1
|
Gomes Paim LM, Bechstedt S. Regulation of microtubule growth rates and their impact on chromosomal instability. Cell Cycle 2025:1-20. [PMID: 40260826 DOI: 10.1080/15384101.2025.2485842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 04/24/2025] Open
Abstract
Microtubules are polymers of α/β tubulin dimers that build the mitotic spindle, which segregates duplicated chromosomes during cell division. Microtubule function is governed by dynamic instability, whereby cycles of growth and shrinkage contribute to the forces necessary for chromosome movement. Regulation of microtubule growth velocity requires cell cycle-dependent changes in expression, localization and activity of microtubule-associated proteins (MAPs) as well as tubulin post-translational modifications that modulate microtubule dynamics. It has become clear that optimal microtubule growth velocities are required for proper chromosome segregation and ploidy maintenance. Suboptimal microtubule growth rates can result from altered activity of MAPs and could lead to aneuploidy, possibly by disrupting the establishment of microtubule bundles at kinetochores and altering the mechanical forces required for sister chromatid segregation. Future work using high-resolution, low-phototoxicity microscopy and novel fluorescent markers will be invaluable in obtaining deeper mechanistic insights into how microtubule processes contribute to chromosome segregation.
Collapse
Affiliation(s)
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
- Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montréal, Canada
| |
Collapse
|
2
|
Uzbekov R, Uzbekova S, Severin F, Prigent C, Arlot-Bonnemains Y. Aurora A Kinase Begins to Localize to the Centrosome in the S-phase of the Cell Cycle in the XL2 Cell Line. FRONT BIOSCI-LANDMRK 2024; 29:317. [PMID: 39344321 DOI: 10.31083/j.fbl2909317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The centrosome is one of the principal cell hubs, where numerous proteins important for intracellular regulatory processes are concentrated. One of them, serine-threonine kinase 6, alias Aurora A, is involved in centrosome duplication and mitotic spindle formation and maintenance. METHODS Long-term vital observations of cells, immunofluorescence analysis of protein localization, synchronization of cells at different phases of the cell cycle, Western blot analysis of protein content were used in the work. RESULTS In this study, we investigated the dynamics of Aurora A protein accumulation and degradation in the XL2 Xenopus cell line during its 28-hour cell cycle. Using Western blot and immunofluorescence analyses, we demonstrated that Aurora A disappeared from the centrosome within one hour following mitosis and was not redistributed to other cell compartments. Using double Aurora A/Bromodeoxyuridine immunofluorescence labeling of the cells with precisely determined cell cycle stages, we observed that Aurora A reappeared in the centrosome during the S-phase, which was earlier than reported for all other known proteins with mitosis-specific centrosomal localization. Moreover, Aurora A accumulation in the centrosomal region and centrosome separation were asynchronous in the sister cells. CONCLUSIONS The reported data allowed us to hypothesize that Aurora A is one of the primary links in coordinating centrosome separation and constructing the mitotic spindle.
Collapse
Affiliation(s)
- Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Electronique, Faculté Médecine, University François Rabelais, 37032 Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119991 Moscow, Russia
| | - Svetlana Uzbekova
- Physiology of Reproduction and Behavior (PRC) mixed Research Unit of National Research Institute for Agriculture, Food and the Environment (INRAE), National Center for Scientific Research (CNRS), French Institute of Horses and Riding (IFCE), University of Tours, 37380 Nouzilly, France
| | - Fedor Severin
- A.N. Belozersky Institute of Physico-chemical Biology, Moscow State University, 119991 Moscow, Russia
| | - Claude Prigent
- University of Montpellier, Centre National de la Recherche Scientifique (CNRS), 34000 Montpellier, France
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), Centre National de la Recherche Scientifique, 34293 Montpellier, France
| | - Yannick Arlot-Bonnemains
- Mixed Research Unit 6290, National Center for Scientific Research (CNRS), Institute of Genetics and Development of Rennes, University of Rennes 1, 35043 Rennes, France
| |
Collapse
|
3
|
Xu K, Li Z, Mao L, Guo Z, Chen Z, Chai Y, Xie C, Yang X, Na J, Li W, Ou G. AlphaFold2-guided engineering of split-GFP technology enables labeling of endogenous tubulins across species while preserving function. PLoS Biol 2024; 22:e3002615. [PMID: 39159282 PMCID: PMC11361732 DOI: 10.1371/journal.pbio.3002615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/29/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024] Open
Abstract
Dynamic properties are essential for microtubule (MT) physiology. Current techniques for in vivo imaging of MTs present intrinsic limitations in elucidating the isotype-specific nuances of tubulins, which contribute to their versatile functions. Harnessing the power of the AlphaFold2 pipeline, we engineered a strategy for the minimally invasive fluorescence labeling of endogenous tubulin isotypes or those harboring missense mutations. We demonstrated that a specifically designed 16-amino acid linker, coupled with sfGFP11 from the split-sfGFP system and integration into the H1-S2 loop of tubulin, facilitated tubulin labeling without compromising MT dynamics, embryonic development, or ciliogenesis in Caenorhabditis elegans. Extending this technique to human cells and murine oocytes, we visualized MTs with the minimal background fluorescence and a pathogenic tubulin isoform with fidelity. The utility of our approach across biological contexts and species set an additional paradigm for studying tubulin dynamics and functional specificity, with implications for understanding tubulin-related diseases known as tubulinopathies.
Collapse
Affiliation(s)
- Kaiming Xu
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Zhiyuan Li
- School of Life Sciences, MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, Tsinghua University, Beijing, China
| | - Linfan Mao
- School of Medicine, Tsinghua University, Beijing, China
| | - Zhengyang Guo
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Zhe Chen
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Yongping Chai
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Chao Xie
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Xuerui Yang
- School of Life Sciences, MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, Tsinghua University, Beijing, China
| | - Jie Na
- School of Medicine, Tsinghua University, Beijing, China
| | - Wei Li
- School of Medicine, Tsinghua University, Beijing, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
Avila J, Marco J, Plascencia-Villa G, Bajic VP, Perry G. Could there be an experimental way to link consciousness and quantum computations of brain microtubules? Front Neurosci 2024; 18:1430432. [PMID: 38979125 PMCID: PMC11228156 DOI: 10.3389/fnins.2024.1430432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024] Open
Affiliation(s)
- Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Jesús Marco
- Instituto de Física de Cantabria (CSIC-UC), Santander, Spain
| | - Germán Plascencia-Villa
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Vladan P. Bajic
- Laboratory for Radiobiology and Molecular Genetics, Department of Health and Environment, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
5
|
Gonzalez SJ, Heckel JM, Goldblum RR, Reid TA, McClellan M, Gardner MK. Rapid binding to protofilament edge sites facilitates tip tracking of EB1 at growing microtubule plus-ends. eLife 2024; 13:e91719. [PMID: 38385657 PMCID: PMC10883673 DOI: 10.7554/elife.91719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
EB1 is a key cellular protein that delivers regulatory molecules throughout the cell via the tip-tracking of growing microtubule plus-ends. Thus, it is important to understand the mechanism for how EB1 efficiently tracks growing microtubule plus-ends. It is widely accepted that EB1 binds with higher affinity to GTP-tubulin subunits at the growing microtubule tip, relative to GDP-tubulin along the microtubule length. However, it is unclear whether this difference in affinity alone is sufficient to explain the tip-tracking of EB1 at growing microtubule tips. Previously, we found that EB1 binds to exposed microtubule protofilament-edge sites at a ~70 fold faster rate than to closed-lattice sites, due to diffusional steric hindrance to binding. Thus, we asked whether rapid protofilament-edge binding could contribute to efficient EB1 tip tracking. A computational simulation with differential EB1 on-rates based on closed-lattice or protofilament-edge binding, and with EB1 off-rates that were dependent on the tubulin hydrolysis state, robustly recapitulated experimental EB1 tip tracking. To test this model, we used cell-free biophysical assays, as well as live-cell imaging, in combination with a Designed Ankyrin Repeat Protein (DARPin) that binds exclusively to protofilament-edge sites, and whose binding site partially overlaps with the EB1 binding site. We found that DARPin blocked EB1 protofilament-edge binding, which led to a decrease in EB1 tip tracking on dynamic microtubules. We conclude that rapid EB1 binding to microtubule protofilament-edge sites contributes to robust EB1 tip tracking at the growing microtubule plus-end.
Collapse
Affiliation(s)
- Samuel J Gonzalez
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Julia M Heckel
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Rebecca R Goldblum
- Department of Biophysics, Molecular Biology, and Biochemistry, University of MinnesotaMinneapolisUnited States
- Medical Scientist Training Program, University of MinnesotaMinneapolisUnited States
| | - Taylor A Reid
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Mark McClellan
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| | - Melissa K Gardner
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| |
Collapse
|
6
|
Wong SS, Wainman A, Saurya S, Raff JW. Regulation of centrosome size by the cell-cycle oscillator in Drosophila embryos. EMBO J 2024; 43:414-436. [PMID: 38233576 PMCID: PMC10898259 DOI: 10.1038/s44318-023-00022-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024] Open
Abstract
Mitotic centrosomes assemble when centrioles recruit large amounts of pericentriolar material (PCM) around themselves. In early C. elegans embryos, mitotic centrosome size appears to be set by the limiting amount of a key component. In Drosophila syncytial embryos, thousands of mitotic centrosomes are assembled as the embryo proceeds through 13 rounds of rapid nuclear division, driven by a core cell cycle oscillator. These divisions slow during nuclear cycles 11-13, and we find that centrosomes respond by reciprocally decreasing their growth rate, but increasing their growth period-so that they grow to a relatively consistent size at each cycle. At the start of each cycle, moderate CCO activity initially promotes centrosome growth, in part by stimulating Polo/PLK1 recruitment to centrosomes. Later in each cycle, high CCO activity inhibits centrosome growth by suppressing the centrosomal recruitment and/or maintenance of centrosome proteins. Thus, in fly embryos, mitotic centrosome size appears to be regulated predominantly by the core cell cycle oscillator, rather than by the depletion of a limiting component.
Collapse
Affiliation(s)
- Siu-Shing Wong
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Alan Wainman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Saroj Saurya
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Jordan W Raff
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| |
Collapse
|
7
|
Gibson C, Jönsson H, Spelman TA. Mean-field theory approach to three-dimensional nematic phase transitions in microtubules. Phys Rev E 2023; 108:064414. [PMID: 38243538 DOI: 10.1103/physreve.108.064414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/16/2023] [Indexed: 01/21/2024]
Abstract
Microtubules are dynamic intracellular fibers that have been observed experimentally to undergo spontaneous self-alignment. We formulate a three-dimensional (3D) mean-field theory model to analyze the nematic phase transition of microtubules growing and interacting within a 3D space, then make a comparison with computational simulations. We identify a control parameter G_{eff} and predict a unique critical value G_{eff}=1.56 for which a phase transition can occur. Furthermore, we show both analytically and using simulations that this predicted critical value does not depend on the presence of zippering. The mean-field theory developed here provides an analytical estimate of microtubule patterning characteristics without running time-consuming simulations and is a step towards bridging scales from microtubule behavior to multicellular simulations.
