1
|
Chew YM, Cross RA. Structural switching of tubulin in the microtubule lattice. Biochem Soc Trans 2025; 53:BST20240360. [PMID: 39910801 DOI: 10.1042/bst20240360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Microtubule (MT) dynamic instability, a cycle of growth, catastrophe, shrinkage and rescue, is driven by the switching of tubulin between two structural states, one stabilised by GTP and the other by GDP. Recent work has uncovered the ancient origins of this structural switch and revealed further fundamental elements of microtubule dynamic instability, whereby switching can be brought about by a range of allosteric effectors, propagate deep within the lattice of assembled MTs, and profoundly affect MT function. Here, we review evidence for structural switching within the MT lattice and discuss current ideas about its mechanisms.
Collapse
Affiliation(s)
- Yean-Ming Chew
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| | - Robert A Cross
- Centre for Mechanochemical Cell Biology, University of Warwick, Warwick Medical School, Coventry CV4 7LA, U.K
| |
Collapse
|
2
|
Kumar P, Khan R, Singh BN, Kumari A, Rai A, Singh AK, Prakash A, Ray S. Hydroxyethylamine based analog targets microtubule assembly: an in silico study for anti-cancerous drug development. Sci Rep 2024; 14:31381. [PMID: 39732970 PMCID: PMC11682412 DOI: 10.1038/s41598-024-82823-8] [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: 08/27/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Microtubules are dynamic cytoskeletal structures essential for cell architecture, cellular transport, cell motility, and cell division. Due to their dynamic nature, known as dynamic instability, microtubules can spontaneously switch between phases of growth and shortening. Disruptions in microtubule functions have been implicated in several diseases, including cancer, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and birth defects. The role of microtubules during various phases of the cell cycle, particularly in cell division, makes them attractive targets for drug development against cancer. Several successful drugs currently on the market are designed to target microtubules. However, the presence of cellular toxicity and the development of multidrug resistance necessitate the search for new microtubule-targeting drugs.Here, a library of 106 biologically active compounds were screened to identify potent microtubule assembly inhibitors. Out of all the screened compounds, the hydroxyethylamine (HEA) analogues are found to be the best hit.We identified three inhibitors, BKS3031A, BKS3045A and BKS3046A, that bind at the same site as the well-known microtubule targeting agent colchicine. These inhibitors were simulated for 100 ns with tubulin complexes, and the results indicated that they remain stable within the binding pocket of α-β tubulin complexes. In addition, we estimated the binding free energy of BKS3031A, BKS3045A and BKS3046A by using molecular mechanics generalized Born surface area (MM-GBSA) calculations, and it was found to be -32.67 ± 6.01, -21.77 ± 5.12 and - 22.92 ± 5.09 kcal/mol, respectively. Our findings suggest that these novel inhibitors have potential to bind and perturb the microtubule network, positioning them as promising microtubule-targeting agents.
Collapse
Affiliation(s)
- Pawan Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Rajni Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, 844102, India
| | - Basant Narain Singh
- Department of Botany, Pandit Deendayal Upadhyaya Shekhawati University, Sikar, Nawalgarh Road, Katrathal, Rajasthan, 332024, India
| | - Anisha Kumari
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India
| | - Ankit Rai
- Department of Medical Biotechnology, Gujrat Biotechnology University, Gandhinagar, 382355, Gujarat, India.
| | - Anil Kumar Singh
- Department of Chemistry, Mahatma Gandhi Central University, Motihari, 845401, India.
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India.
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India.
| |
Collapse
|
3
|
Pantaleone LC, Calicchia E, Martinelli J, Stuart MCA, Lopatina YY, Browne WR, Portale G, Tych KM, Kudernac T. Exerting pulling forces in fluids by directional disassembly of microcrystalline fibres. NATURE NANOTECHNOLOGY 2024; 19:1507-1513. [PMID: 39075290 PMCID: PMC11486658 DOI: 10.1038/s41565-024-01742-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/02/2024] [Indexed: 07/31/2024]
Abstract
Biomolecular polymerization motors are biochemical systems that use supramolecular (de-)polymerization to convert chemical potential into useful mechanical work. With the intent to explore new chemomechanical transduction strategies, here we show a synthetic molecular system that can generate forces via the controlled disassembly of self-organized molecules in a crystal lattice, as they are freely suspended in a fluid. An amphiphilic monomer self-assembles into rigid, high-aspect-ratio microcrystalline fibres. The assembly process is regulated by a coumarin-based pH switching motif. The microfibre crystal morphology determines the monomer reactivity at the interface, resulting in anisotropic etching. This effect exerts a directional pulling force on microscopic beads adsorbed on the crystal surface through weak multivalent interactions. We use optical-tweezers-based force spectroscopy to extract mechanistic insights into this process, quantifying a stall force of 2.3 pN (±0.1 pN) exerted by the ratcheting mechanism produced by the disassembly of the microfibres.
Collapse
Affiliation(s)
- L C Pantaleone
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - E Calicchia
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, Netherlands
| | - J Martinelli
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - M C A Stuart
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Y Y Lopatina
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
- Institute of Physics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - W R Browne
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - G Portale
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, Netherlands
| | - K M Tych
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands.
| | - T Kudernac
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
4
|
Sanchez-Fernandez A, Insua I, Montenegro J. Supramolecular fibrillation in coacervates and other confined systems towards biomimetic function. Commun Chem 2024; 7:223. [PMID: 39349583 PMCID: PMC11442845 DOI: 10.1038/s42004-024-01308-x] [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: 12/30/2023] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
As in natural cytoskeletons, the cooperative assembly of fibrillar networks can be hosted inside compartments to engineer biomimetic functions, such as mechanical actuation, transport, and reaction templating. Coacervates impose an optimal liquid-liquid phase separation within the aqueous continuum, functioning as membrane-less compartments that can organise such self-assembling processes as well as the exchange of information with their environment. Furthermore, biological fibrillation can often be controlled or assisted by intracellular compartments. Thus, the reconstitution of analogues of natural filaments in simplified artificial compartments, such as coacervates, offer a suitable model to unravel, mimic, and potentially exploit cellular functions. This perspective summarises the latest developments towards assembling fibrillar networks under confinement inside coacervates and related compartments, including a selection of examples ranging from biological to fully synthetic monomers. Comparative analysis between coacervates, lipid vesicles, and droplet emulsions showcases the interplay between supramolecular fibres and the boundaries of the corresponding compartment. Combining inspiration from natural systems and the custom properties of tailored synthetic fibrillators, rational monomer and compartment design will contribute towards engineering increasingly complex and more realistic artificial protocells.
