into higher-order networks determines the traffic rules for motor-based transport in different animal cell types. We describe the interplay between microtubule network organization and motor-based transport within epithelial cells, oocytes, neurons, cilia, and the spindle apparatus.


Subject(s)
Microtubules/metabolism , Animals , Cell Polarity , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/chemistry , Molecular Motor Proteins/metabolism , Protein Transport , Spindle Apparatus/chemistry , Spindle Apparatus/metabolism
8.
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
9.
Annu Rev Cell Dev Biol ; 33: 1-22, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28992437

ABSTRACT

During my graduate work with Keith Porter, I became fascinated by the mitotic spindle, an interest that has motivated much of my scientific work ever since. I began spindle studies by using electron microscopes, instruments that have made significant contributions to our understanding of spindle organization. Such instruments have helped to elucidate the distributions of spindle microtubules, the interactions among them, their molecular polarity, and their associations with both kinetochores and spindle poles. Our lab has also investigated some processes of spindle physiology: microtubule dynamics, the actions of microtubule-associated proteins (including motor enzymes), the character of forces generated by specific spindle components, and factors that control mitotic progression. Here, I give a personal perspective on some of this intellectual history and on what recent discoveries imply about the mechanisms of chromosome motion.


Subject(s)
Chromosomes/metabolism , Enzymes/metabolism , Microtubules/metabolism , Mitosis , Molecular Motor Proteins/metabolism , Motion , Animals , Chromosomes/ultrastructure , Humans , Microtubules/ultrastructure
10.
Mol Cell ; 83(13): 2290-2302.e13, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37295431

ABSTRACT

Microtubules play crucial roles in cellular architecture, intracellular transport, and mitosis. The availability of free tubulin subunits affects polymerization dynamics and microtubule function. When cells sense excess free tubulin, they trigger degradation of the encoding mRNAs, which requires recognition of the nascent polypeptide by the tubulin-specific ribosome-binding factor TTC5. How TTC5 initiates the decay of tubulin mRNAs is unknown. Here, our biochemical and structural analysis reveals that TTC5 recruits the poorly studied protein SCAPER to the ribosome. SCAPER, in turn, engages the CCR4-NOT deadenylase complex through its CNOT11 subunit to trigger tubulin mRNA decay. SCAPER mutants that cause intellectual disability and retinitis pigmentosa in humans are impaired in CCR4-NOT recruitment, tubulin mRNA degradation, and microtubule-dependent chromosome segregation. Our findings demonstrate how recognition of a nascent polypeptide on the ribosome is physically linked to mRNA decay factors via a relay of protein-protein interactions, providing a paradigm for specificity in cytoplasmic gene regulation.


Subject(s)
Ribosomes , Tubulin , Humans , Tubulin/genetics , Tubulin/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Microtubules/metabolism , Homeostasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Stability , Carrier Proteins/metabolism , Transcription Factors/metabolism
11.
Annu Rev Cell Dev Biol ; 32: 173-195, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27362645

ABSTRACT

Objects are commonly moved within the cell by either passive diffusion or active directed transport. A third possibility is advection, in which objects within the cytoplasm are moved with the flow of the cytoplasm. Bulk movement of the cytoplasm, or streaming, as required for advection, is more common in large cells than in small cells. For example, streaming is observed in elongated plant cells and the oocytes of several species. In the Drosophila oocyte, two stages of streaming are observed: relatively slow streaming during mid-oogenesis and streaming that is approximately ten times faster during late oogenesis. These flows are implicated in two processes: polarity establishment and mixing. In this review, I discuss the underlying mechanism of streaming, how slow and fast streaming are differentiated, and what we know about the physiological roles of the two types of streaming.


Subject(s)
Cytoplasmic Streaming , Drosophila/cytology , Oocytes/cytology , Animals , Cell Polarity , Oogenesis
12.
Mol Cell ; 82(1): 75-89.e9, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34942120

ABSTRACT

Circular RNAs (circRNAs) are widely expressed in eukaryotes and are regulated in many biological processes. Although several studies indicate their activity as microRNA (miRNA) and protein sponges, little is known about their ability to directly control mRNA homeostasis. We show that the widely expressed circZNF609 directly interacts with several mRNAs and increases their stability and/or translation by favoring the recruitment of the RNA-binding protein ELAVL1. Particularly, the interaction with CKAP5 mRNA, which interestingly overlaps the back-splicing junction, enhances CKAP5 translation, regulating microtubule function in cancer cells and sustaining cell-cycle progression. Finally, we show that circZNF609 downregulation increases the sensitivity of several cancer cell lines to different microtubule-targeting chemotherapeutic drugs and that locked nucleic acid (LNA) protectors against the pairing region on circZNF609 phenocopy such effects. These data set an example of how the small effects tuned by circZNF609/CKAP5 mRNA interaction might have a potent output in tumor growth and drug response.