Collapse
Affiliation(s)
- Cameron Gibson
- Sainsbury Laboratory, University of Cambridge, Cambridge, CB2 1LR, United Kingdom
- Department of Physics and Astronomy, Rice University, Houston, Texas 77005, USA
- Department of Physics and Astronomy, Texas A&M University, College Station, Texas 77843, USA
| | - Henrik Jönsson
- Sainsbury Laboratory, University of Cambridge, Cambridge, CB2 1LR, United Kingdom
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, CB3 0WA, United Kingdom
- Centre for Environmental and Climate Science, Lund University, SE-223 62 Lund, Sweden
| | - Tamsin A Spelman
- Sainsbury Laboratory, University of Cambridge, Cambridge, CB2 1LR, United Kingdom
| |
Collapse
|
8
|
Zhu Z, Becam I, Tovey CA, Elfarkouchi A, Yen EC, Bernard F, Guichet A, Conduit PT. Multifaceted modes of γ-tubulin complex recruitment and microtubule nucleation at mitotic centrosomes. J Cell Biol 2023; 222:e202212043. [PMID: 37698931 PMCID: PMC10497398 DOI: 10.1083/jcb.202212043] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 09/14/2023] Open
Abstract
Microtubule nucleation is mediated by γ-tubulin ring complexes (γ-TuRCs). In most eukaryotes, a GCP4/5/4/6 "core" complex promotes γ-tubulin small complex (γ-TuSC) association to generate cytosolic γ-TuRCs. Unlike γ-TuSCs, however, this core complex is non-essential in various species and absent from budding yeasts. In Drosophila, Spindle defective-2 (Spd-2) and Centrosomin (Cnn) redundantly recruit γ-tubulin complexes to mitotic centrosomes. Here, we show that Spd-2 recruits γ-TuRCs formed via the GCP4/5/4/6 core, but Cnn can recruit γ-TuSCs directly via its well-conserved CM1 domain, similar to its homologs in budding yeast. When centrosomes fail to recruit γ-tubulin complexes, they still nucleate microtubules via the TOG domain protein Mini-spindles (Msps), but these microtubules have different dynamic properties. Our data, therefore, help explain the dispensability of the GCP4/5/4/6 core and highlight the robustness of centrosomes as microtubule organizing centers. They also suggest that the dynamic properties of microtubules are influenced by how they are nucleated.
Collapse
Affiliation(s)
- Zihan Zhu
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Isabelle Becam
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Corinne A. Tovey
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Abir Elfarkouchi
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Eugenie C. Yen
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Fred Bernard
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Antoine Guichet
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Paul T. Conduit
- Department of Zoology, University of Cambridge, Cambridge, UK
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| |
Collapse
|
9
|
Mann JR, McKenna ED, Mawrie D, Papakis V, Alessandrini F, Anderson EN, Mayers R, Ball HE, Kaspi E, Lubinski K, Baron DM, Tellez L, Landers JE, Pandey UB, Kiskinis E. Loss of function of the ALS-associated NEK1 kinase disrupts microtubule homeostasis and nuclear import. SCIENCE ADVANCES 2023; 9:eadi5548. [PMID: 37585529 PMCID: PMC10431718 DOI: 10.1126/sciadv.adi5548] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Loss-of-function variants in NIMA-related kinase 1 (NEK1) constitute a major genetic cause of amyotrophic lateral sclerosis (ALS), accounting for 2 to 3% of all cases. However, how NEK1 mutations cause motor neuron (MN) dysfunction is unknown. Using mass spectrometry analyses for NEK1 interactors and NEK1-dependent expression changes, we find functional enrichment for proteins involved in the microtubule cytoskeleton and nucleocytoplasmic transport. We show that α-tubulin and importin-β1, two key proteins involved in these processes, are phosphorylated by NEK1 in vitro. NEK1 is essential for motor control and survival in Drosophila models in vivo, while using several induced pluripotent stem cell (iPSC)-MN models, including NEK1 knockdown, kinase inhibition, and a patient mutation, we find evidence for disruptions in microtubule homeostasis and nuclear import. Notably, stabilizing microtubules with two distinct classes of drugs restored NEK1-dependent deficits in both pathways. The capacity of NEK1 to modulate these processes that are critically involved in ALS pathophysiology renders this kinase a formidable therapeutic candidate.
Collapse
Affiliation(s)
- Jacob R. Mann
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elizabeth D. McKenna
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Darilang Mawrie
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Vasileios Papakis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Francesco Alessandrini
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eric N. Anderson
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Ryan Mayers
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hannah E. Ball
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Evan Kaspi
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Katherine Lubinski
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Desiree M. Baron
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Liana Tellez
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Udai B. Pandey
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Ansari S, Gergely ZR, Flynn P, Li G, Moore JK, Betterton MD. Quantifying Yeast Microtubules and Spindles Using the Toolkit for Automated Microtubule Tracking (TAMiT). Biomolecules 2023; 13:939. [PMID: 37371519 DOI: 10.3390/biom13060939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 06/29/2023] Open
Abstract
Fluorescently labeled proteins absorb and emit light, appearing as Gaussian spots in fluorescence imaging. When fluorescent tags are added to cytoskeletal polymers such as microtubules, a line of fluorescence and even non-linear structures results. While much progress has been made in techniques for imaging and microscopy, image analysis is less well-developed. Current analysis of fluorescent microtubules uses either manual tools, such as kymographs, or automated software. As a result, our ability to quantify microtubule dynamics and organization from light microscopy remains limited. Despite the development of automated microtubule analysis tools for in vitro studies, analysis of images from cells often depends heavily on manual analysis. One of the main reasons for this disparity is the low signal-to-noise ratio in cells, where background fluorescence is typically higher than in reconstituted systems. Here, we present the Toolkit for Automated Microtubule Tracking (TAMiT), which automatically detects, optimizes, and tracks fluorescent microtubules in living yeast cells with sub-pixel accuracy. Using basic information about microtubule organization, TAMiT detects linear and curved polymers using a geometrical scanning technique. Images are fit via an optimization problem for the microtubule image parameters that are solved using non-linear least squares in Matlab. We benchmark our software using simulated images and show that it reliably detects microtubules, even at low signal-to-noise ratios. Then, we use TAMiT to measure monopolar spindle microtubule bundle number, length, and lifetime in a large dataset that includes several S. pombe mutants that affect microtubule dynamics and bundling. The results from the automated analysis are consistent with previous work and suggest a direct role for CLASP/Cls1 in bundling spindle microtubules. We also illustrate automated tracking of single curved astral microtubules in S. cerevisiae, with measurement of dynamic instability parameters. The results obtained with our fully-automated software are similar to results using hand-tracked measurements. Therefore, TAMiT can facilitate automated analysis of spindle and microtubule dynamics in yeast cells.
Collapse
Affiliation(s)
- Saad Ansari
- Department of Physics, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Zachary R Gergely
- Department of Physics, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Patrick Flynn
- Department of Physics, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Gabriella Li
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey K Moore
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Meredith D Betterton
- Department of Physics, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
11
|
Jeruzalska E, Mazur AJ. The Role of non-muscle actin paralogs in cell cycle progression and proliferation. Eur J Cell Biol 2023; 102:151315. [PMID: 37099935 DOI: 10.1016/j.ejcb.2023.151315] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Uncontrolled cell proliferation leads to several pathologies, including cancer. Thus, this process must be tightly regulated. The cell cycle accounts for cell proliferation, and its progression is coordinated with changes in cell shape, for which cytoskeleton reorganization is responsible. Rearrangement of the cytoskeleton allows for its participation in the precise division of genetic material and cytokinesis. One of the main cytoskeletal components is filamentous actin-based structures. Mammalian cells have at least six actin paralogs, four of which are muscle-specific, while two, named β- and γ-actin, are abundantly present in all types of cells. This review summarizes the findings that establish the role of non-muscle actin paralogs in regulating cell cycle progression and proliferation. We discuss studies showing that the level of a given non-muscle actin paralog in a cell influences the cell's ability to progress through the cell cycle and, thus, proliferation. Moreover, we elaborate on the non-muscle actins' role in regulating gene transcription, interactions of actin paralogs with proteins involved in controlling cell proliferation, and the contribution of non-muscle actins to different structures in a dividing cell. The data cited in this review show that non-muscle actins regulate the cell cycle and proliferation through varying mechanisms. We point to the need for further studies addressing these mechanisms.
Collapse
Affiliation(s)
- Estera Jeruzalska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| |
Collapse
|
12
|
Ansari S, Gergely ZR, Flynn P, Li G, Moore JK, Betterton MD. Quantifying yeast microtubules and spindles using the Toolkit for Automated Microtubule Tracking (TAMiT). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527544. [PMID: 36798368 PMCID: PMC9934621 DOI: 10.1101/2023.02.07.527544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Fluorescently labeled proteins absorb and emit light, appearing as Gaussian spots in fluorescence imaging. When fluorescent tags are added to cytoskeletal polymers such as microtubules, a line of fluorescence and even non-linear structures results. While much progress has been made in techniques for imaging and microscopy, image analysis is less well developed. Current analysis of fluorescent microtubules uses either manual tools, such as kymographs, or automated software. As a result, our ability to quantify microtubule dynamics and organization from light microscopy remains limited. Despite development of automated microtubule analysis tools for in vitro studies, analysis of images from cells often depends heavily on manual analysis. One of the main reasons for this disparity is the low signal-to-noise ratio in cells, where background fluorescence is typically higher than in reconstituted systems. Here, we present the Toolkit for Automated Microtubule Tracking (TAMiT), which automatically detects, optimizes and tracks fluorescent microtubules in living yeast cells with sub-pixel accuracy. Using basic information about microtubule organization, TAMiT detects linear and curved polymers using a geometrical scanning technique. Images are fit via an optimization problem for the microtubule image parameters that is solved using non-linear least squares in Matlab. We benchmark our software using simulated images and show that it reliably detects microtubules, even at low signal-to-noise ratios. Then, we use TAMiT to measure monopolar spindle microtubule bundle number, length, and lifetime in a large dataset that includes several S. pombe mutants that affect microtubule dynamics and bundling. The results from the automated analysis are consistent with previous work, and suggest a direct role for CLASP/Cls1 in bundling spindle microtubules. We also illustrate automated tracking of single curved astral microtubules in S. cerevisiae , with measurement of dynamic instability parameters. The results obtained with our fully-automated software are similar to results using hand-tracked measurements. Therefore, TAMiT can facilitate automated analysis of spindle and microtubule dynamics in yeast cells.
Collapse
|
13
|
Timón Pérez K, Scrofani J, Vernos I. NEDD1-S411 phosphorylation plays a critical function in the coordination of microtubule nucleation during mitosis. Biol Open 2022; 11:278477. [PMID: 36318115 PMCID: PMC9836086 DOI: 10.1242/bio.059474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
During mitosis, spindle assembly relies on centrosomal and acentrosomal microtubule nucleation pathways that all require the γ-Tubulin Ring Complex (γ-TuRC) and its adaptor protein NEDD1. The activity of these different pathways needs to be coordinated to ensure bipolar spindle assembly ( Cavazza et al., 2016) but the underlying mechanism is still unclear. Previous studies have identified three sites in NEDD1 (S377, S405 and S411) that when phosphorylated drive MT nucleation at the centrosomes, around the chromosomes and on pre-existing MTs respectively ( Lüders et al., 2006; Pinyol et al., 2013; Sdelci et al., 2012). Here we aimed at getting additional insights into the mechanism that coordinates the different MT nucleation pathways in dividing cells using a collection of HeLa stable inducible cell lines expressing NEDD1 phospho-variants at these three sites and Xenopus egg extracts. Our results provide further support for the essential role of phosphorylation at the three residues. Moreover, we directly demonstrate that S411 phosphorylation is essential for MT branching using TIRF microscopy in Xenopus egg extracts and we show that it plays a crucial role in ensuring the balance between centrosome and chromosome-dependent MT nucleation required for bipolar spindle assembly in mitotic cells.
Collapse
Affiliation(s)
- Krystal Timón Pérez
- Quantitative Cell Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Jacopo Scrofani
- Quantitative Cell Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain,Author for correspondence ()
| | - Isabelle Vernos
- Quantitative Cell Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain,Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain,ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain,Author for correspondence ()
| |
Collapse
|
14
|
Naghsh F, Monajjemi M. Thermodynamic Study of Assembling ↔ Disassembling of Microtubules via the Monte Carlo Simulation. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY A 2022; 96:1474-1483. [DOI: 10.1134/s0036024422070111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/30/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023]
|
15
|
McAlear TS, Bechstedt S. The mitotic spindle protein CKAP2 potently increases formation and stability of microtubules. eLife 2022; 11:72202. [PMID: 35029146 PMCID: PMC8798059 DOI: 10.7554/elife.72202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cells increase microtubule dynamics to make large rearrangements to their microtubule cytoskeleton during cell division. Changes in microtubule dynamics are essential for the formation and function of the mitotic spindle, and misregulation can lead to aneuploidy and cancer. Using in vitro reconstitution assays we show that the mitotic spindle protein Cytoskeleton-Associated Protein 2 (CKAP2) has a strong effect on nucleation of microtubules by lowering the critical tubulin concentration 100-fold. CKAP2 increases the apparent rate constant ka of microtubule growth by 50-fold and increases microtubule growth rates. In addition, CKAP2 strongly suppresses catastrophes. Our results identify CKAP2 as the most potent microtubule growth factor to date. These finding help explain CKAP2's role as an important spindle protein, proliferation marker, and oncogene.
Collapse
Affiliation(s)
- Thomas S McAlear
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| |
Collapse
|
16
|
Ryniawec JM, Rogers GC. Centrosome instability: when good centrosomes go bad. Cell Mol Life Sci 2021; 78:6775-6795. [PMID: 34476544 PMCID: PMC8560572 DOI: 10.1007/s00018-021-03928-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
The centrosome is a tiny cytoplasmic organelle that organizes and constructs massive molecular machines to coordinate diverse cellular processes. Due to its many roles during both interphase and mitosis, maintaining centrosome homeostasis is essential to normal health and development. Centrosome instability, divergence from normal centrosome number and structure, is a common pathognomonic cellular state tightly associated with cancers and other genetic diseases. As novel connections are investigated linking the centrosome to disease, it is critical to understand the breadth of centrosome functions to inspire discovery. In this review, we provide an introduction to normal centrosome function and highlight recent discoveries that link centrosome instability to specific disease states.
Collapse
Affiliation(s)
- John M Ryniawec
- University of Arizona Cancer Center, University of Arizona, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Gregory C Rogers
- University of Arizona Cancer Center, University of Arizona, 1515 N. Campbell Ave., Tucson, AZ, 85724, USA.
| |
Collapse
|
17
|
Asthana J, Cade NI, Normanno D, Lim WM, Surrey T. Gradual compaction of the central spindle decreases its dynamicity in PRC1 and EB1 gene-edited cells. Life Sci Alliance 2021; 4:4/12/e202101222. [PMID: 34580180 PMCID: PMC8500333 DOI: 10.26508/lsa.202101222] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/24/2022] Open
Abstract
Although different anaphase proteins bind with characteristically different strength to the central spindle, the overall central spindle dynamicity slows down as mitosis proceeds. During mitosis, the spindle undergoes morphological and dynamic changes. It reorganizes at the onset of the anaphase when the antiparallel bundler PRC1 accumulates and recruits central spindle proteins to the midzone. Little is known about how the dynamic properties of the central spindle change during its morphological changes in human cells. Using gene editing, we generated human cells that express from their endogenous locus fluorescent PRC1 and EB1 to quantify their native spindle distribution and binding/unbinding turnover. EB1 plus end tracking revealed a general slowdown of microtubule growth, whereas PRC1, similar to its yeast orthologue Ase1, binds increasingly strongly to compacting antiparallel microtubule overlaps. KIF4A and CLASP1 bind more dynamically to the central spindle, but also show slowing down turnover. These results show that the central spindle gradually becomes more stable during mitosis, in agreement with a recent “bundling, sliding, and compaction” model of antiparallel midzone bundle formation in the central spindle during late mitosis.
Collapse
Affiliation(s)
- Jayant Asthana
- The Francis Crick Institute, London, UK.,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Davide Normanno
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Wei Ming Lim
- The Francis Crick Institute, London, UK.,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Thomas Surrey
- The Francis Crick Institute, London, UK .,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
18
|
Centrosome, the Newly Identified Passenger through Tunneling Nanotubes, Increases Binucleation and Proliferation Marker in Receiving Cells. Int J Mol Sci 2021; 22:ijms22189680. [PMID: 34575851 PMCID: PMC8467045 DOI: 10.3390/ijms22189680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Type 1 tunneling nanotubes (TNTs-1) are long, cytoplasmic protrusions containing actin, microtubules and intermediate filaments that provide a bi-directional road for the transport of various components between distant cells. TNT-1 formation is accompanied by dramatic cytoskeletal reorganization offering mechanical support for intercellular communication. Although the centrosome is the major microtubule nucleating center and also a signaling hub, the relationship between the centrosome and TNTs-1 is still unexplored. We provide here the first evidence of centrosome localization and orientation towards the TNTs-1 protrusion site, which is implicated in TNT-1 formation. We also envision a model whereby synchronized reorientation of the Golgi apparatus along with the centrosome towards TNTs-1 ensures effective polarized trafficking through TNTs-1. Furthermore, using immunohistochemistry and live imaging, we observed for the first time the movement of an extra centrosome within TNTs-1. In this regard, we hypothesize a novel role for TNTs-1 as a critical pathway serving to displace extra centrosomes and potentially to either protect malignant cells against aberrant centrosome amplification or contribute to altering cells in the tumor environment. Indeed, we have observed the increase in binucleation and proliferation markers in receiving cells. The fact that the centrosome can be both as the base and the user of TNTs-1 offers new perspectives and new opportunities to follow in order to improve our knowledge of the pathophysiological mechanisms under TNT control.
Collapse
|
19
|
Jiang X, Ho DBT, Mahe K, Mia J, Sepulveda G, Antkowiak M, Jiang L, Yamada S, Jao LE. Condensation of pericentrin proteins in human cells illuminates phase separation in centrosome assembly. J Cell Sci 2021; 134:jcs258897. [PMID: 34308971 PMCID: PMC8349556 DOI: 10.1242/jcs.258897] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022] Open
Abstract
At the onset of mitosis, centrosomes expand the pericentriolar material (PCM) to maximize their microtubule-organizing activity. This step, termed centrosome maturation, ensures proper spindle organization and faithful chromosome segregation. However, as the centrosome expands, how PCM proteins are recruited and held together without membrane enclosure remains elusive. We found that endogenously expressed pericentrin (PCNT), a conserved PCM scaffold protein, condenses into dynamic granules during late G2/early mitosis before incorporating into mitotic centrosomes. Furthermore, the N-terminal portion of PCNT, enriched with conserved coiled-coils (CCs) and low-complexity regions (LCRs), phase separates into dynamic condensates that selectively recruit PCM proteins and nucleate microtubules in cells. We propose that CCs and LCRs, two prevalent sequence features in the centrosomal proteome, are preserved under evolutionary pressure in part to mediate liquid-liquid phase separation, a process that bestows upon the centrosome distinct properties critical for its assembly and functions.
Collapse
Affiliation(s)
- Xueer Jiang
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Dac Bang Tam Ho
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Karan Mahe
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Jennielee Mia
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Guadalupe Sepulveda
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Mark Antkowiak
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Linhao Jiang
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| |
Collapse
|
20
|
Mercadante DL, Manning AL, Olson SD. Modeling reveals cortical dynein-dependent fluctuations in bipolar spindle length. Biophys J 2021; 120:3192-3210. [PMID: 34197801 DOI: 10.1016/j.bpj.2021.05.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022] Open
Abstract
Proper formation and maintenance of the mitotic spindle is required for faithful cell division. Although much work has been done to understand the roles of the key molecular components of the mitotic spindle, identifying the consequences of force perturbations in the spindle remains a challenge. We develop a computational framework accounting for the minimal force requirements of mitotic progression. To reflect early spindle formation, we model microtubule dynamics and interactions with major force-generating motors, excluding chromosome interactions that dominate later in mitosis. We directly integrate our experimental data to define and validate the model. We then use simulations to analyze individual force components over time and their relationship to spindle dynamics, making it distinct from previously published models. We show through both model predictions and biological manipulation that rather than achieving and maintaining a constant bipolar spindle length, fluctuations in pole-to-pole distance occur that coincide with microtubule binding and force generation by cortical dynein. Our model further predicts that high dynein activity is required for spindle bipolarity when kinesin-14 (HSET) activity is also high. To the best of our knowledge, our results provide novel insight into the role of cortical dynein in the regulation of spindle bipolarity.
Collapse
Affiliation(s)
- Dayna L Mercadante
- Bioinformatics and Computational Biology Program, Worcester, Massachusetts
| | - Amity L Manning
- Department of Biology and Biotechnology, Worcester, Massachusetts.
| | - Sarah D Olson
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, Massachusetts.
| |
Collapse
|
21
|
Sharma A, Dagar S, Mylavarapu SVS. Transgelin-2 and phosphoregulation of the LIC2 subunit of dynein govern mitotic spindle orientation. J Cell Sci 2020; 133:jcs239673. [PMID: 32467330 DOI: 10.1242/jcs.239673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/29/2020] [Indexed: 08/31/2023] Open
Abstract
The molecular motor dynein is essential for mitotic spindle orientation, which defines the axis of cell division. The light intermediate chain subunits, LIC1 and LIC2, define biochemically and functionally distinct vertebrate dynein complexes, with LIC2-dynein playing a crucial role in ensuring spindle orientation. We reveal a novel, mitosis-specific interaction of LIC2-dynein with the cortical actin-bundling protein transgelin-2. Transgelin-2 is required for maintaining proper spindle length, equatorial metaphase chromosome alignment, spindle orientation and timely anaphase onset. We show that transgelin-2 stabilizes the cortical recruitment of LGN-NuMA, which together with dynein is required for spindle orientation. The opposing actions of transgelin-2 and LIC2-dynein maintain optimal cortical levels of LGN-NuMA. In addition, we show that the highly conserved serine 194 phosphorylation of LIC2 is required for proper spindle orientation, by maintaining mitotic centrosome integrity to ensure optimal astral microtubule nucleation. The work reveals two specific mechanisms through which LIC2-dynein regulates mitotic spindle orientation; namely, through a new interactor transgelin-2, which is required for engagement of LGN-NuMA with the actin cortex, and through mitotic phosphoregulation of LIC2 to control microtubule nucleation from the poles.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Amit Sharma
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sunayana Dagar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
- Affiliated to the Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
22
|
Phosphorylation of keratin 18 serine 52 regulates mother-daughter centriole engagement and microtubule nucleation by cell cycle-dependent accumulation at the centriole. Histochem Cell Biol 2020; 153:307-321. [PMID: 32078038 DOI: 10.1007/s00418-020-01849-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
Serine-52 (Ser52) is the major physiologic site of keratin 18 (K18) phosphorylation. Here, we report that serine-52 phosphorylated K18 (phospho-Ser52 K18) accumulated on centrosomes in a cell cycle-dependent manner. Moreover, we found that phospho-Ser52 K18 was located at the proximal end of the mother centriole. Transfection with the K18 Ser52 → Ala (K18 S52A) mutant prevented centriole localization of phospho-Ser52 K18 and resulted in separation of the mother-daughter centrioles. Inhibition of microtubule polymerization led to the disappearance of aggregated phospho-Ser52 K18 on the centrosome; removal of inhibitors resulted in reaccumulation of phospho-Ser52 K18 in microtubule-organizing centers. Transfection with a K18 S52A mutant inhibited microtubule nucleation. These results reveal a cell cycle-dependent change in centrosome localization of phospho-Ser52 k18 and strongly suggest that the phosphorylation status of Ser52 K18 of mother centrioles plays a critical role in maintaining a tight engagement between mother and daughter centrioles and also contributes to microtubule nucleation.
Collapse
|
23
|
Palumbo V, Tariq A, Borgal L, Metz J, Brancaccio M, Gatti M, Wakefield JG, Bonaccorsi S. Drosophila Morgana is an Hsp90-interacting protein with a direct role in microtubule polymerisation. J Cell Sci 2020; 133:jcs236786. [PMID: 31907206 PMCID: PMC6983718 DOI: 10.1242/jcs.236786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022] Open
Abstract
Morgana (Mora, also known as CHORD in flies) and its mammalian homologue, called CHORDC1 or CHP1, is a highly conserved cysteine and histidine-rich domain (CHORD)-containing protein that has been proposed to function as an Hsp90 co-chaperone. Morgana deregulation promotes carcinogenesis in both mice and humans while, in Drosophila, loss of mora causes lethality and a complex mitotic phenotype that is rescued by a human morgana transgene. Here, we show that Drosophila Mora localises to mitotic spindles and co-purifies with the Hsp90-R2TP-TTT supercomplex and with additional well-known Hsp90 co-chaperones. Acute inhibition of Mora function in the early embryo results in a dramatic reduction in centrosomal microtubule stability, leading to small spindles nucleated from mitotic chromatin. Purified Mora binds to microtubules directly and promotes microtubule polymerisation in vitro, suggesting that Mora directly regulates spindle dynamics independently of its Hsp90 co-chaperone role.
Collapse
Affiliation(s)
- Valeria Palumbo
- Dipartimento di Biologia e Biotecnologie Sapienza, Università di Roma, 00185 Rome, Italy
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Ammarah Tariq
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Lori Borgal
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Jeremy Metz
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Mara Brancaccio
- Dipartimento di Genetica, Biologia e Biochimica, Università di Torino, 10126 Torino, Italy
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie Sapienza, Università di Roma, 00185 Rome, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, 00185 Rome, Italy
| | - James G Wakefield
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Silvia Bonaccorsi
- Dipartimento di Biologia e Biotecnologie Sapienza, Università di Roma, 00185 Rome, Italy
| |
Collapse
|
24
|
Thoppil RJ, Sanders AAWM, Kaverina I. Detection of Microtubule Nucleation Hotspots at the Golgi. Methods Mol Biol 2020; 2101:179-189. [PMID: 31879905 PMCID: PMC7015078 DOI: 10.1007/978-1-0716-0219-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cell polarization is important for multiple physiological processes. In motile cells, microtubules (MTs) are organized as a polarized array, which is to a large extent comprised of Golgi-derived MTs (GDMTs), which asymmetrically extend toward the cell front. We have recently found that GDMT asymmetry is based on a nonrandom positioning of spatially restricted nucleation hotspots, where MTs form in a cooperative manner. Here, we summarize methods used for GDMT identification including microtubule regrowth after complete drug-induced depolymerization and tracking of growing microtubules using fluorescent MT plus-end-tracking proteins (+TIPs) in living cells, and subsequent detection of those GDMTs that originate from the nucleation hotspots. These approaches can be used for quantification of the spatial distribution of MT nucleation events associated with the Golgi or another large structure.
Collapse
Affiliation(s)
- Roslin J Thoppil
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Anna A W M Sanders
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
25
|
Prahl LS, Bangasser PF, Stopfer LE, Hemmat M, White FM, Rosenfeld SS, Odde DJ. Microtubule-Based Control of Motor-Clutch System Mechanics in Glioma Cell Migration. Cell Rep 2019; 25:2591-2604.e8. [PMID: 30485822 PMCID: PMC6345402 DOI: 10.1016/j.celrep.2018.10.101] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/25/2018] [Accepted: 10/26/2018] [Indexed: 11/30/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are widely used chemotherapy drugs capable of disrupting microtubule-dependent cellular functions, such as division and migration. We show that two clinically approved MTAs, paclitaxel and vinblastine, each suppress stiffness-sensitive migration and polarization characteristic of human glioma cells on compliant hydrogels. MTAs influence microtubule dynamics and cell traction forces by nearly opposite mechanisms, the latter of which can be explained by a combination of changes in myosin motor and adhesion clutch number. Our results support a microtubule-dependent signaling-based model for controlling traction forces through a motor-clutch mechanism, rather than microtubules directly relieving tension within F-actin and adhesions. Computational simulations of cell migration suggest that increasing protrusion number also impairs stiffness-sensitive migration, consistent with experimental MTA effects. These results provide a theoretical basis for the role of microtubules and mechanisms of MTAs in controlling cell migration. Prahl et al. examine the mechanisms by which microtubule-targeting drugs inhibit glioma cell migration. They find that dynamic microtubules regulate actin-based protrusion dynamics that facilitate cell polarity and migration. Changes in net microtubule assembly alter cell traction forces via signaling-based regulation of a motor-clutch system.
Collapse
Affiliation(s)
- Louis S Prahl
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Patrick F Bangasser
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lauren E Stopfer
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research and Physical Sciences-Oncology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mahya Hemmat
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research and Physical Sciences-Oncology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Steven S Rosenfeld
- Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA; Brain Tumor and Neuro-Oncology Center and Department of Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Physical Sciences-Oncology Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
26
|
Lin TC, Kuo HH, Wu YC, Pan TS, Yih LH. Phosphatidylinositol-5-phosphate 4-kinase gamma accumulates at the spindle pole and prevents microtubule depolymerization. Cell Div 2019; 14:9. [PMID: 31452676 PMCID: PMC6702725 DOI: 10.1186/s13008-019-0053-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/13/2019] [Indexed: 11/10/2022] Open
Abstract
Background A previous screen of a human kinase and phosphatase shRNA library to select genes that mediate arsenite induction of spindle abnormalities resulted in the identification of phosphatidylinositol-5-phosphate 4-kinase type-2 gamma (PIP4KIIγ), a phosphatidylinositol 4,5-bisphosphate (PIP2)-synthesizing enzyme. In this study, we explored how PIP4KIIγ regulates the assembly of mitotic spindles. Results PIP4KIIγ accumulates at the spindle pole before anaphase, and is required for the assembly of functional bipolar spindles. Depletion of PIP4KIIγ enhanced the spindle pole accumulation of mitotic centromere-associated kinesin (MCAK), a microtubule (MT)-depolymerizing kinesin, and resulted in a less stable spindle pole-associated MT. Depletion of MCAK can ameliorate PIP4KIIγ depletion-induced spindle abnormalities. In addition, PIP2 binds to polo-like kinase (PLK1) and reduces PLK1-mediated phosphorylation of MCAK. These results indicate that PIP4KIIγ and PIP2 may negatively regulate the MT depolymerization activity of MCAK by reducing PLK1-mediated phosphorylation of MCAK. Consequently, depletion of PLK1 has been shown to counteract the PIP4KIIγ depletion-induced instability of spindle pole-associated MT and cell resistance to arsenite. Conclusions Our current results imply that PIP4KIIγ may restrain MT depolymerization at the spindle pole through attenuating PLK1-mediated activation of MCAK before anaphase onset.
Collapse
Affiliation(s)
- Tz-Chi Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Hsiao-Hui Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Yi-Chen Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Tiffany S Pan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| | - Ling-Huei Yih
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115 Taiwan
| |
Collapse
|
27
|
Farina F, Ramkumar N, Brown L, Samandar Eweis D, Anstatt J, Waring T, Bithell J, Scita G, Thery M, Blanchoin L, Zech T, Baum B. Local actin nucleation tunes centrosomal microtubule nucleation during passage through mitosis. EMBO J 2019; 38:e99843. [PMID: 31015335 PMCID: PMC6545563 DOI: 10.15252/embj.201899843] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Cells going through mitosis undergo precisely timed changes in cell shape and organisation, which serve to ensure the fair partitioning of cellular components into the two daughter cells. These structural changes are driven by changes in actin filament and microtubule dynamics and organisation. While most evidence suggests that the two cytoskeletal systems are remodelled in parallel during mitosis, recent work in interphase cells has implicated the centrosome in both microtubule and actin nucleation, suggesting the potential for regulatory crosstalk between the two systems. Here, by using both in vitro and in vivo assays to study centrosomal actin nucleation as cells pass through mitosis, we show that mitotic exit is accompanied by a burst in cytoplasmic actin filament formation that depends on WASH and the Arp2/3 complex. This leads to the accumulation of actin around centrosomes as cells enter anaphase and to a corresponding reduction in the density of centrosomal microtubules. Taken together, these data suggest that the mitotic regulation of centrosomal WASH and the Arp2/3 complex controls local actin nucleation, which may function to tune the levels of centrosomal microtubules during passage through mitosis.
Collapse
Affiliation(s)
- Francesca Farina
- MRC-LMCB, UCL, London, UK
- IPLS, UCL, London, UK
- IFOM, the FIRC Institute of Molecular Oncology, University of Milan, Milan, Italy
- University of Grenoble, Grenoble, France
| | | | - Louise Brown
- Institute of Translational Medicine, Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | | | | | - Thomas Waring
- Institute of Translational Medicine, Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Jessica Bithell
- Institute of Translational Medicine, Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Giorgio Scita
- IFOM, the FIRC Institute of Molecular Oncology, University of Milan, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | | | - Tobias Zech
- Institute of Translational Medicine, Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Buzz Baum
- MRC-LMCB, UCL, London, UK
- IPLS, UCL, London, UK
| |
Collapse
|
28
|
Thawani A, Stone HA, Shaevitz JW, Petry S. Spatiotemporal organization of branched microtubule networks. eLife 2019; 8:43890. [PMID: 31066674 PMCID: PMC6519983 DOI: 10.7554/elife.43890] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/07/2019] [Indexed: 11/13/2022] Open
Abstract
To understand how chromosomes are segregated, it is necessary to explain the precise spatiotemporal organization of microtubules (MTs) in the mitotic spindle. We use Xenopus egg extracts to study the nucleation and dynamics of MTs in branched networks, a process that is critical for spindle assembly. Surprisingly, new branched MTs preferentially originate near the minus-ends of pre-existing MTs. A sequential reaction model, consisting of deposition of nucleation sites on an existing MT, followed by rate-limiting nucleation of branches, reproduces the measured spatial profile of nucleation, the distribution of MT plus-ends and tubulin intensity. By regulating the availability of the branching effectors TPX2, augmin and γ-TuRC, combined with single-molecule observations, we show that first TPX2 is deposited on pre-existing MTs, followed by binding of augmin/γ-TuRC to result in the nucleation of branched MTs. In sum, regulating the localization and kinetics of nucleation effectors governs the architecture of branched MT networks.
Collapse
Affiliation(s)
- Akanksha Thawani
- Department of Chemical and Biological Engineering, Princeton University, Princeton, United States
| | - Howard A Stone
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, United States
| | - Joshua W Shaevitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States.,Department of Physics, Princeton University, Princeton, United States
| | - Sabine Petry
- Department of Molecular Biology, Princeton University, Princeton, United States
| |
Collapse
|
29
|
Wang Y, Rui M, Tang Q, Bu S, Yu F. Patronin governs minus-end-out orientation of dendritic microtubules to promote dendrite pruning in Drosophila. eLife 2019; 8:39964. [PMID: 30920370 PMCID: PMC6438692 DOI: 10.7554/elife.39964] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 03/08/2019] [Indexed: 01/09/2023] Open
Abstract
Class IV ddaC neurons specifically prune larval dendrites without affecting axons during Drosophila metamorphosis. ddaCs distribute the minus ends of microtubules (MTs) to dendrites but the plus ends to axons. However, a requirement of MT minus-end-binding proteins in dendrite-specific pruning remains completely unknown. Here, we identified Patronin, a minus-end-binding protein, for its crucial and dose-sensitive role in ddaC dendrite pruning. The CKK domain is important for Patronin’s function in dendrite pruning. Moreover, we show that both patronin knockdown and overexpression resulted in a drastic decrease of MT minus ends and a concomitant increase of plus-end-out MTs in ddaC dendrites, suggesting that Patronin stabilizes dendritic minus-end-out MTs. Consistently, attenuation of Klp10A MT depolymerase in patronin mutant neurons significantly restored minus-end-out MTs in dendrites and thereby rescued dendrite-pruning defects. Thus, our study demonstrates that Patronin orients minus-end-out MT arrays in dendrites to promote dendrite-specific pruning mainly through antagonizing Klp10A activity. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that minor issues remain unresolved (see decision letter).
Collapse
Affiliation(s)
- Yan Wang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Menglong Rui
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - Quan Tang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Shufeng Bu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore.,Neuroscience and Behavioral Disorder Program, Duke-NUS Graduate Medical School, National University of Singapore, Singapore, Singapore
| |
Collapse
|
30
|
Hasegawa K, Nazarov O, Porter E. LSPR Biosensing Approach for the Detection of Microtubule Nucleation. SENSORS 2019; 19:s19061436. [PMID: 30909588 PMCID: PMC6471214 DOI: 10.3390/s19061436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 11/16/2022]
Abstract
Microtubules are dynamic protein filaments that are involved in a number of cellular processes. Here, we report the development of a novel localized surface plasmon resonance (LSPR) biosensing approach for investigating one aspect of microtubule dynamics that is not well understood, namely, nucleation. Using a modified Mie theory with radially variable refractive index, we construct a theoretical model to describe the optical response of gold nanoparticles when microtubules form around them. The model predicts that the extinction maximum wavelength is sensitive to a change in the local refractive index induced by microtubule nucleation within a few tens of nanometers from the nanoparticle surface, but insensitive to a change in the refractive index outside this region caused by microtubule elongation. As a proof of concept to demonstrate that LSPR can be used for detecting microtubule nucleation experimentally, we induce spontaneous microtubule formation around gold nanoparticles by immobilizing tubulin subunits on the nanoparticles. We find that, consistent with the theoretical model, there is a redshift in the extinction maximum wavelength upon the formation of short microtubules around the nanoparticles, but no significant change in maximum wavelength when the microtubules are elongated. We also perform kinetic experiments and demonstrate that the maximum wavelength is sensitive to the microtubule nuclei assembly even when microtubules are too small to be detected from an optical density measurement.
Collapse
Affiliation(s)
- Keisuke Hasegawa
- Department of Physics, Grinnell College, 1116 Eighth Avenue, Grinnell, IA 50112, USA.
| | - Otabek Nazarov
- Department of Physics, Grinnell College, 1116 Eighth Avenue, Grinnell, IA 50112, USA.
| | - Evan Porter
- Department of Physics, Grinnell College, 1116 Eighth Avenue, Grinnell, IA 50112, USA.
| |
Collapse
|
31
|
Dwivedi D, Kumari A, Rathi S, Mylavarapu SVS, Sharma M. The dynein adaptor Hook2 plays essential roles in mitotic progression and cytokinesis. J Cell Biol 2019; 218:871-894. [PMID: 30674580 PMCID: PMC6400558 DOI: 10.1083/jcb.201804183] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/29/2018] [Accepted: 12/07/2018] [Indexed: 12/16/2022] Open
Abstract
Hook proteins are evolutionarily conserved dynein adaptors that promote assembly of highly processive dynein-dynactin motor complexes. Mammals express three Hook paralogs, namely Hook1, Hook2, and Hook3, that have distinct subcellular localizations and expectedly, distinct cellular functions. Here we demonstrate that Hook2 binds to and promotes dynein-dynactin assembly specifically during mitosis. During the late G2 phase, Hook2 mediates dynein-dynactin localization at the nuclear envelope (NE), which is required for centrosome anchoring to the NE. Independent of its binding to dynein, Hook2 regulates microtubule nucleation at the centrosome; accordingly, Hook2-depleted cells have reduced astral microtubules and spindle positioning defects. Besides the centrosome, Hook2 localizes to and recruits dynactin and dynein to the central spindle. Dynactin-dependent targeting of centralspindlin complex to the midzone is abrogated upon Hook2 depletion; accordingly, Hook2 depletion results in cytokinesis failure. We find that the zebrafish Hook2 homologue promotes dynein-dynactin association and was essential for zebrafish early development. Together, these results suggest that Hook2 mediates assembly of the dynein-dynactin complex and regulates mitotic progression and cytokinesis.
Collapse
Affiliation(s)
- Devashish Dwivedi
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Amrita Kumari
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, Faridabad, India.,Affiliated to Manipal Academy of Higher Education, Manipal, India
| | - Siddhi Rathi
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, Faridabad, India.,Affiliated to Manipal Academy of Higher Education, Manipal, India
| | - Mahak Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| |
Collapse
|
32
|
Liu Y, Pelletier L. A magic bullet for targeting cancers with supernumerary centrosomes. EMBO J 2019; 38:embj.2018101134. [PMID: 30538103 DOI: 10.15252/embj.2018101134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Yi Liu
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Laurence Pelletier
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Mariappan A, Soni K, Schorpp K, Zhao F, Minakar A, Zheng X, Mandad S, Macheleidt I, Ramani A, Kubelka T, Dawidowski M, Golfmann K, Wason A, Yang C, Simons J, Schmalz HG, Hyman AA, Aneja R, Ullrich R, Urlaub H, Odenthal M, Büttner R, Li H, Sattler M, Hadian K, Gopalakrishnan J. Inhibition of CPAP-tubulin interaction prevents proliferation of centrosome-amplified cancer cells. EMBO J 2018; 38:embj.201899876. [PMID: 30530478 PMCID: PMC6331730 DOI: 10.15252/embj.201899876] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 11/29/2022] Open
Abstract
Centrosome amplification is a hallmark of human cancers that can trigger cancer cell invasion. To survive, cancer cells cluster amplified extra centrosomes and achieve pseudobipolar division. Here, we set out to prevent clustering of extra centrosomes. Tubulin, by interacting with the centrosomal protein CPAP, negatively regulates CPAP‐dependent peri‐centriolar material recruitment, and concurrently microtubule nucleation. Screening for compounds that perturb CPAP–tubulin interaction led to the identification of CCB02, which selectively binds at the CPAP binding site of tubulin. Genetic and chemical perturbation of CPAP–tubulin interaction activates extra centrosomes to nucleate enhanced numbers of microtubules prior to mitosis. This causes cells to undergo centrosome de‐clustering, prolonged multipolar mitosis, and cell death. 3D‐organotypic invasion assays reveal that CCB02 has broad anti‐invasive activity in various cancer models, including tyrosine kinase inhibitor (TKI)‐resistant EGFR‐mutant non‐small‐cell lung cancers. Thus, we have identified a vulnerability of cancer cells to activation of extra centrosomes, which may serve as a global approach to target various tumors, including drug‐resistant cancers exhibiting high incidence of centrosome amplification.
Collapse
Affiliation(s)
- Aruljothi Mariappan
- Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany.,Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Komal Soni
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Biomolecular NMR at Center for Integrated Protein Science Munich and Department Chemie, Technische Universität München, Garching, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Fan Zhao
- Department of Basic Medical Sciences, Center for Structural Biology, School of Medicine, Beijing, China.,MOE Key Laboratory of Protein Sciences, School of Life Sciences, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Amin Minakar
- Department of Chemistry, University of Cologne, Cologne, Germany
| | - Xiangdong Zheng
- Department of Basic Medical Sciences, Center for Structural Biology, School of Medicine, Beijing, China.,MOE Key Laboratory of Protein Sciences, School of Life Sciences, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Sunit Mandad
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Bioanalytics, University Medical Center Goettingen, Goettingen, Germany.,Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Iris Macheleidt
- Institute of Pathology and Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Anand Ramani
- Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany.,IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tomáš Kubelka
- Biomolecular NMR at Center for Integrated Protein Science Munich and Department Chemie, Technische Universität München, Garching, Germany
| | - Maciej Dawidowski
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Biomolecular NMR at Center for Integrated Protein Science Munich and Department Chemie, Technische Universität München, Garching, Germany.,Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Kristina Golfmann
- Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Arpit Wason
- Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Chunhua Yang
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Judith Simons
- Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | | | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Roland Ullrich
- Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Bioanalytics, University Medical Center Goettingen, Goettingen, Germany
| | - Margarete Odenthal
- Institute of Pathology and Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Reinhardt Büttner
- Institute of Pathology and Center for Molecular Medicine of the University of Cologne, Cologne, Germany
| | - Haitao Li
- Department of Basic Medical Sciences, Center for Structural Biology, School of Medicine, Beijing, China.,MOE Key Laboratory of Protein Sciences, School of Life Sciences, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Biomolecular NMR at Center for Integrated Protein Science Munich and Department Chemie, Technische Universität München, Garching, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Jay Gopalakrishnan
- Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany .,Center for Molecular Medicine of the University of Cologne, Cologne, Germany.,IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| |
Collapse
|
34
|
Abstract
For efficient replication of the influenza virus genome and its post-replicational processes, not only viral factors but also host-derived cellular factors (host factors) are required. The influenza virus genome exists as viral ribonucleoprotein (vRNP) complexes with viral RNA-dependent RNA polymerases and nucleoprotein (NP). Using biochemical and proteomics approaches, we have identified host factors which are required for the vRNP replication and the progeny vRNP transport. We found that MCM complex, a cellular DNA replication licensing factor, is required for successful viral genome replication. In concert with the replication reaction, the nascent RNA chains are encapsidated with NP by cellular splicing factor UAP56. Further, after nuclear export of vRNP, we revealed that vRNP is transported to the plasma membrane using cholesterol-enriched recycling endosomes through cell cycle-independent activation of the centrosome by YB-1, which is a mitotic centrosomal protein. Depletion of YB-1 shows that the cholesterol-enriched endosomes are important for clustering of viral structural proteins at lipid rafts to assemble the virus particles concomitantly with the arrival of vRNP beneath the plasma membrane.
Collapse
|
35
|
Cassimeris L, Leung JC, Odde DJ. Monte Carlo simulations of microtubule arrays: The critical roles of rescue transitions, the cell boundary, and tubulin concentration in shaping microtubule distributions. PLoS One 2018; 13:e0197538. [PMID: 29782540 PMCID: PMC5962052 DOI: 10.1371/journal.pone.0197538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 01/12/2023] Open
Abstract
Microtubules are dynamic polymers required for a number of processes, including chromosome movement in mitosis. While regulators of microtubule dynamics have been well characterized, we lack a convenient way to predict how the measured dynamic parameters shape the entire microtubule system within a cell, or how the system responds when specific parameters change in response to internal or external signals. Here we describe a Monte Carlo model to simulate an array of dynamic microtubules from parameters including the cell radius, total tubulin concentration, microtubule nucleation rate from the centrosome, and plus end dynamic instability. The algorithm also allows dynamic instability or position of the cell edge to vary during the simulation. Outputs from simulations include free tubulin concentration, average microtubule lengths, length distributions, and individual length changes over time. Using this platform and reported parameters measured in interphase LLCPK1 epithelial cells, we predict that sequestering ~ 15-20% of total tubulin results in fewer microtubules, but promotes dynamic instability of those remaining. Simulations also predict that lowering nucleation rate will increase the stability and average length of the remaining microtubules. Allowing the position of the cell's edge to vary over time changed the average length but not the number of microtubules and generated length distributions consistent with experimental measurements. Simulating the switch from interphase to prophase demonstrated that decreased rescue frequency at prophase is the critical factor needed to rapidly clear the cell of interphase microtubules prior to mitotic spindle assembly. Finally, consistent with several previous simulations, our results demonstrate that microtubule nucleation and dynamic instability in a confined space determines the partitioning of tubulin between monomer and polymer pools. The model and simulations will be useful for predicting changes to the entire microtubule array after modification to one or more parameters, including predicting the effects of tubulin-targeted chemotherapies.
Collapse
Affiliation(s)
- Lynne Cassimeris
- Dept. of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Jessica C Leung
- Dept. of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - David J Odde
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
36
|
Sepulveda G, Antkowiak M, Brust-Mascher I, Mahe K, Ou T, Castro NM, Christensen LN, Cheung L, Jiang X, Yoon D, Huang B, Jao LE. Co-translational protein targeting facilitates centrosomal recruitment of PCNT during centrosome maturation in vertebrates. eLife 2018; 7:34959. [PMID: 29708497 PMCID: PMC5976437 DOI: 10.7554/elife.34959] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022] Open
Abstract
As microtubule-organizing centers of animal cells, centrosomes guide the formation of the bipolar spindle that segregates chromosomes during mitosis. At mitosis onset, centrosomes maximize microtubule-organizing activity by rapidly expanding the pericentriolar material (PCM). This process is in part driven by the large PCM protein pericentrin (PCNT), as its level increases at the PCM and helps recruit additional PCM components. However, the mechanism underlying the timely centrosomal enrichment of PCNT remains unclear. Here, we show that PCNT is delivered co-translationally to centrosomes during early mitosis by cytoplasmic dynein, as evidenced by centrosomal enrichment of PCNT mRNA, its translation near centrosomes, and requirement of intact polysomes for PCNT mRNA localization. Additionally, the microtubule minus-end regulator, ASPM, is also targeted co-translationally to mitotic spindle poles. Together, these findings suggest that co-translational targeting of cytoplasmic proteins to specific subcellular destinations may be a generalized protein targeting mechanism. Before a cell divides, it creates a copy of its genetic material (DNA) and evenly distributes it between the new ‘daughter’ cells with the help of a complex called the mitotic spindle. This complex is made of long cable-like protein chains called microtubules. To ensure that each daughter cell receives an equal amount of DNA, structures known as centrosomes organize the microtubules during the division process. Centrosomes have two rigid cores, called centrioles, which are surrounded by a matrix of proteins called the pericentriolar material. It is from this material that the microtubules are organized. The pericentriolar material is a dynamic structure and changes its size by assembling and disassembling its protein components. The larger the pericentriolar material, the more microtubules can form. Before a cell divides, it rapidly expands in a process called centrosome maturation. A protein called pericentrin initiates the maturation by helping to recruit other proteins to the centrosome. Pericentrin molecules are large, and it takes the cell between 10 and 20 minutes to make each one. Nevertheless, the cell can produce and deliver large quantities of pericentrin to the centrosome in a matter of minutes. We do not yet know how this happens. To investigate this further, Sepulveda, Antkowiak, Brust-Mascher et al. used advanced microscopy to study zebrafish embryos and human cells grown in the laboratory. The results showed that cells build and transport pericentrin at the same time. Cells use messenger RNA molecules as templates to build proteins. These feed into protein factories called ribosomes, which assemble the building blocks in the correct order. Rather than waiting for the pericentrin production to finish, the cell moves the active factories to the centrosome with the help of a molecular motor called dynein. By the time the pericentrin molecules are completely made by ribosomes, they are already at the centrosome, ready to help with the recruitment of other proteins during centrosome maturation. These findings improve our understanding of centrosome maturation. The next step is to find out how the cell coordinates this process with the recruitment of other proteins to the centrosome. It is also possible that the cell uses similar processes to deliver other proteins to different parts of the cell.
Collapse
Affiliation(s)
- Guadalupe Sepulveda
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Mark Antkowiak
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, University of California, Davis School of Veterinary Medicine, Davis, United States
| | - Karan Mahe
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Tingyoung Ou
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Noemi M Castro
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Lana N Christensen
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Lee Cheung
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Xueer Jiang
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Daniel Yoon
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Chan Zuckerberg Biohub, San Francisco, United States
| | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, United States
| |
Collapse
|
37
|
Haverfield J, Dean NL, Nöel D, Rémillard-Labrosse G, Paradis V, Kadoch IJ, FitzHarris G. Tri-directional anaphases as a novel chromosome segregation defect in human oocytes. Hum Reprod 2018; 32:1293-1303. [PMID: 28449121 DOI: 10.1093/humrep/dex083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/07/2017] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION What are the chromosome segregation errors in human oocyte meiosis-I that may underlie oocyte aneuploidy? SUMMARY ANSWER Multiple modes of chromosome segregation error were observed, including tri-directional anaphases, which we attribute to loss of bipolar spindle structure at anaphase-I. WHAT IS KNOWN ALREADY Oocyte aneuploidy is common and associated with infertility, but mechanistic information on the chromosome segregation errors underlying these defects is scarce. Lagging chromosomes were recently reported as a possible mechanism by which segregation errors occur. STUDY DESIGN, SIZE, DURATION Long-term confocal imaging of chromosome dynamics in 50 human oocytes collected between January 2015 and May 2016. PARTICIPANTS/MATERIALS, SETTING, METHODS Germinal vesicle (GV) stage oocytes were collected from women undergoing intracytoplasmic sperm injection cycles and also CD1 mice. Oocytes were microinjected with complementary RNAs to label chromosomes, and in a subset of oocytes, the meiotic spindle. Oocytes were imaged live through meiosis-I using confocal microscopy. 3D image reconstruction was used to classify chromosome segregation phenotypes at anaphase-I. Segregation phenotypes were related to spindle dynamics and cell cycle timings. MAIN RESULTS AND THE ROLE OF CHANCE Most (87%) mouse oocytes segregated chromosomes with no obvious defects. We found that 20% of human oocytes segregated chromosomes bi-directionally with no lagging chromosomes. The rest were categorised as bi-directional anaphase with lagging chromosomes (20%), bi-directional anaphase with chromatin mass separation (34%) or tri-directional anaphase (26%). Segregation errors correlated with chromosome misalignment prior to anaphase. Spindles were tripolar when tri-directional anaphases occurred. Anaphase phenotypes did not correlate with meiosis-I duration (P = 0.73). LARGE SCALE DATA Not applicable. LIMITATIONS, REASONS FOR CAUTION Oocytes were recovered at GV stage after gonadotrophin-stimulation, and the usual oocyte quality caveats apply. Whilst the possibility that imaging may affect oocyte physiology cannot be formally excluded, detailed controls and justifications are presented. WIDER IMPLICATIONS OF THE FINDINGS This is one of the first reports of live imaging of chromosome dynamics in human oocytes, introducing tri-directional anaphases as a novel potential mechanism for oocyte aneuploidy. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by grants from Fondation Jean-Louis Lévesque (Canada), CIHR (MOP142334) and CFI (32711) to GF. JH is supported by Postdoctoral Fellowships from The Lalor Foundation and CIHR (146703). The authors have no conflict of interest.
Collapse
Affiliation(s)
- Jenna Haverfield
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montréal, 900 Rue St Denis, Montréal, Québec, Canada H2X 0A9.,Department of Obstetrics and Gynaecology, University of Montréal, Montréal, Québec, Canada H3T 1J4
| | - Nicola L Dean
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montréal, 900 Rue St Denis, Montréal, Québec, Canada H2X 0A9.,Clinique de Procréation Assistée (CPA) du CHUM, Montréal, Québec, Canada H2L 4S8
| | - Diana Nöel
- Clinique de Procréation Assistée (CPA) du CHUM, Montréal, Québec, Canada H2L 4S8
| | - Gaudeline Rémillard-Labrosse
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montréal, 900 Rue St Denis, Montréal, Québec, Canada H2X 0A9
| | - Veronique Paradis
- Clinique de Procréation Assistée (CPA) du CHUM, Montréal, Québec, Canada H2L 4S8
| | - Isaac-Jacques Kadoch
- Department of Obstetrics and Gynaecology, University of Montréal, Montréal, Québec, Canada H3T 1J4.,Clinique de Procréation Assistée (CPA) du CHUM, Montréal, Québec, Canada H2L 4S8
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montréal, 900 Rue St Denis, Montréal, Québec, Canada H2X 0A9.,Department of Obstetrics and Gynaecology, University of Montréal, Montréal, Québec, Canada H3T 1J4
| |
Collapse
|
38
|
Zenker J, White MD, Templin RM, Parton RG, Thorn-Seshold O, Bissiere S, Plachta N. A microtubule-organizing center directing intracellular transport in the early mouse embryo. Science 2018; 357:925-928. [PMID: 28860385 DOI: 10.1126/science.aam9335] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/03/2017] [Accepted: 08/01/2017] [Indexed: 12/26/2022]
Abstract
The centrosome is the primary microtubule-organizing center (MTOC) of most animal cells; however, this organelle is absent during early mammalian development. Therefore, the mechanism by which the mammalian embryo organizes its microtubules (MTs) is unclear. We visualize MT bridges connecting pairs of cells and show that the cytokinetic bridge does not undergo stereotypical abscission after cell division. Instead, it serves as scaffold for the accumulation of the MT minus-end-stabilizing protein CAMSAP3 throughout interphase, thereby transforming this structure into a noncentrosomal MTOC. Transport of the cell adhesion molecule E-cadherin to the membrane is coordinated by this MTOC and is required to form the pluripotent inner mass. Our study reveals a noncentrosomal form of MT organization that directs intracellular transport and is essential for mammalian development.
Collapse
Affiliation(s)
- J Zenker
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - M D White
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - R M Templin
- Institute for Molecular Biosciences and Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Queensland, Australia
| | - R G Parton
- Institute for Molecular Biosciences and Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Queensland, Australia
| | - O Thorn-Seshold
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - S Bissiere
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - N Plachta
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
39
|
Mchedlishvili N, Matthews HK, Corrigan A, Baum B. Two-step interphase microtubule disassembly aids spindle morphogenesis. BMC Biol 2018; 16:14. [PMID: 29361957 PMCID: PMC5778756 DOI: 10.1186/s12915-017-0478-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Entry into mitosis triggers profound changes in cell shape and cytoskeletal organisation. Here, by studying microtubule remodelling in human flat mitotic cells, we identify a two-step process of interphase microtubule disassembly. RESULTS First, a microtubule-stabilising protein, Ensconsin/MAP7, is inactivated in prophase as a consequence of its phosphorylation downstream of Cdk1/cyclin B. This leads to a reduction in interphase microtubule stability that may help to fuel the growth of centrosomally nucleated microtubules. The peripheral interphase microtubules that remain are then rapidly lost as the concentration of tubulin heterodimers falls following dissolution of the nuclear compartment boundary. Finally, we show that a failure to destabilise microtubules in prophase leads to the formation of microtubule clumps, which interfere with spindle assembly. CONCLUSIONS This analysis highlights the importance of the step-wise remodelling of the microtubule cytoskeleton and the significance of permeabilisation of the nuclear envelope in coordinating the changes in cellular organisation and biochemistry that accompany mitotic entry.
Collapse
Affiliation(s)
- Nunu Mchedlishvili
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Helen K Matthews
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Adam Corrigan
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK
| | - Buzz Baum
- MRC Laboratory of Molecular Cell Biology and the IPLS, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
40
|
Foteinopoulos P, Mulder BM. A microtubule-based minimal model for spontaneous and persistent spherical cell polarity. PLoS One 2017; 12:e0184706. [PMID: 28931032 PMCID: PMC5607169 DOI: 10.1371/journal.pone.0184706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
We propose a minimal model for the spontaneous and persistent generation of polarity in a spherical cell based on dynamic microtubules and a single mobile molecular component. This component, dubbed the polarity factor, binds to microtubules nucleated from a centrosome located in the center of the cell, is subsequently delivered to the cell membrane, where it diffuses until it unbinds. The only feedback mechanism we impose is that the residence time of the microtubules at the membrane increases with the local density of the polarity factor. We show analytically that this system supports a stable unipolar symmetry-broken state for a wide range of parameters. We validate the predictions of the model by 2D particle-based simulations. Our model provides a route towards the creation of polarity in a minimal cell-like environment using a biochemical reconstitution approach.
Collapse
Affiliation(s)
| | - Bela M. Mulder
- Systems Biophysics Department, Institute AMOLF, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
41
|
Liu Y, Visetsouk M, Mynlieff M, Qin H, Lechtreck KF, Yang P. H +- and Na +- elicited rapid changes of the microtubule cytoskeleton in the biflagellated green alga Chlamydomonas. eLife 2017; 6:26002. [PMID: 28875932 PMCID: PMC5779235 DOI: 10.7554/elife.26002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/05/2017] [Indexed: 12/27/2022] Open
Abstract
Although microtubules are known for dynamic instability, the dynamicity is considered to be tightly controlled to support a variety of cellular processes. Yet diverse evidence suggests that this is not applicable to Chlamydomonas, a biflagellate fresh water green alga, but intense autofluorescence from photosynthesis pigments has hindered the investigation. By expressing a bright fluorescent reporter protein at the endogenous level, we demonstrate in real time discreet sweeping changes in algal microtubules elicited by rises of intracellular H+ and Na+. These results from this model organism with characteristics of animal and plant cells provide novel explanations regarding how pH may drive cellular processes; how plants may respond to, and perhaps sense stresses; and how organisms with a similar sensitive cytoskeleton may be susceptible to environmental changes.
Collapse
Affiliation(s)
- Yi Liu
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Mike Visetsouk
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Michelle Mynlieff
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Hongmin Qin
- Department of Biology, Texas A&M University, College Station, United States
| | - Karl F Lechtreck
- Department of Cellular Biology, University of Georgia, Athen, United States
| | - Pinfen Yang
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| |
Collapse
|
42
|
Mustyatsa VV, Boyakhchyan AV, Ataullakhanov FI, Gudimchuk NB. EB-family proteins: Functions and microtubule interaction mechanisms. BIOCHEMISTRY (MOSCOW) 2017; 82:791-802. [DOI: 10.1134/s0006297917070045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Semenova I, Gupta D, Usui T, Hayakawa I, Cowan A, Rodionov V. Stimulation of microtubule-based transport by nucleation of microtubules on pigment granules. Mol Biol Cell 2017; 28:1418-1425. [PMID: 28381426 PMCID: PMC5449142 DOI: 10.1091/mbc.e16-08-0571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 11/11/2022] Open
Abstract
In Xenopus melanophores, nucleation of microtubules on pigment granules provides a positive feedback loop that enhances their transport to the cell center during pigment aggregation. Microtubule (MT)-based transport can be regulated through changes in organization of MT transport tracks, but the mechanisms that regulate these changes are poorly understood. In Xenopus melanophores, aggregation of pigment granules in the cell center involves their capture by the tips of MTs growing toward the cell periphery, and granule aggregation signals facilitate capture by increasing the number of growing MT tips. This increase could be explained by stimulation of MT nucleation either on the centrosome or on the aggregate of pigment granules that gradually forms in the cell center. We blocked movement of pigment granules to the cell center and compared the MT-nucleation activity of the centrosome in the same cells in two signaling states. We found that granule aggregation signals did not stimulate MT nucleation on the centrosome but did increase MT nucleation activity of pigment granules. Elevation of MT-nucleation activity correlated with the recruitment to pigment granules of a major component of MT-nucleation templates, γ-tubulin, and was suppressed by γ-tubulin inhibitors. We conclude that generation of new MT transport tracks by concentration of the leading pigment granules provides a positive feedback loop that enhances delivery of trailing granules to the cell center.
Collapse
Affiliation(s)
- Irina Semenova
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Dipika Gupta
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Takeo Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Ichiro Hayakawa
- Division of Applied Chemistry, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ann Cowan
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| | - Vladimir Rodionov
- R. D. Berlin Center for Cell Analysis and Modeling and Department of Cell Biology, UConn Health, Farmington, CT 06030
| |
Collapse
|
44
|
Ory EC, Bhandary L, Boggs AE, Chakrabarti KR, Parker J, Losert W, Martin SS. Analysis of microtubule growth dynamics arising from altered actin network structure and contractility in breast tumor cells. Phys Biol 2017; 14:026005. [PMID: 28092269 PMCID: PMC5738915 DOI: 10.1088/1478-3975/aa59a2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The periphery of epithelial cells is shaped by opposing cytoskeletal physical forces generated predominately by two dynamic force generating systems-growing microtubule ends push against the boundary from the cell center, and the actin cortex contracts the attached plasma membrane. Here we investigate how changes to the structure and dynamics of the actin cortex alter the dynamics of microtubules. Current drugs target actin polymerization and contraction to reduce cell division and invasiveness; however, the impacts on microtubule dynamics remain incompletely understood. Using human MCF-7 breast tumor cells expressing GFP-tagged microtubule end-binding-protein-1 (EB1) and coexpression of cytoplasmic fluorescent protein mCherry, we map the trajectories of growing microtubule ends and cytoplasmic boundary respectively. Based on EB1 tracks and cytoplasmic boundary outlines, we calculate the speed, distance from cytoplasmic boundary, and straightness of microtubule growth. Actin depolymerization with Latrunculin-A reduces EB1 growth speed as well as allows the trajectories to extend beyond the cytoplasmic boundary. Blebbistatin, a direct myosin-II inhibitor, reduced EB1 speed and yielded less straight EB1 trajectories. Inhibiting signaling upstream of myosin-II contractility via the Rho-kinase inhibitor, Y-27632, altered EB1 dynamics differently from Blebbistatin. These results indicate that reduced actin cortex integrity can induce distinct alterations in microtubule dynamics. Given recent findings that tumor stem cell characteristics are increased by drugs which reduce actin contractility or stabilize microtubules, it remains important to clearly define how cytoskeletal drugs alter the interactions between these two filament systems in tumor cells.
Collapse
Affiliation(s)
- Eleanor C Ory
- Department of Physics, IPST, and IREAP, University of Maryland, College Park, MD, United States of America
| | - Lekhana Bhandary
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States of America
- Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States of America
| | - Amanda E Boggs
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States of America
- Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States of America
| | - Kristi R Chakrabarti
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States of America
- Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States of America
| | - Joshua Parker
- Department of Physics, IPST, and IREAP, University of Maryland, College Park, MD, United States of America
| | - Wolfgang Losert
- Department of Physics, IPST, and IREAP, University of Maryland, College Park, MD, United States of America
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States of America
| | - Stuart S Martin
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States of America
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore Street, Bressler Bldg. Rm 10-29, Baltimore, MD 21201, United States of America
| |
Collapse
|
45
|
Ilton M, DiMaria C, Dalnoki-Veress K. Direct Measurement of the Critical Pore Size in a Model Membrane. PHYSICAL REVIEW LETTERS 2016; 117:257801. [PMID: 28036217 DOI: 10.1103/physrevlett.117.257801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Indexed: 05/27/2023]
Abstract
We study pore nucleation in a model membrane system, a freestanding polymer film. Nucleated pores smaller than a critical size close, while pores larger than the critical size grow. Holes of varying size were purposefully prepared in liquid polymer films, and their evolution in time was monitored using optical and atomic force microscopy to extract a critical radius. The critical radius scales linearly with film thickness for a homopolymer film. The results agree with a simple model which takes into account the energy cost due to surface area at the edge of the pore. The energy cost at the edge of the pore is experimentally varied by using a lamellar-forming diblock copolymer membrane. The underlying molecular architecture causes increased frustration at the pore edge resulting in an enhanced cost of pore formation.
Collapse
Affiliation(s)
- Mark Ilton
- Department of Physics & Astronomy, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Christian DiMaria
- Department of Physics & Astronomy, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Kari Dalnoki-Veress
- Department of Physics & Astronomy, McMaster University, Hamilton, Ontario L8S 4M1, Canada
- Laboratoire de Physico-Chimie Théorique, UMR CNRS Gulliver 7083, ESPCI ParisTech, PSL Research University, 75005 Paris, France
| |
Collapse
|
46
|
Mann BJ, Balchand SK, Wadsworth P. Regulation of Kif15 localization and motility by the C-terminus of TPX2 and microtubule dynamics. Mol Biol Cell 2016; 28:65-75. [PMID: 27852894 PMCID: PMC5221630 DOI: 10.1091/mbc.e16-06-0476] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
Mitotic motor proteins generate force to establish and maintain spindle bipolarity, but how they are temporally and spatially regulated in vivo is unclear. Prior work demonstrated that a microtubule-associated protein, TPX2, targets kinesin-5 and kinesin-12 motors to spindle microtubules. The C-terminal domain of TPX2 contributes to the localization and motility of the kinesin-5, Eg5, but it is not known whether this domain regulates kinesin-12, Kif15. We found that the C-terminal domain of TPX2 contributes to the localization of Kif15 to spindle microtubules in cells and suppresses motor walking in vitro. Kif15 and Eg5 are partially redundant motors, and overexpressed Kif15 can drive spindle formation in the absence of Eg5 activity. Kif15-dependent bipolar spindle formation in vivo requires the C-terminal domain of TPX2. In the spindle, fluorescent puncta of GFP-Kif15 move toward the equatorial region at a rate equivalent to microtubule growth. Reduction of microtubule growth with paclitaxel suppresses GFP-Kif15 motility, demonstrating that dynamic microtubules contribute to Kif15 behavior. Our results show that the C-terminal region of TPX2 regulates Kif15 in vitro, contributes to motor localization in cells, and is required for Kif15 force generation in vivo and further reveal that dynamic microtubules contribute to Kif15 behavior in vivo.
Collapse
Affiliation(s)
- Barbara J Mann
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Sai K Balchand
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Patricia Wadsworth
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
47
|
Fang CT, Kuo HH, Pan TS, Yu FC, Yih LH. HSP70 regulates the function of mitotic centrosomes. Cell Mol Life Sci 2016; 73:3949-60. [PMID: 27137183 PMCID: PMC11108311 DOI: 10.1007/s00018-016-2236-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/16/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
To establish a functional bipolar mitotic spindle, the centrosome expands and matures, acquiring enhanced activities for microtubule (MT) nucleation and assembly at the onset of mitosis. However, the regulatory mechanisms of centrosome maturation and MT assembly from the matured centrosome are largely unknown. In this study, we showed that heat shock protein (HSP) 70 considerably accumulates at the mitotic centrosome during prometaphase to metaphase and is required for bipolar spindle assembly. Inhibition or depletion of HSP70 impaired the function of mitotic centrosome and disrupted MT nucleation and polymerization from the spindle pole, and may thus result in formation of abnormal mitotic spindles. In addition, HSP70 may associate with NEDD1 and γ-tubulin, two pericentriolar material (PCM) components essential for centrosome maturation and MT nucleation. Loss of HSP70 function disrupted the interaction between NEDD1 and γ-tubulin, and reduced their accumulation at the mitotic centrosome. Our results thus demonstrate a role for HSP70 in regulating centrosome integrity during mitosis, and indicate that HSP70 is required for the maintenance of a functional mitotic centrosome that supports the assembly of a bipolar mitotic spindle.
Collapse
Affiliation(s)
- Chieh-Ting Fang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Hsiao-Hui Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Tiffany S Pan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Fu-Chi Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Ling-Huei Yih
- Department of Life Science, National Taiwan University, Taipei, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
48
|
Letort G, Nedelec F, Blanchoin L, Théry M. Centrosome centering and decentering by microtubule network rearrangement. Mol Biol Cell 2016; 27:2833-43. [PMID: 27440925 PMCID: PMC5025270 DOI: 10.1091/mbc.e16-06-0395] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/11/2016] [Indexed: 11/11/2022] Open
Abstract
Numerical simulations are used to investigate the role of microtubule network architecture in centrosome positioning. Microtubule gliding along cell edges and pivoting around the centrosome are key regulators of the orientation of pushing forces, the magnitude of which depends on the number, dynamics, and stiffness of microtubules. The centrosome is positioned at the cell center by pushing and pulling forces transmitted by microtubules (MTs). Centrosome decentering is often considered to result from asymmetric, cortical pulling forces exerted in particular by molecular motors on MTs and controlled by external cues affecting the cell cortex locally. Here we used numerical simulations to investigate the possibility that it could equally result from the redistribution of pushing forces due to a reorientation of MTs. We first showed that MT gliding along cell edges and pivoting around the centrosome regulate MT rearrangement and thereby direct the spatial distribution of pushing forces, whereas the number, dynamics, and stiffness of MTs determine the magnitude of these forces. By modulating these parameters, we identified different regimes, involving both pushing and pulling forces, characterized by robust centrosome centering, robust off-centering, or “reactive” positioning. In the last-named conditions, weak asymmetric cues can induce a misbalance of pushing and pulling forces, resulting in an abrupt transition from a centered to an off-centered position. Taken together, these results point to the central role played by the configuration of the MTs on the distribution of pushing forces that position the centrosome. We suggest that asymmetric external cues should not be seen as direct driver of centrosome decentering and cell polarization but instead as inducers of an effective reorganization of the MT network, fostering centrosome motion to the cell periphery.
Collapse
Affiliation(s)
- Gaëlle Letort
- CytoMorpho Lab, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, 38054 Grenoble, France
| | - Francois Nedelec
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Laurent Blanchoin
- CytoMorpho Lab, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, 38054 Grenoble, France CytoMorpho Lab, Hopital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/Université Paris Diderot, 75010 Paris, France
| | - Manuel Théry
- CytoMorpho Lab, Biosciences and Biotechnology Institute of Grenoble, UMR5168, CEA/INRA/CNRS/Université Grenoble-Alpes, 38054 Grenoble, France CytoMorpho Lab, Hopital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/Université Paris Diderot, 75010 Paris, France
| |
Collapse
|
49
|
Vora SM, Phillips BT. The benefits of local depletion: The centrosome as a scaffold for ubiquitin-proteasome-mediated degradation. Cell Cycle 2016; 15:2124-2134. [PMID: 27294844 DOI: 10.1080/15384101.2016.1196306] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The centrosome is the major microtubule-organizing center in animal cells but is dispensable for proper microtubule spindle formation in many biological contexts and is thus thought to fulfill additional functions. Recent observations suggest that the centrosome acts as a scaffold for proteasomal degradation in the cell to regulate a variety of biological processes including cell fate acquisition, cell cycle control, stress response, and cell morphogenesis. Here, we review the body of studies indicating a role for the centrosome in promoting proteasomal degradation of ubiquitin-proteasome substrates and explore the functional relevance of this system in different biological contexts. We discuss a potential role for the centrosome in coordinating local degradation of proteasomal substrates, allowing cells to achieve stringent spatiotemporal control over various signaling processes.
Collapse
Affiliation(s)
- Setu M Vora
- a Department of Biological Sciences, University of Iowa , Iowa City , IA , USA
| | - Bryan T Phillips
- a Department of Biological Sciences, University of Iowa , Iowa City , IA , USA
| |
Collapse
|
50
|
Muroyama A, Seldin L, Lechler T. Divergent regulation of functionally distinct γ-tubulin complexes during differentiation. J Cell Biol 2016; 213:679-92. [PMID: 27298324 PMCID: PMC4915192 DOI: 10.1083/jcb.201601099] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/29/2016] [Indexed: 11/22/2022] Open
Abstract
Differentiation induces the formation of noncentrosomal microtubule arrays in diverse tissues. The formation of these arrays requires loss of microtubule-organizing activity (MTOC) at the centrosome, but the mechanisms regulating this transition remain largely unexplored. Here, we use the robust loss of centrosomal MTOC activity in the epidermis to identify two pools of γ-tubulin that are biochemically and functionally distinct and differentially regulated. Nucleation-competent CDK5RAP2-γ-tubulin complexes were maintained at centrosomes upon initial epidermal differentiation. In contrast, Nedd1-γ-tubulin complexes did not promote nucleation but were required for anchoring of microtubules, a previously uncharacterized activity for this complex. Cell cycle exit specifically triggered loss of Nedd1-γ-tubulin complexes, providing a mechanistic link connecting MTOC activity and differentiation. Collectively, our studies demonstrate that distinct γ-tubulin complexes regulate different microtubule behaviors at the centrosome and show that differential regulation of these complexes drives loss of centrosomal MTOC activity.
Collapse
Affiliation(s)
- Andrew Muroyama
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710 Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| | - Lindsey Seldin
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710 Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| | - Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710 Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|