Collapse
Affiliation(s)
- Adrian Sanchez-Fernandez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Departamento de Enxeñaría Química, Universidade de Santaigo de Compostela, Santiago de Compostela, Spain
| | - Ignacio Insua
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Departamento de Farmacoloxía, Farmacia e Tecnoloxía Farmacéutica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
- Departamento de Química Orgánica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
5
|
Kreis NN, Moon HH, Wordeman L, Louwen F, Solbach C, Yuan J, Ritter A. KIF2C/MCAK a prognostic biomarker and its oncogenic potential in malignant progression, and prognosis of cancer patients: a systematic review and meta-analysis as biomarker. Crit Rev Clin Lab Sci 2024; 61:404-434. [PMID: 38344808 PMCID: PMC11815995 DOI: 10.1080/10408363.2024.2309933] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 03/24/2024]
Abstract
KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Ha Hyung Moon
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Frank Louwen
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
6
|
Aspenström P. Miro GTPases at the Crossroads of Cytoskeletal Dynamics and Mitochondrial Trafficking. Cells 2024; 13:647. [PMID: 38607086 PMCID: PMC11012113 DOI: 10.3390/cells13070647] [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: 03/07/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
Miro GTPases are key components in the machinery responsible for transporting mitochondria and peroxisomes along microtubules, and also play important roles in regulating calcium homeostasis and organizing contact sites between mitochondria and the endoplasmic reticulum. Moreover, Miro GTPases have been shown to interact with proteins that actively regulate cytoskeletal organization and dynamics, suggesting that these GTPases participate in organizing cytoskeletal functions and organelle transport. Derailed mitochondrial transport is associated with neuropathological conditions such as Parkinson's and Alzheimer's diseases. This review explores our recent understanding of the diverse roles of Miro GTPases under cytoskeletal control, both under normal conditions and during the course of human diseases such as neuropathological disorders.
Collapse
Affiliation(s)
- Pontus Aspenström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology (IGP), Uppsala University, SE 751 85 Uppsala, Sweden
| |
Collapse
|
7
|
Laporte D, Massoni-Laporte A, Lefranc C, Dompierre J, Mauboules D, Nsamba ET, Royou A, Gal L, Schuldiner M, Gupta ML, Sagot I. A stable microtubule bundle formed through an orchestrated multistep process controls quiescence exit. eLife 2024; 12:RP89958. [PMID: 38527106 PMCID: PMC10963028 DOI: 10.7554/elife.89958] [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: 03/27/2024] Open
Abstract
Cells fine-tune microtubule assembly in both space and time to give rise to distinct edifices with specific cellular functions. In proliferating cells, microtubules are highly dynamics, and proliferation cessation often leads to their stabilization. One of the most stable microtubule structures identified to date is the nuclear bundle assembled in quiescent yeast. In this article, we characterize the original multistep process driving the assembly of this structure. This Aurora B-dependent mechanism follows a precise temporality that relies on the sequential actions of kinesin-14, kinesin-5, and involves both microtubule-kinetochore and kinetochore-kinetochore interactions. Upon quiescence exit, the microtubule bundle is disassembled via a cooperative process involving kinesin-8 and its full disassembly is required prior to cells re-entry into proliferation. Overall, our study provides the first description, at the molecular scale, of the entire life cycle of a stable microtubule structure in vivo and sheds light on its physiological function.
Collapse
Affiliation(s)
| | | | | | | | | | - Emmanuel T Nsamba
- Genetics, Development, and Cell Biology, Iowa State UniversityAmesUnited States
| | - Anne Royou
- Univ. Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance
| | - Lihi Gal
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Mohan L Gupta
- Genetics, Development, and Cell Biology, Iowa State UniversityAmesUnited States
| | | |
Collapse
|
8
|
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
|
9
|
McCormick LA, Cleary JM, Hancock WO, Rice LM. Interface-acting nucleotide controls polymerization dynamics at microtubule plus- and minus-ends. eLife 2024; 12:RP89231. [PMID: 38180336 PMCID: PMC10945504 DOI: 10.7554/elife.89231] [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: 01/06/2024] Open
Abstract
GTP-tubulin is preferentially incorporated at growing microtubule ends, but the biochemical mechanism by which the bound nucleotide regulates the strength of tubulin:tubulin interactions is debated. The 'self-acting' (cis) model posits that the nucleotide (GTP or GDP) bound to a particular tubulin dictates how strongly that tubulin interacts, whereas the 'interface-acting' (trans) model posits that the nucleotide at the interface of two tubulin dimers is the determinant. We identified a testable difference between these mechanisms using mixed nucleotide simulations of microtubule elongation: with a self-acting nucleotide, plus- and minus-end growth rates decreased in the same proportion to the amount of GDP-tubulin, whereas with interface-acting nucleotide, plus-end growth rates decreased disproportionately. We then experimentally measured plus- and minus-end elongation rates in mixed nucleotides and observed a disproportionate effect of GDP-tubulin on plus-end growth rates. Simulations of microtubule growth were consistent with GDP-tubulin binding at and 'poisoning' plus-ends but not at minus-ends. Quantitative agreement between simulations and experiments required nucleotide exchange at terminal plus-end subunits to mitigate the poisoning effect of GDP-tubulin there. Our results indicate that the interfacial nucleotide determines tubulin:tubulin interaction strength, thereby settling a longstanding debate over the effect of nucleotide state on microtubule dynamics.
Collapse
Affiliation(s)
- Lauren A McCormick
- Department of Biophysics and Biochemistry, the University of Texas Southwestern Medical CenterDallasUnited States
| | - Joseph M Cleary
- Department of Biomedical Engineering, Pennsylvania State UniversityState CollegeUnited States
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State UniversityState CollegeUnited States
| | - Luke M Rice
- Department of Biophysics and Biochemistry, the University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
10
|
Leeds BK, Kostello KF, Liu YY, Nelson CR, Biggins S, Asbury CL. Mechanical coupling coordinates microtubule growth. eLife 2023; 12:RP89467. [PMID: 38150374 PMCID: PMC10752587 DOI: 10.7554/elife.89467] [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: 12/29/2023] Open
Abstract
During mitosis, kinetochore-attached microtubules form bundles (k-fibers) in which many filaments grow and shorten in near-perfect unison to align and segregate each chromosome. However, individual microtubules grow at intrinsically variable rates, which must be tightly regulated for a k-fiber to behave as a single unit. This exquisite coordination might be achieved biochemically, via selective binding of polymerases and depolymerases, or mechanically, because k-fiber microtubules are coupled through a shared load that influences their growth. Here, we use a novel dual laser trap assay to show that microtubule pairs growing in vitro are coordinated by mechanical coupling. Kinetic analyses show that microtubule growth is interrupted by stochastic, force-dependent pauses and indicate persistent heterogeneity in growth speed during non-pauses. A simple model incorporating both force-dependent pausing and persistent growth speed heterogeneity explains the measured coordination of microtubule pairs without any free fit parameters. Our findings illustrate how microtubule growth may be synchronized during mitosis and provide a basis for modeling k-fiber bundles with three or more microtubules, as found in many eukaryotes.
Collapse
Affiliation(s)
- Bonnibelle K Leeds
- Department of Physiology & Biophysics, University of WashingtonSeattleUnited States
| | - Katelyn F Kostello
- Department of Physiology & Biophysics, University of WashingtonSeattleUnited States
| | - Yuna Y Liu
- Department of Physiology & Biophysics, University of WashingtonSeattleUnited States
| | - Christian R Nelson
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Sue Biggins
- Basic Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Charles L Asbury
- Department of Physiology & Biophysics, University of WashingtonSeattleUnited States
| |
Collapse
|
11
|
Romeiro Motta M, Biswas S, Schaedel L. Beyond uniformity: Exploring the heterogeneous and dynamic nature of the microtubule lattice. Eur J Cell Biol 2023; 102:151370. [PMID: 37922811 DOI: 10.1016/j.ejcb.2023.151370] [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: 08/14/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
A fair amount of research on microtubules since their discovery in 1963 has focused on their dynamic tips. In contrast, the microtubule lattice was long believed to be highly regular and static, and consequently received far less attention. Yet, as it turned out, the microtubule lattice is neither as regular, nor as static as previously believed: structural studies uncovered the remarkable wealth of different conformations the lattice can accommodate. In the last decade, the microtubule lattice was shown to be labile and to spontaneously undergo renovation, a phenomenon that is intimately linked to structural defects and was called "microtubule self-repair". Following this breakthrough discovery, further recent research provided a deeper understanding of the lattice self-repair mechanism, which we review here. Instrumental to these discoveries were in vitro microtubule reconstitution assays, in which microtubules are grown from the minimal components required for their dynamics. In this review, we propose a shift from the term "lattice self-repair" to "lattice dynamics", since this phenomenon is an inherent property of microtubules and can happen without microtubule damage. We focus on how in vitro microtubule reconstitution assays helped us learn (1) which types of structural variations microtubules display, (2) how these structural variations influence lattice dynamics and microtubule damage caused by mechanical stress, (3) how lattice dynamics impact tip dynamics, and (4) how microtubule-associated proteins (MAPs) can play a role in structuring the lattice. Finally, we discuss the unanswered questions about lattice dynamics and how technical advances will help us tackle these questions.
Collapse
Affiliation(s)
- Mariana Romeiro Motta
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany; Laboratoire Reproduction et Développement des Plantes, Université de Lyon, École normale supérieure de Lyon, Lyon 69364, France
| | - Subham Biswas
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany
| | - Laura Schaedel
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany.
| |
Collapse
|
12
|
Leeds BK, Kostello KF, Liu YY, Nelson CR, Biggins S, Asbury CL. Mechanical coupling coordinates microtubule growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547092. [PMID: 37905093 PMCID: PMC10614740 DOI: 10.1101/2023.06.29.547092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
During mitosis, kinetochore-attached microtubules form bundles (k-fibers) in which many filaments grow and shorten in near-perfect unison to align and segregate each chromosome. However, individual microtubules grow at intrinsically variable rates, which must be tightly regulated for a k-fiber to behave as a single unit. This exquisite coordination might be achieved biochemically, via selective binding of polymerases and depolymerases, or mechanically, because k-fiber microtubules are coupled through a shared load that influences their growth. Here, we use a novel dual laser trap assay to show that microtubule pairs growing in vitro are coordinated by mechanical coupling. Kinetic analyses show that microtubule growth is interrupted by stochastic, force-dependent pauses and indicate persistent heterogeneity in growth speed during non-pauses. A simple model incorporating both force-dependent pausing and persistent growth speed heterogeneity explains the measured coordination of microtubule pairs without any free fit parameters. Our findings illustrate how microtubule growth may be synchronized during mitosis and provide a basis for modeling k-fiber bundles with three or more microtubules, as found in many eukaryotes.
Collapse
Affiliation(s)
- Bonnibelle K. Leeds
- Physiology & Biophysics Department, University of Washington School of Medicine, Seattle WA, USA
| | - Katelyn F. Kostello
- Physiology & Biophysics Department, University of Washington School of Medicine, Seattle WA, USA
| | - Yuna Y. Liu
- Physiology & Biophysics Department, University of Washington School of Medicine, Seattle WA, USA
| | | | | | - Charles L. Asbury
- Physiology & Biophysics Department, University of Washington School of Medicine, Seattle WA, USA
| |
Collapse
|
13
|
McCormick LA, Cleary JM, Hancock WO, Rice LM. Interface-acting nucleotide controls polymerization dynamics at microtubule plus- and minus-ends. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539131. [PMID: 37205370 PMCID: PMC10187237 DOI: 10.1101/2023.05.03.539131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
GTP-tubulin is preferentially incorporated at growing microtubule ends, but the biochemical mechanism by which the bound nucleotide regulates the strength of tubulin:tubulin interactions is debated. The 'self-acting' (cis) model posits that the nucleotide (GTP or GDP) bound to a particular tubulin dictates how strongly that tubulin interacts, whereas the 'interface-acting' (trans) model posits that the nucleotide at the interface of two tubulin dimers is the determinant. We identified a testable difference between these mechanisms using mixed nucleotide simulations of microtubule elongation: with self-acting nucleotide, plus- and minus-end growth rates decreased in the same proportion to the amount of GDP-tubulin, whereas with interface-acting nucleotide, plus-end growth rates decreased disproportionately. We then experimentally measured plus- and minus-end elongation rates in mixed nucleotides and observed a disproportionate effect of GDP-tubulin on plus-end growth rates. Simulations of microtubule growth were consistent with GDP-tubulin binding at and 'poisoning' plus-ends but not at minus-ends. Quantitative agreement between simulations and experiments required nucleotide exchange at terminal plus-end subunits to mitigate the poisoning effect of GDP-tubulin there. Our results indicate that the interfacial nucleotide determines tubulin:tubulin interaction strength, thereby settling a longstanding debate over the effect of nucleotide state on microtubule dynamics.
Collapse
Affiliation(s)
- Lauren A McCormick
- Department of Biophysics and Biochemistry, the University of Texas Southwestern Medical Center, Dallas, TX
| | - Joseph M Cleary
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA
| | - Luke M Rice
- Department of Biophysics and Biochemistry, the University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
14
|
Amiri A, Dietz C, Rapp A, Cardoso MC, Stark RW. The cyto-linker and scaffolding protein "plectin" mis-localization leads to softening of cancer cells. NANOSCALE 2023; 15:15008-15026. [PMID: 37668423 DOI: 10.1039/d3nr02226a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Discovering tools to prevent cancer progression requires understanding the fundamental differences between normal and cancer cells. More than a decade ago, atomic force microscopy (AFM) revealed cancer cells' softer body compared to their healthy counterparts. Here, we investigated the mechanism underlying the softening of cancerous cells in comparison with their healthy counterparts based on AFM high resolution stiffness tomography and 3D confocal microscopy. We showed microtubules (MTs) network in invasive ductal carcinoma cell cytoskeleton is basally located and segmented for around 400 nm from the cell periphery. Additionally, the cytoskeleton scaffolding protein plectin exhibits a mis-localization from the cytoplasm to the surface of cells in the carcinoma which justifies the dissociation of the MT network from the cell's cortex. Furthermore, the assessment of MTs' persistence length using a worm-like-chain (WLC) model in high resolution AFM images showed lower persistence length of the single MTs in ductal carcinoma compared to that in the normal state. Overall, these tuned mechanics support the invasive cells to ascertain more flexibility under compressive forces in small deformations. These data provide new insights into the structural origins of cancer aids in progression.
Collapse
Affiliation(s)
- Anahid Amiri
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Christian Dietz
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Alexander Rapp
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - M Cristina Cardoso
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Robert W Stark
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| |
Collapse
|
15
|
Chew YM, Cross RA. Taxol acts differently on different tubulin isotypes. Commun Biol 2023; 6:946. [PMID: 37717119 PMCID: PMC10505170 DOI: 10.1038/s42003-023-05306-y] [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: 03/20/2023] [Accepted: 08/31/2023] [Indexed: 09/18/2023] Open
Abstract
Taxol is a small molecule effector that allosterically locks tubulin into the microtubule lattice. We show here that taxol has different effects on different single-isotype microtubule lattices. Using in vitro reconstitution, we demonstrate that single-isotype α1β4 GDP-tubulin lattices are stabilised and expanded by 10 µM taxol, as reported by accelerated microtubule gliding in kinesin motility assays, whereas single-isotype α1β3 GDP-tubulin lattices are stabilised but not expanded. This isotype-specific action of taxol drives gliding of segmented-isotype GDP-taxol microtubules along convoluted, sinusoidal paths, because their expanded α1β4 segments try to glide faster than their compacted α1β3 segments. In GMPCPP, single-isotype α1β3 and α1β4 lattices both show accelerated gliding, indicating that both can in principle be driven to expand. We therefore propose that taxol-induced lattice expansion requires a higher taxol occupancy than taxol-induced stabilisation, and that higher taxol occupancies are accessible to α1β4 but not α1β3 single-isotype lattices.
Collapse
Affiliation(s)
- Yean Ming Chew
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, CV4 7LA, UK
| | - Robert A Cross
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Coventry, CV4 7LA, UK.
| |
Collapse
|
16
|
Ráduly Z, Szabó L, Dienes B, Szentesi P, Bana ÁV, Hajdú T, Kókai E, Hegedűs C, Csernoch L, Gönczi M. Migration of Myogenic Cells Is Highly Influenced by Cytoskeletal Septin7. Cells 2023; 12:1825. [PMID: 37508490 PMCID: PMC10378681 DOI: 10.3390/cells12141825] [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: 05/30/2023] [Revised: 06/19/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Septin7 as a unique member of the GTP binding protein family, is widely expressed in the eukaryotic cells and considered to be essential in the formation of hetero-oligomeric septin complexes. As a cytoskeletal component, Septin7 is involved in many important cellular processes. However, its contribution in striated muscle physiology is poorly described. In skeletal muscle, a highly orchestrated process of migration is crucial in the development of functional fibers and in regeneration. Here, we describe the pronounced appearance of Septin7 filaments and a continuous change of Septin7 protein architecture during the migration of myogenic cells. In Septin7 knockdown C2C12 cultures, the basic parameters of migration are significantly different, and the intracellular calcium concentration change in migrating cells are lower compared to that of scrambled cultures. Using a plant cytokinin, forchlorfenuron, to dampen septin dynamics, the altered behavior of the migrating cells is described, where Septin7-depleted cells are more resistant to the treatment. These results indicate the functional relevance of Septin7 in the migration of myoblasts, implying its contribution to muscle myogenesis and regeneration.
Collapse
Affiliation(s)
- Zsolt Ráduly
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Szabó
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágnes Viktória Bana
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Endre Kókai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Prasad M, Hazra B, Mandal R, Das S, Tarafdar PK. ATP-Assisted Protocellular Membrane Formation with Ethanolamine-Based Amphiphiles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023. [PMID: 37421360 DOI: 10.1021/acs.langmuir.3c00600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Prebiotic membranes are one of the essential elements of the origin of life because they build compartments to keep genetic materials and metabolic machinery safe. Since modern cell membranes are made up of ethanolamine-based phospholipids, prebiotic membrane formation with ethanolamine-based amphiphiles and phosphates might act as a bridge between the prebiotic and contemporary eras. Here, we report the prebiotic synthesis of O-lauroyl ethanolamine (OLEA), O-lauroyl methyl ethanolamine (OLMEA), and O-lauroyl dimethylethanolamine (OLDMEA) under wet-dry cycles. Turbidimetric, NMR, DLS, fluorescence, microscopy, and glucose encapsulation studies highlighted that OLEA-ATP and OLMEA-ATP form protocellular membranes in a 3:1 ratio, where ATP acts as a template. OLDMEA with a dimethyl group did not form any membrane in the presence of ATP. ADP can also template OLEA to form vesicles in a 2:1 ratio, but the ADP-templated vesicles were smaller. This suggests the critical role of the phosphate backbone in controlling the curvature of supramolecular assembly. The mechanisms of hierarchical assembly and transient dissipative assembly are discussed based on templated-complex formation via electrostatic, hydrophobic, and H-bonding interactions. Our results suggest that N-methylethanolamine-based amphiphiles could be used to form prebiotic vesicles, but the superior H-bonding ability of the ethanolamine moiety likely provides an evolutionary advantage for stable protocell formation during the fluctuating environments of early earth.
Collapse
Affiliation(s)
- Mahesh Prasad
- Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Bibhas Hazra
- Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Raki Mandal
- Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subrata Das
- Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Pradip K Tarafdar
- Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
18
|
Shi K, Wang Y, Meng D, Zhang Y, Wang Y, An J. Cytosolic JNK-dependent microtubule reassembly protects Jurkat leukemia cells from selenite-induced apoptosis. J Trace Elem Med Biol 2023; 79:127214. [PMID: 37224747 DOI: 10.1016/j.jtemb.2023.127214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND Selenite at high dosage exhibits great potential in curing tumors. It has been shown that selenite inhibits tumor growth through regulation of microtubule dynamics, however, the exact underlying mechanisms remained to be fully elucidated. METHODS & RESULTS Western blots were carried out to evaluate expression level of different molecules. Our current study discovered that selenite induced microtubule disassembly, cell cycle arrest and finally resulted in apoptosis in Jurkat leukemia cells, while during this process disassembled tubulins were re-organized after long-term exposure to selenite. Furthermore, JNK was activated in the cytoplasm of selenite-treated Jurkat cells, and inhibition of JNK activity successfully prevented the process of microtubule re-assembly. Moreover, inactivation of JNK further enhanced selenite-induced cell cycle arrest and apoptosis. According to the results from cell counting-8 assay, blockage of microtubule re-assembly by colchicine further inhibited Jurkat cell viability after exposure to selenite. Experiments in a xenograft model also proved that selenite could alter JNK activity, destroy microtubule structure and inhibit cell division in vivo. Moreover, TP53, MAPT and YWHAZ were identified to be three most confident interactors that link JNK to microtubule assembly using PPIs analysis. CONCLUSION Our study indicated that cytosolic JNK-dependent microtubule re-organization took a protective function during selenite-induced apoptosis, while inhibition of this process would finally enhance the anti-tumor effect of selenite.
Collapse
Affiliation(s)
- Kejian Shi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China; State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medicine Sciences & School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, PR China
| | - Yang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Di Meng
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, PR China
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China.
| | - Ying Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, PR China.
| | - Jiajia An
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou 256603, PR China.
| |
Collapse
|
19
|
Verhey KJ, Ohi R. Causes, costs and consequences of kinesin motors communicating through the microtubule lattice. J Cell Sci 2023; 136:293511. [PMID: 36866642 PMCID: PMC10022682 DOI: 10.1242/jcs.260735] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Microtubules are critical for a variety of important functions in eukaryotic cells. During intracellular trafficking, molecular motor proteins of the kinesin superfamily drive the transport of cellular cargoes by stepping processively along the microtubule surface. Traditionally, the microtubule has been viewed as simply a track for kinesin motility. New work is challenging this classic view by showing that kinesin-1 and kinesin-4 proteins can induce conformational changes in tubulin subunits while they are stepping. These conformational changes appear to propagate along the microtubule such that the kinesins can work allosterically through the lattice to influence other proteins on the same track. Thus, the microtubule is a plastic medium through which motors and other microtubule-associated proteins (MAPs) can communicate. Furthermore, stepping kinesin-1 can damage the microtubule lattice. Damage can be repaired by the incorporation of new tubulin subunits, but too much damage leads to microtubule breakage and disassembly. Thus, the addition and loss of tubulin subunits are not restricted to the ends of the microtubule filament but rather, the lattice itself undergoes continuous repair and remodeling. This work leads to a new understanding of how kinesin motors and their microtubule tracks engage in allosteric interactions that are critical for normal cell physiology.
Collapse
Affiliation(s)
- Kristen J. Verhey
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Author for correspondence ()
| | - Ryoma Ohi
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Luchniak A, Kuo YW, McGuinness C, Sutradhar S, Orbach R, Mahamdeh M, Howard J. Dynamic microtubules slow down during their shrinkage phase. Biophys J 2023; 122:616-623. [PMID: 36659852 PMCID: PMC9989939 DOI: 10.1016/j.bpj.2023.01.020] [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: 07/22/2022] [Revised: 11/25/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Microtubules are dynamic polymers that undergo stochastic transitions between growing and shrinking phases. The structural and chemical properties of these phases remain poorly understood. The transition from growth to shrinkage, termed catastrophe, is not a first-order reaction but rather a multistep process whose frequency increases with the growth time: the microtubule ages as the older microtubule tip becomes more unstable. Aging shows that the growing phase is not a single state but comprises several substates of increasing instability. To investigate whether the shrinking phase is also multistate, we characterized the kinetics of microtubule shrinkage following catastrophe using an in vitro reconstitution assay with purified tubulins. We found that the shrinkage speed is highly variable across microtubules and that the shrinkage speed of individual microtubules slows down over time by as much as several fold. The shrinkage slowdown was observed in both fluorescently labeled and unlabeled microtubules as well as in microtubules polymerized from tubulin purified from different species, suggesting that the shrinkage slowdown is a general property of microtubules. These results indicate that microtubule shrinkage, like catastrophe, is time dependent and that the shrinking microtubule tip passes through a succession of states of increasing stability. We hypothesize that the shrinkage slowdown is due to destabilizing events that took place during growth, which led to multistep catastrophe. This suggests that the aging associated with growth is also manifested during shrinkage, with the older, more unstable growing tip being associated with a faster depolymerizing shrinking tip.
Collapse
Affiliation(s)
- Anna Luchniak
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Yin-Wei Kuo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Catherine McGuinness
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Ron Orbach
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Mohammed Mahamdeh
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut.
| |
Collapse
|
21
|
Gazzola M, Schaeffer A, Butler-Hallissey C, Friedl K, Vianay B, Gaillard J, Leterrier C, Blanchoin L, Théry M. Microtubules self-repair in living cells. Curr Biol 2023; 33:122-133.e4. [PMID: 36565699 DOI: 10.1016/j.cub.2022.11.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/21/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
Microtubule self-repair has been studied both in vitro and in vivo as an underlying mechanism of microtubule stability. The turnover of tubulin dimers along the microtubule has challenged the pre-existing dogma that only growing ends are dynamic. However, although there is clear evidence of tubulin incorporation into the shaft of polymerized microtubules in vitro, the possibility of such events occurring in living cells remains uncertain. In this study, we investigated this possibility by microinjecting purified tubulin dimers labeled with a red fluorophore into the cytoplasm of cells expressing GFP-tubulin. We observed the appearance of red dots along the pre-existing green microtubule within minutes. We found that the fluorescence intensities of these red dots were inversely correlated with the green signal, suggesting that the red dimers were incorporated into the microtubules and replaced the pre-existing green dimers. Lateral distance from the microtubule center was similar to that in incorporation sites and in growing ends. The saturation of the size and spatial frequency of incorporations as a function of injected tubulin concentration and post-injection delay suggested that the injected dimers incorporated into a finite number of damaged sites. By our low estimate, within a few minutes of the injections, free dimers incorporated into major repair sites every 70 μm of microtubules. Finally, we mapped the location of these sites in micropatterned cells and found that they were more concentrated in regions where the actin filament network was less dense and where microtubules exhibited greater lateral fluctuations.
Collapse
Affiliation(s)
- Morgan Gazzola
- University of Paris, INSERM, CEA, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, 10 Avenue Claude Vellefaux, 75010 Paris, France
| | - Alexandre Schaeffer
- University of Paris, INSERM, CEA, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, 10 Avenue Claude Vellefaux, 75010 Paris, France
| | - Ciarán Butler-Hallissey
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto Lab, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Karoline Friedl
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto Lab, 27 Boulevard Jean Moulin, 13385 Marseille, France; Abbelight, 191 Avenue Aristide Briand, 94230 Cachan, France
| | - Benoit Vianay
- University of Paris, INSERM, CEA, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, 10 Avenue Claude Vellefaux, 75010 Paris, France
| | - Jérémie Gaillard
- University of Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, 17 rue des Martyrs, 38054 Grenoble, France
| | - Christophe Leterrier
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto Lab, 27 Boulevard Jean Moulin, 13385 Marseille, France
| | - Laurent Blanchoin
- University of Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, 17 rue des Martyrs, 38054 Grenoble, France.
| | - Manuel Théry
- University of Paris, INSERM, CEA, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, 10 Avenue Claude Vellefaux, 75010 Paris, France.
| |
Collapse
|
22
|
Farmer VJ, Zanic M. Beyond the GTP-cap: Elucidating the molecular mechanisms of microtubule catastrophe. Bioessays 2023; 45:e2200081. [PMID: 36398561 PMCID: PMC10648283 DOI: 10.1002/bies.202200081] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022]
Abstract
Almost 40 years since the discovery of microtubule dynamic instability, the molecular mechanisms underlying microtubule dynamics remain an area of intense research interest. The "standard model" of microtubule dynamics implicates a "cap" of GTP-bound tubulin dimers at the growing microtubule end as the main determinant of microtubule stability. Loss of the GTP-cap leads to microtubule "catastrophe," a switch-like transition from microtubule growth to shrinkage. However, recent studies, using biochemical in vitro reconstitution, cryo-EM, and computational modeling approaches, challenge the simple GTP-cap model. Instead, a new perspective on the mechanisms of microtubule dynamics is emerging. In this view, highly dynamic transitions between different structural conformations of the growing microtubule end - which may or may not be directly linked to the nucleotide content at the microtubule end - ultimately drive microtubule catastrophe.
Collapse
Affiliation(s)
- Veronica J. Farmer
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biomolecular and Chemical Engineering, Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
23
|
Guyomar C, Bousquet C, Ku S, Heumann JM, Guilloux G, Gaillard N, Heichette C, Duchesne L, Steinmetz MO, Gibeaux R, Chrétien D. Changes in seam number and location induce holes within microtubules assembled from porcine brain tubulin and in Xenopus egg cytoplasmic extracts. eLife 2022; 11:e83021. [PMID: 36503602 PMCID: PMC9788831 DOI: 10.7554/elife.83021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Microtubules are tubes of about 25 nm in diameter that are critically involved in a variety of cellular functions, including motility, compartmentalization, and division. They are considered as pseudo-helical polymers whose constituent αβ-tubulin heterodimers share lateral homotypic interactions, except at one unique region called the seam. Here, we used a segmented sub-tomogram averaging strategy to reassess this paradigm and analyze the organization of the αβ-tubulin heterodimers in microtubules assembled from purified porcine brain tubulin in the presence of GTP and GMPCPP, and in Xenopus egg cytoplasmic extracts. We find that in almost all conditions, microtubules incorporate variable protofilament and/or tubulin subunit helical-start numbers, as well as variable numbers of seams. Strikingly, the seam number and location vary along individual microtubules, generating holes of one to a few subunits in size within their lattices. Together, our results reveal that the formation of mixed and discontinuous microtubule lattices is an intrinsic property of tubulin that requires the formation of unique lateral interactions without longitudinal ones. They further suggest that microtubule assembly is tightly regulated in a cytoplasmic environment.
Collapse
Affiliation(s)
- Charlotte Guyomar
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Clément Bousquet
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Siou Ku
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - John M Heumann
- Department of Molecular, Cellular and Developmental Biology, University of Colorado BoulderBoulderUnited States
| | - Gabriel Guilloux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Natacha Gaillard
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer InstituteVilligenSwitzerland
| | - Claire Heichette
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Laurence Duchesne
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer InstituteVilligenSwitzerland
- University of Basel, BiozentrumBaselSwitzerland
| | - Romain Gibeaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| | - Denis Chrétien
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000RennesFrance
| |
Collapse
|
24
|
Hoff KJ, Neumann AJ, Moore JK. The molecular biology of tubulinopathies: Understanding the impact of variants on tubulin structure and microtubule regulation. Front Cell Neurosci 2022; 16:1023267. [PMID: 36406756 PMCID: PMC9666403 DOI: 10.3389/fncel.2022.1023267] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/30/2022] [Indexed: 11/24/2022] Open
Abstract
Heterozygous, missense mutations in both α- and β-tubulin genes have been linked to an array of neurodevelopment disorders, commonly referred to as "tubulinopathies." To date, tubulinopathy mutations have been identified in three β-tubulin isotypes and one α-tubulin isotype. These mutations occur throughout the different genetic domains and protein structures of these tubulin isotypes, and the field is working to address how this molecular-level diversity results in different cellular and tissue-level pathologies. Studies from many groups have focused on elucidating the consequences of individual mutations; however, the field lacks comprehensive models for the molecular etiology of different types of tubulinopathies, presenting a major gap in diagnosis and treatment. This review highlights recent advances in understanding tubulin structural dynamics, the roles microtubule-associated proteins (MAPs) play in microtubule regulation, and how these are inextricably linked. We emphasize the value of investigating interactions between tubulin structures, microtubules, and MAPs to understand and predict the impact of tubulinopathy mutations at the cell and tissue levels. Microtubule regulation is multifaceted and provides a complex set of controls for generating a functional cytoskeleton at the right place and right time during neurodevelopment. Understanding how tubulinopathy mutations disrupt distinct subsets of those controls, and how that ultimately disrupts neurodevelopment, will be important for establishing mechanistic themes among tubulinopathies that may lead to insights in other neurodevelopment disorders and normal neurodevelopment.
Collapse
Affiliation(s)
| | | | - Jeffrey K. Moore
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
25
|
Foroutan A, Corazzari M, Grolla AA, Colombo G, Travelli C, Genazzani AA, Theeramunkong S, Galli U, Tron GC. Identification of novel aza-analogs of TN-16 as disrupters of microtubule dynamics through a multicomponent reaction. Eur J Med Chem 2022; 245:114895. [DOI: 10.1016/j.ejmech.2022.114895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/14/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
|
26
|
Zhang M, Sun L, Zhang Z, Shentu L, Zhang Y, Li Z, Zhang Y, Zhang Y. Alpha-lipoic acid supplementation restores the meiotic competency and fertilization capacity of porcine oocytes induced by arsenite. Front Cell Dev Biol 2022; 10:943757. [PMID: 36263016 PMCID: PMC9574060 DOI: 10.3389/fcell.2022.943757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Arsenite is known as a well-known endocrine disrupting chemicals, and reported to be associated with an increased incidence of negative health effects, including reproductive disorders and dysfunction of the endocrine system. However, it still lacks of the research regarding the beneficial effects of ALA on arsenite exposed oocytes, and the underlying mechanisms have not been determined. Here, we report that supplementation of alpha-lipoic acid (ALA), a strong antioxidant naturally present in all cells of the humans, is able to restore the declined meiotic competency and fertilization capacity of porcine oocytes induced by arsenite. Notably, ALA recovers the defective nuclear and cytoplasmic maturation of porcine oocytes caused by arsenite exposure, including the impaired spindle formation and actin polymerization, the defective mitochondrion integrity and cortical granules distribution. Also, ALA recovers the compromised sperm binding ability to maintain the fertilization potential of arsenite-exposed oocytes. Importantly, ALA suppresses the oxidative stress by reducing the levels of ROS and inhibits the occurrence of DNA damage along with apoptosis. Above all, we provide a new perspective for the application of ALA in effectively preventing the declined oocyte quality induced by environmental EDCs.
Collapse
|
27
|
Xie S, Li J, Sun S, Chen W, Cheng H, Song Y, Li Y, Liu M, Zhu X, Liang X, Zhou J. TUBright: A Peptide Probe for Imaging Microtubules. Anal Chem 2022; 94:11168-11174. [PMID: 35917443 DOI: 10.1021/acs.analchem.2c01285] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vitro assays using reconstituted microtubules have provided molecular insights into the principles of microtubule dynamics and the roles of microtubule-associated proteins. Emerging questions that further uncover the complexity in microtubule dynamics, especially those on tubulin isotypes and post-translational modifications, raise new technical challenges on how to visualize microtubules composed of tubulin purified from limited sources, primarily due to the low efficiency of the conventional tubulin labeling protocol. Here, we develop a peptide probe, termed TUBright, that labels in vitro reconstituted microtubules. TUBright, when coupled with different fluorescent dyes, provides flexible labeling of microtubules with a high signal-to-noise ratio. TUBright does not interfere with the dynamic behaviors of microtubules and microtubule-associated proteins. Therefore, TUBright is a useful tool for imaging microtubules, making it feasible to use tubulin from limited sources for answering many open questions on microtubule dynamics.
Collapse
Affiliation(s)
- Songbo Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Jingrui Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Shuang Sun
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Wei Chen
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haisu Cheng
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yinlong Song
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuyang Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Min Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin Liang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China.,State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
28
|
Liu GY, Chen S, Lee G, Shaiv K, Chen P, Cheng H, Hong S, Yang W, Huang S, Chang Y, Wang H, Kao C, Sun P, Chao M, Lee Y, Tang M, Lin Y. Precise control of microtubule disassembly in living cells. EMBO J 2022; 41:e110472. [PMID: 35686621 PMCID: PMC9340485 DOI: 10.15252/embj.2021110472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Microtubules tightly regulate various cellular activities. Our understanding of microtubules is largely based on experiments using microtubule-targeting agents, which, however, are insufficient to dissect the dynamic mechanisms of specific microtubule populations, due to their slow effects on the entire pool of microtubules. To overcome this technological limitation, we have used chemo and optogenetics to disassemble specific microtubule subtypes, including tyrosinated microtubules, primary cilia, mitotic spindles, and intercellular bridges, by rapidly recruiting engineered microtubule-cleaving enzymes onto target microtubules in a reversible manner. Using this approach, we show that acute microtubule disassembly swiftly halts vesicular trafficking and lysosomal dynamics. It also immediately triggers Golgi and ER reorganization and slows the fusion/fission of mitochondria without affecting mitochondrial membrane potential. In addition, cell rigidity is increased after microtubule disruption owing to increased contractile stress fibers. Microtubule disruption furthermore prevents cell division, but does not cause cell death during interphase. Overall, the reported tools facilitate detailed analysis of how microtubules precisely regulate cellular architecture and functions.
Collapse
Affiliation(s)
- Grace Y Liu
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shiau‐Chi Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Gang‐Hui Lee
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Kritika Shaiv
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Yu Chen
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsuan Cheng
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shi‐Rong Hong
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Wen‐Ting Yang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Shih‐Han Huang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ya‐Chu Chang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsien‐Chu Wang
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ching‐Lin Kao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Pin‐Chiao Sun
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Hong Chao
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Yian‐Ying Lee
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
| | - Ming‐Jer Tang
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Yu‐Chun Lin
- Institute of Molecular MedicineNational Tsing Hua UniversityHsinchuTaiwan
- Department of Medical ScienceNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
29
|
Kikuchi K, Sakamoto Y, Uezu A, Yamamoto H, Ishiguro KI, Shimamura K, Saito T, Hisanaga SI, Nakanishi H. Map7D2 and Map7D1 facilitate microtubule stabilization through distinct mechanisms in neuronal cells. Life Sci Alliance 2022; 5:5/8/e202201390. [PMID: 35470240 PMCID: PMC9039348 DOI: 10.26508/lsa.202201390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 12/05/2022] Open
Abstract
The microtubule-associated proteins Map7D2 and Map7D1, which belong to the MAP7 family, stabilize microtubules through distinct mechanisms for the control of cell motility and neurite outgrowth. Microtubule (MT) dynamics are modulated through the coordinated action of various MT-associated proteins (MAPs). However, the regulatory mechanisms underlying MT dynamics remain unclear. We show that the MAP7 family protein Map7D2 stabilizes MTs to control cell motility and neurite outgrowth. Map7D2 directly bound to MTs through its N-terminal half and stabilized MTs in vitro. Map7D2 localized prominently to the centrosome and partially on MTs in mouse N1-E115 neuronal cells, which expresses two of the four MAP7 family members, Map7D2 and Map7D1. Map7D2 loss decreased the resistance to the MT-destabilizing agent nocodazole without affecting acetylated/detyrosinated stable MTs, suggesting that Map7D2 stabilizes MTs via direct binding. In addition, Map7D2 loss increased the rate of random cell migration and neurite outgrowth, presumably by disturbing the balance between MT stabilization and destabilization. Map7D1 exhibited similar subcellular localization and gene knockdown phenotypes to Map7D2. However, in contrast to Map7D2, Map7D1 was required for the maintenance of acetylated stable MTs. Taken together, our data suggest that Map7D2 and Map7D1 facilitate MT stabilization through distinct mechanisms in cell motility and neurite outgrowth.
Collapse
Affiliation(s)
- Koji Kikuchi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhisa Sakamoto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akiyoshi Uezu
- Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Hideyuki Yamamoto
- Department of Biochemistry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Taro Saito
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Hiroyuki Nakanishi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
30
|
Cleary JM, Kim T, Cook ASI, McCormick LA, Hancock WO, Rice LM. Measurements and simulations of microtubule growth imply strong longitudinal interactions and reveal a role for GDP on the elongating end. eLife 2022; 11:75931. [PMID: 35420545 PMCID: PMC9064298 DOI: 10.7554/elife.75931] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubule polymerization dynamics result from the biochemical interactions of αβ-tubulin with the polymer end, but a quantitative understanding has been challenging to establish. We used interference reflection microscopy to make improved measurements of microtubule growth rates and growth fluctuations in the presence and absence of GTP hydrolysis. In the absence of GTP hydrolysis, microtubules grew steadily with very low fluctuations. These data were best described by a computational model implementing slow assembly kinetics, such that the rate of microtubule elongation is primarily limited by the rate of αβ-tubulin associations. With GTPase present, microtubules displayed substantially larger growth fluctuations than expected based on the no GTPase measurements. Our modeling showed that these larger fluctuations occurred because exposure of GDP-tubulin on the microtubule end transiently 'poisoned' growth, yielding a wider range of growth rates compared to GTP only conditions. Our experiments and modeling point to slow association kinetics (strong longitudinal interactions), such that drugs and regulatory proteins that alter microtubule dynamics could do so by modulating either the association or dissociation rate of tubulin from the microtubule tip. By causing slower growth, exposure of GDP tubulin at the growing microtubule end may be an important early event determining catastrophe.
Collapse
Affiliation(s)
- Joseph M Cleary
- Department of Biomedical Engineering, Pennsylvania State University, University Park, United States
| | - Tae Kim
- Departments of Biophysics and Biochemistry, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Annan S I Cook
- Department of Biomedical Engineering, Pennsylvania State University, University Park, United States
| | - Lauren A McCormick
- Departments of Biophysics and Biochemistry, The University of Texas Southwestern Medical Center, Dallas, United States
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State University, University Park, United States
| | - Luke M Rice
- Departments of Biophysics and Biochemistry, The University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
31
|
Huang L, Peng Y, Tao X, Ding X, Li R, Jiang Y, Zuo W. Microtubule Organization Is Essential for Maintaining Cellular Morphology and Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1623181. [PMID: 35295719 PMCID: PMC8920689 DOI: 10.1155/2022/1623181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/10/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microtubules (MTs) are highly dynamic polymers essential for a wide range of cellular physiologies, such as acting as directional railways for intracellular transport and position, guiding chromosome segregation during cell division, and controlling cell polarity and morphogenesis. Evidence has established that maintaining microtubule (MT) stability in neurons is vital for fundamental cellular and developmental processes, such as neurodevelopment, degeneration, and regeneration. To fulfill these diverse functions, the nervous system employs an arsenal of microtubule-associated proteins (MAPs) to control MT organization and function. Subsequent studies have identified that the disruption of MT function in neurons is one of the most prevalent and important pathological features of traumatic nerve damage and neurodegenerative diseases and that this disruption manifests as a reduction in MT polymerization and concomitant deregulation of the MT cytoskeleton, as well as downregulation of microtubule-associated protein (MAP) expression. A variety of MT-targeting agents that reverse this pathological condition, which is regarded as a therapeutic opportunity to intervene the onset and development of these nervous system abnormalities, is currently under development. Here, we provide an overview of the MT-intrinsic organization process and how MAPs interact with the MT cytoskeleton to promote MT polymerization, stabilization, and bundling. We also highlight recent advances in MT-targeting therapeutic agents applied to various neurological disorders. Together, these findings increase our current understanding of the function and regulation of MT organization in nerve growth and regeneration.
Collapse
Affiliation(s)
- Lijiang Huang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| | - Yan Peng
- Hangzhou Institute for Food and Drug Control, Hangzhou, Zhejiang, China
| | - Xuetao Tao
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xiaoxiao Ding
- Department of Pharmacy, The People's Hospital of Beilun District, Ningbo, Zhejiang 315807, China
| | - Rui Li
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongsheng Jiang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| | - Wei Zuo
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, No. 291 Donggu Road, Xiangshan County, Zhejiang 315000, China
| |
Collapse
|