Subject(s)
Carcinogenesis , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neoplasms/metabolism , RNA, Circular/metabolism , RNA, Messenger/metabolism , Animals , Antineoplastic Agents/pharmacology , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Male , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubules/drug effects , Microtubules/genetics , Microtubules/pathology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA, Circular/genetics , RNA, Messenger/genetics , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
13.
Trends Biochem Sci ; 49(4): 318-332, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38350804

ABSTRACT

To fulfill their actual cellular role, individual microtubules become functionally specialized through a broad range of mechanisms. The 'search and capture' model posits that microtubule dynamics and functions are specified by cellular targets that they capture (i.e., a posteriori), independently of the microtubule-organizing center (MTOC) they emerge from. However, work in budding yeast indicates that MTOCs may impart a functional identity to the microtubules they nucleate, a priori. Key effectors in this process are microtubule plus-end tracking proteins (+TIPs), which track microtubule tips to regulate their dynamics and facilitate their targeted interactions. In this review, we discuss potential mechanisms of a priori microtubule specialization, focusing on recent findings indicating that +TIP networks may undergo liquid biomolecular condensation in different cell types.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism
14.
EMBO J ; 43(13): 2733-2758, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38831122

ABSTRACT

Organ morphogenesis depends on mechanical interactions between cells and tissues. These interactions generate forces that can be sensed by cells and affect key cellular processes. However, how mechanical forces, together with biochemical signals, contribute to the shaping of complex organs is still largely unclear. We address this question using the seed of Arabidopsis as a model system. We show that seeds first experience a phase of rapid anisotropic growth that is dependent on the response of cortical microtubule (CMT) to forces, which guide cellulose deposition according to shape-driven stresses in the outermost layer of the seed coat. However, at later stages of development, we show that seed growth is isotropic and depends on the properties of an inner layer of the seed coat that stiffens its walls in response to tension but has isotropic material properties. Finally, we show that the transition from anisotropic to isotropic growth is due to the dampening of cortical microtubule responses to shape-driven stresses. Altogether, our work supports a model in which spatiotemporally distinct mechanical responses control the shape of developing seeds in Arabidopsis.


Subject(s)
Arabidopsis , Microtubules , Seeds , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis/genetics , Seeds/growth & development , Seeds/metabolism , Microtubules/metabolism , Biomechanical Phenomena , Stress, Mechanical , Anisotropy , Cellulose/metabolism
15.
EMBO J ; 43(7): 1214-1243, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38388748

ABSTRACT

Regulation of directed axon guidance and branching during development is essential for the generation of neuronal networks. However, the molecular mechanisms that underlie interstitial (or collateral) axon branching in the mammalian brain remain unresolved. Here, we investigate interstitial axon branching in vivo using an approach for precise labeling of layer 2/3 callosal projection neurons (CPNs). This method allows for quantitative analysis of axonal morphology at high acuity and also manipulation of gene expression in well-defined temporal windows. We find that the GSK3ß serine/threonine kinase promotes interstitial axon branching in layer 2/3 CPNs by releasing MAP1B-mediated inhibition of axon branching. Further, we find that the tubulin tyrosination cycle is a key downstream component of GSK3ß/MAP1B signaling. These data suggest a cell-autonomous molecular regulation of cortical neuron axon morphology, in which GSK3ß can release a MAP1B-mediated brake on interstitial axon branching upstream of the posttranslational tubulin code.


Subject(s)
Carrier Proteins , Tubulin , Animals , Tubulin/metabolism , Carrier Proteins/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Microtubules/metabolism , Axons/metabolism , Cells, Cultured , Mammals
16.
Mol Cell ; 79(1): 191-198.e3, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619469

ABSTRACT

We recently used CRISPRi/a-based chemical-genetic screens and cell biological, biochemical, and structural assays to determine that rigosertib, an anti-cancer agent in phase III clinical trials, kills cancer cells by destabilizing microtubules. Reddy and co-workers (Baker et al., 2020, this issue of Molecular Cell) suggest that a contaminating degradation product in commercial formulations of rigosertib is responsible for the microtubule-destabilizing activity. Here, we demonstrate that cells treated with pharmaceutical-grade rigosertib (>99.9% purity) or commercially obtained rigosertib have qualitatively indistinguishable phenotypes across multiple assays. The two formulations have indistinguishable chemical-genetic interactions with genes that modulate microtubule stability, both destabilize microtubules in cells and in vitro, and expression of a rationally designed tubulin mutant with a mutation in the rigosertib binding site (L240F TUBB) allows cells to proliferate in the presence of either formulation. Importantly, the specificity of the L240F TUBB mutant for microtubule-destabilizing agents has been confirmed independently. Thus, rigosertib kills cancer cells by destabilizing microtubules, in agreement with our original findings.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation , Glycine/analogs & derivatives , Microtubules/drug effects , Neoplasms/pathology , Pharmaceutical Preparations/metabolism , Sulfones/pharmacology , Tubulin/metabolism , Cells, Cultured , Crystallography, X-Ray , Drug Contamination , Glycine/pharmacology , Humans , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Pharmaceutical Preparations/chemistry , Protein Conformation , Tubulin/chemistry , Tubulin/genetics
17.
Trends Biochem Sci ; 48(4): 315-316, 2023 04.
Article in English | MEDLINE | ID: mdl-36754682

ABSTRACT

In a recent study, Chaaban and Carter use cryo-electron microscopy (cryo-EM) and an innovative data-processing pipeline to determine the first high-resolution structure of the dynein-dynactin-BICDR1 complex assembled on microtubules. The structure of the complex reveals novel stoichiometry and provides new mechanistic insight into dynein function and mechanism.


Subject(s)
Dyneins , Microtubule-Associated Proteins , Dyneins/metabolism , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Cryoelectron Microscopy , Microtubules/chemistry , Microtubules/metabolism , Dynactin Complex/analysis , Dynactin Complex/chemistry , Dynactin Complex/metabolism
18.
Trends Genet ; 40(7): 558-559, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749881

ABSTRACT

Twin studies suggest that additive genetic effects account for about a quarter of the variance in handedness. Recently, Schijven et al. used exome-wide sequencing to provide evidence for a role of rare protein-coding variants in handedness. These included the gene encoding beta-tubulin, TUBB4B, suggesting that microtubules are relevant for handedness ontogenesis.


Subject(s)
Functional Laterality , Tubulin , Tubulin/genetics , Humans , Functional Laterality/genetics , Genetic Variation , Microtubules/genetics
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL