1
|
Hannaford MR, Rusan NM. Positioning centrioles and centrosomes. J Cell Biol 2024; 223:e202311140. [PMID: 38512059 PMCID: PMC10959756 DOI: 10.1083/jcb.202311140] [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: 12/14/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Centrosomes are the primary microtubule organizer in eukaryotic cells. In addition to shaping the intracellular microtubule network and the mitotic spindle, centrosomes are responsible for positioning cilia and flagella. To fulfill these diverse functions, centrosomes must be properly located within cells, which requires that they undergo intracellular transport. Importantly, centrosome mispositioning has been linked to ciliopathies, cancer, and infertility. The mechanisms by which centrosomes migrate are diverse and context dependent. In many cells, centrosomes move via indirect motor transport, whereby centrosomal microtubules engage anchored motor proteins that exert forces on those microtubules, resulting in centrosome movement. However, in some cases, centrosomes move via direct motor transport, whereby the centrosome or centriole functions as cargo that directly binds molecular motors which then walk on stationary microtubules. In this review, we summarize the mechanisms of centrosome motility and the consequences of centrosome mispositioning and identify key questions that remain to be addressed.
Collapse
Affiliation(s)
- Matthew R. Hannaford
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nasser M. Rusan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Adler A, Bangera M, Beugelink JW, Bahri S, van Ingen H, Moores CA, Baldus M. A structural and dynamic visualization of the interaction between MAP7 and microtubules. Nat Commun 2024; 15:1948. [PMID: 38431715 PMCID: PMC10908866 DOI: 10.1038/s41467-024-46260-5] [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: 06/02/2023] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Microtubules (MTs) are key components of the eukaryotic cytoskeleton and are essential for intracellular organization, organelle trafficking and mitosis. MT tasks depend on binding and interactions with MT-associated proteins (MAPs). MT-associated protein 7 (MAP7) has the unusual ability of both MT binding and activating kinesin-1-mediated cargo transport along MTs. Additionally, the protein is reported to stabilize MTs with its 112 amino-acid long MT-binding domain (MTBD). Here we investigate the structural basis of the interaction of MAP7 MTBD with the MT lattice. Using a combination of solid and solution-state nuclear magnetic resonance (NMR) spectroscopy with electron microscopy, fluorescence anisotropy and isothermal titration calorimetry, we shed light on the binding mode of MAP7 to MTs at an atomic level. Our results show that a combination of interactions between MAP7 and MT lattice extending beyond a single tubulin dimer and including tubulin C-terminal tails contribute to formation of the MAP7-MT complex.
Collapse
Affiliation(s)
- Agnes Adler
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Mamata Bangera
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck, University of London, London, WC1E 7HX, UK
| | - J Wouter Beugelink
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Salima Bahri
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hugo van Ingen
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck, University of London, London, WC1E 7HX, UK.
| | - Marc Baldus
- NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Berisha AM, Eot-Houllier G, Giet R. Imaging and Analysis of Drosophila Neural Stem Cell Asymmetric Division. Methods Mol Biol 2024; 2740:229-242. [PMID: 38393479 DOI: 10.1007/978-1-0716-3557-5_14] [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] [Indexed: 02/25/2024]
Abstract
Cell division is a conserved process among eukaryotes. It is designed to segregate chromosomes into future daughter cells and involves a complex rearrangement of the cytoskeleton, including microtubules and actin filaments. An additional level of complexity is present in asymmetric dividing stem cells because cytoskeleton elements are also regulated by polarity cues. The neural stem cell system of the fruit fly represents a simple model to dissect the mechanisms that control cytoskeleton reorganization during asymmetric division. In this chapter, we propose to describe protocols that allow accurate analysis of microtubule reorganization during cell division in this model.
Collapse
Affiliation(s)
- Anne-Marie Berisha
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR6290-U1305, Rennes, France
| | - Gregory Eot-Houllier
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR6290-U1305, Rennes, France
| | - Régis Giet
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR6290-U1305, Rennes, France.
| |
Collapse
|
4
|
Tan Z, Yue Y, Leprevost F, Haynes S, Basrur V, Nesvizhskii AI, Verhey KJ, Cianfrocco MA. Autoinhibited kinesin-1 adopts a hierarchical folding pattern. eLife 2023; 12:RP86776. [PMID: 37910016 PMCID: PMC10619981 DOI: 10.7554/elife.86776] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Conventional kinesin-1 is the primary anterograde motor in cells for transporting cellular cargo. While there is a consensus that the C-terminal tail of kinesin-1 inhibits motility, the molecular architecture of a full-length autoinhibited kinesin-1 remains unknown. Here, we combine crosslinking mass spectrometry (XL-MS), electron microscopy (EM), and AlphaFold structure prediction to determine the architecture of the full-length autoinhibited kinesin-1 homodimer (kinesin-1 heavy chain [KHC]) and kinesin-1 heterotetramer (KHC bound to kinesin light chain 1 [KLC1]). Our integrative analysis shows that kinesin-1 forms a compact, bent conformation through a break in coiled-coil 3. Moreover, our XL-MS analysis demonstrates that kinesin light chains stabilize the folded inhibited state rather than inducing a new structural state. Using our structural model, we show that disruption of multiple interactions between the motor, stalk, and tail domains is required to activate the full-length kinesin-1. Our work offers a conceptual framework for understanding how cargo adaptors and microtubule-associated proteins relieve autoinhibition to promote activation.
Collapse
Affiliation(s)
- Zhenyu Tan
- Department of Biophysics, University of MichiganAnn ArborUnited States
- Life Sciences Institute, University of MichiganAnn ArborUnited States
| | - Yang Yue
- Department of Cell & Developmental Biology, University of MichiganAnn ArborUnited States
| | - Felipe Leprevost
- Department of Pathology, University of MichiganAnn ArborUnited States
| | - Sarah Haynes
- Department of Pathology, University of MichiganAnn ArborUnited States
| | - Venkatesha Basrur
- Department of Pathology, University of MichiganAnn ArborUnited States
| | - Alexey I Nesvizhskii
- Department of Pathology, University of MichiganAnn ArborUnited States
- Department of Computational Medicine and Bioinformatics, University of MichiganAnn ArborUnited States
| | - Kristen J Verhey
- Department of Cell & Developmental Biology, University of MichiganAnn ArborUnited States
| | - Michael A Cianfrocco
- Life Sciences Institute, University of MichiganAnn ArborUnited States
- Department of Biological Chemistry, University of MichiganAnn ArborUnited States
| |
Collapse
|
5
|
Tan Z, Yue Y, da Veiga Leprevost F, Haynes SE, Basrur V, Nesvizhskii AI, Verhey KJ, Cianfrocco MA. Autoinhibited kinesin-1 adopts a hierarchical folding pattern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525761. [PMID: 36747757 PMCID: PMC9901034 DOI: 10.1101/2023.01.26.525761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Conventional kinesin-1 is the primary anterograde motor in cells for transporting cellular cargo. While there is a consensus that the C-terminal tail of kinesin-1 inhibits motility, the molecular architecture of a full-length autoinhibited kinesin-1 remains unknown. Here, we combine cross-linking mass spectrometry (XL-MS), electron microscopy (EM), and AlphaFold structure prediction to determine the architecture of the full-length autoinhibited kinesin-1 homodimer [kinesin-1 heavy chain (KHC)] and kinesin-1 heterotetramer [KHC bound to kinesin light chain 1 (KLC1)]. Our integrative analysis shows that kinesin-1 forms a compact, bent conformation through a break in coiled coil 3. Moreover, our XL-MS analysis demonstrates that kinesin light chains stabilize the folded inhibited state rather than inducing a new structural state. Using our structural model, we show that disruption of multiple interactions between the motor, stalk, and tail domains is required to activate the full-length kinesin-1. Our work offers a conceptual framework for understanding how cargo adaptors and microtubule-associated proteins relieve autoinhibition to promote activation.
Collapse
Affiliation(s)
- Zhenyu Tan
- Department of Biophysics, University of Michigan
- Life Sciences Institute, University of Michigan
| | - Yang Yue
- Department of Cell & Developmental Biology, University of Michigan
| | | | | | | | - Alexey I. Nesvizhskii
- Department of Pathology, University of Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan
| | | | - Michael A. Cianfrocco
- Life Sciences Institute, University of Michigan
- Department of Biological Chemistry, University of Michigan
| |
Collapse
|
6
|
Adler A, Kjaer LF, Beugelink JW, Baldus M, van Ingen H. Resonance assignments of the microtubule-binding domain of the microtubule-associated protein 7 (MAP7). BIOMOLECULAR NMR ASSIGNMENTS 2023; 17:10.1007/s12104-023-10124-8. [PMID: 37099260 DOI: 10.1007/s12104-023-10124-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 06/02/2023]
Abstract
The microtubule-associated protein 7 (MAP7) is a protein involved in cargo transport along microtubules (MTs) by interacting with kinesin-1 through the C-terminal kinesin-binding domain. Moreover, the protein is reported to stabilize MT, thereby playing a key role in axonal branch development. An important element for this latter function is the 112 amino-acid long N-terminal microtubule-binding domain (MTBD) of MAP7. Here we report NMR backbone and side-chain assignments that suggest a primarily alpha-helical secondary fold of this MTBD in solution. The MTBD contains a central long α-helical segment that includes a short four-residue 'hinge' sequence with decreased helicity and increased flexibility. Our data represent a first step towards analysing the complex interaction of MAP7 with MTs at an atomic level via NMR spectroscopy.
Collapse
Affiliation(s)
- Agnes Adler
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Lenette F Kjaer
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands
- Institute of Structural Biology Grenoble, Grenoble, Auvergne-Rhône-Alpes, France
| | - J Wouter Beugelink
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands
| | - Marc Baldus
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands.
| | - Hugo van Ingen
- Bijvoet Center for Biomolecular Research, NMR Spectroscopy, Utrecht University, Padualaan 8, Utrecht, 3584 CH, The Netherlands.
| |
Collapse
|
7
|
Dullovi A, Ozgencil M, Rajvee V, Tse WY, Cutillas PR, Martin SA, Hořejší Z. Microtubule-associated proteins MAP7 and MAP7D1 promote DNA double-strand break repair in the G1 cell cycle phase. iScience 2023; 26:106107. [PMID: 36852271 PMCID: PMC9958362 DOI: 10.1016/j.isci.2023.106107] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
The DNA-damage response is a complex signaling network that guards genomic integrity. The microtubule cytoskeleton is involved in the repair of DNA double-strand breaks; however, little is known about which cytoskeleton-related proteins are involved in DNA repair and how. Using quantitative proteomics, we discovered that microtubule associated proteins MAP7 and MAP7D1 interact with several DNA repair proteins including DNA double-strand break repair proteins RAD50, BRCA1 and 53BP1. We observed that downregulation of MAP7 and MAP7D1 leads to increased phosphorylation of p53 after γ-irradiation. Moreover, we determined that the downregulation of MAP7D1 leads to a strong G1 arrest and that the downregulation of MAP7 and MAP7D1 in G1 arrested cells negatively affects DNA repair, recruitment of RAD50 to chromatin and localization of 53BP1 to the sites of damage. These findings describe for the first time a novel function of MAP7 and MAP7D1 in cell cycle regulation and repair of DNA double-strand breaks.
Collapse
Affiliation(s)
- Arlinda Dullovi
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Meryem Ozgencil
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Vinothini Rajvee
- Mass Spectrometry Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Wai Yiu Tse
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro R. Cutillas
- Centre for Genomics & Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sarah A. Martin
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Zuzana Hořejší
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK,Corresponding author
| |
Collapse
|
8
|
Hannaford MR, Liu R, Billington N, Swider ZT, Galletta BJ, Fagerstrom CJ, Combs C, Sellers JR, Rusan NM. Pericentrin interacts with Kinesin-1 to drive centriole motility. J Cell Biol 2022; 221:e202112097. [PMID: 35929834 PMCID: PMC9361567 DOI: 10.1083/jcb.202112097] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 06/02/2022] [Accepted: 07/12/2022] [Indexed: 12/23/2022] Open
Abstract
Centrosome positioning is essential for their function. Typically, centrosomes are transported to various cellular locations through the interaction of centrosomal microtubules (MTs) with motor proteins anchored at the cortex or the nuclear surface. However, it remains unknown how centrioles migrate in cellular contexts in which they do not nucleate MTs. Here, we demonstrate that during interphase, inactive centrioles move directly along the interphase MT network as Kinesin-1 cargo. We identify Pericentrin-Like-Protein (PLP) as a novel Kinesin-1 interacting molecule essential for centriole motility. In vitro assays show that PLP directly interacts with the cargo binding domain of Kinesin-1, allowing PLP to migrate on MTs. Binding assays using purified proteins revealed that relief of Kinesin-1 autoinhibition is critical for its interaction with PLP. Finally, our studies of neural stem cell asymmetric divisions in the Drosophila brain show that the PLP-Kinesin-1 interaction is essential for the timely separation of centrioles, the asymmetry of centrosome activity, and the age-dependent centrosome inheritance.
Collapse
Affiliation(s)
- Matthew R. Hannaford
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Rong Liu
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Neil Billington
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Zachary T. Swider
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Brian J. Galletta
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Carey J. Fagerstrom
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christian Combs
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - James R. Sellers
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Nasser M. Rusan
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
9
|
Thomas A, Giet R. Live imaging of Drosophila melanogaster neural stem cells with photo-ablated centrosomes. STAR Protoc 2022; 3:101493. [PMID: 35776653 PMCID: PMC9249821 DOI: 10.1016/j.xpro.2022.101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/26/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Alexandre Thomas
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, ERL U1305, 35000 Rennes, France
| | - Régis Giet
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR6290, ERL U1305, 35000 Rennes, France.
| |
Collapse
|
10
|
Popova JV, Pavlova GA, Razuvaeva AV, Yarinich LA, Andreyeva EN, Anders AF, Galimova YA, Renda F, Somma MP, Pindyurin AV, Gatti M. Genetic Control of Kinetochore-Driven Microtubule Growth in Drosophila Mitosis. Cells 2022; 11:cells11142127. [PMID: 35883570 PMCID: PMC9323100 DOI: 10.3390/cells11142127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/08/2023] Open
Abstract
Centrosome-containing cells assemble their spindles exploiting three main classes of microtubules (MTs): MTs nucleated by the centrosomes, MTs generated near the chromosomes/kinetochores, and MTs nucleated within the spindle by the augmin-dependent pathway. Mammalian and Drosophila cells lacking the centrosomes generate MTs at kinetochores and eventually form functional bipolar spindles. However, the mechanisms underlying kinetochore-driven MT formation are poorly understood. One of the ways to elucidate these mechanisms is the analysis of spindle reassembly following MT depolymerization. Here, we used an RNA interference (RNAi)-based reverse genetics approach to dissect the process of kinetochore-driven MT regrowth (KDMTR) after colcemid-induced MT depolymerization. This MT depolymerization procedure allows a clear assessment of KDMTR, as colcemid disrupts centrosome-driven MT regrowth but not KDMTR. We examined KDMTR in normal Drosophila S2 cells and in S2 cells subjected to RNAi against conserved genes involved in mitotic spindle assembly: mast/orbit/chb (CLASP1), mei-38 (TPX2), mars (HURP), dgt6 (HAUS6), Eb1 (MAPRE1/EB1), Patronin (CAMSAP2), asp (ASPM), and Klp10A (KIF2A). RNAi-mediated depletion of Mast/Orbit, Mei-38, Mars, Dgt6, and Eb1 caused a significant delay in KDMTR, while loss of Patronin had a milder negative effect on this process. In contrast, Asp or Klp10A deficiency increased the rate of KDMTR. These results coupled with the analysis of GFP-tagged proteins (Mast/Orbit, Mei-38, Mars, Eb1, Patronin, and Asp) localization during KDMTR suggested a model for kinetochore-dependent spindle reassembly. We propose that kinetochores capture the plus ends of MTs nucleated in their vicinity and that these MTs elongate at kinetochores through the action of Mast/Orbit. The Asp protein binds the MT minus ends since the beginning of KDMTR, preventing excessive and disorganized MT regrowth. Mei-38, Mars, Dgt6, Eb1, and Patronin positively regulate polymerization, bundling, and stabilization of regrowing MTs until a bipolar spindle is reformed.
Collapse
Affiliation(s)
- Julia V. Popova
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia
| | - Gera A. Pavlova
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Alyona V. Razuvaeva
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Lyubov A. Yarinich
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Evgeniya N. Andreyeva
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
| | - Alina F. Anders
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
| | - Yuliya A. Galimova
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
| | - Fioranna Renda
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy; (F.R.); (M.P.S.)
| | - Maria Patrizia Somma
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy; (F.R.); (M.P.S.)
| | - Alexey V. Pindyurin
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Correspondence: (A.V.P.); (M.G.)
| | - Maurizio Gatti
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (J.V.P.); (G.A.P.); (A.V.R.); (L.A.Y.); (E.N.A.); (A.F.A.); (Y.A.G.)
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome, 00185 Rome, Italy; (F.R.); (M.P.S.)
- Correspondence: (A.V.P.); (M.G.)
| |
Collapse
|
11
|
Gallaud E, Richard-Parpaillon L, Bataillé L, Pascal A, Métivier M, Archambault V, Giet R. The spindle assembly checkpoint and the spatial activation of Polo kinase determine the duration of cell division and prevent tumor formation. PLoS Genet 2022; 18:e1010145. [PMID: 35377889 PMCID: PMC9009772 DOI: 10.1371/journal.pgen.1010145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/14/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023] Open
Abstract
The maintenance of a restricted pool of asymmetrically dividing stem cells is essential for tissue homeostasis. This process requires the control of mitotic progression that ensures the accurate chromosome segregation. In addition, this event is coupled to the asymmetric distribution of cell fate determinants in order to prevent stem cell amplification. How this coupling is regulated remains poorly described. Here, using asymmetrically dividing Drosophila neural stem cells (NSCs), we show that Polo kinase activity levels determine timely Cyclin B degradation and mitotic progression independent of the spindle assembly checkpoint (SAC). This event is mediated by the direct phosphorylation of Polo kinase by Aurora A at spindle poles and Aurora B kinases at centromeres. Furthermore, we show that Aurora A-dependent activation of Polo is the major event that promotes NSC polarization and together with the SAC prevents brain tumor growth. Altogether, our results show that an Aurora/Polo kinase module couples NSC mitotic progression and polarization for tissue homeostasis.
Collapse
Affiliation(s)
- Emmanuel Gallaud
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
| | - Laurent Richard-Parpaillon
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
| | - Laetitia Bataillé
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
| | - Aude Pascal
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
| | - Mathieu Métivier
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada
| | - Régis Giet
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes) UMR 6290, ERL U1305, Rennes, France
- * E-mail:
| |
Collapse
|
12
|
Ferro LS, Fang Q, Eshun-Wilson L, Fernandes J, Jack A, Farrell DP, Golcuk M, Huijben T, Costa K, Gur M, DiMaio F, Nogales E, Yildiz A. Structural and functional insight into regulation of kinesin-1 by microtubule-associated protein MAP7. Science 2022; 375:326-331. [PMID: 35050657 PMCID: PMC8985661 DOI: 10.1126/science.abf6154] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microtubule (MT)-associated protein 7 (MAP7) is a required cofactor for kinesin-1-driven transport of intracellular cargoes. Using cryo-electron microscopy and single-molecule imaging, we investigated how MAP7 binds MTs and facilitates kinesin-1 motility. The MT-binding domain (MTBD) of MAP7 bound MTs as an extended α helix between the protofilament ridge and the site of lateral contact. Unexpectedly, the MTBD partially overlapped with the binding site of kinesin-1 and inhibited its motility. However, by tethering kinesin-1 to the MT, the projection domain of MAP7 prevented dissociation of the motor and facilitated its binding to available neighboring sites. The inhibitory effect of the MTBD dominated as MTs became saturated with MAP7. Our results reveal biphasic regulation of kinesin-1 by MAP7 in the context of their competitive binding to MTs.
Collapse
Affiliation(s)
- Luke S Ferro
- Department of Molecular and Cellular Biology, University of California, Berkeley CA, USA
| | - Qianglin Fang
- Department of Molecular and Cellular Biology, University of California, Berkeley CA, USA
| | - Lisa Eshun-Wilson
- Department of Molecular and Cellular Biology, University of California, Berkeley CA, USA
| | | | - Amanda Jack
- Biophysics Graduate Group, University of California, Berkeley CA, USA
| | - Daniel P Farrell
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mert Golcuk
- Department of Mechanical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Teun Huijben
- Department of Imaging Physics, Delft University of Technology, Delft, Netherlands
| | | | - Mert Gur
- Department of Mechanical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Eva Nogales
- Department of Molecular and Cellular Biology, University of California, Berkeley CA, USA
- Biophysics Graduate Group, University of California, Berkeley CA, USA
- Howard Hughes Medical Institute, Chevy Chase MD, USA
| | - Ahmet Yildiz
- Department of Molecular and Cellular Biology, University of California, Berkeley CA, USA
- Biophysics Graduate Group, University of California, Berkeley CA, USA
- Physics Department, University of California, Berkeley CA, USA
| |
Collapse
|
13
|
Lacroix B, Dumont J. Spatial and Temporal Scaling of Microtubules and Mitotic Spindles. Cells 2022; 11:cells11020248. [PMID: 35053364 PMCID: PMC8774166 DOI: 10.3390/cells11020248] [Citation(s) in RCA: 2] [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: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 02/01/2023] Open
Abstract
During cell division, the mitotic spindle, a macromolecular structure primarily comprised of microtubules, drives chromosome alignment and partitioning between daughter cells. Mitotic spindles can sense cellular dimensions in order to adapt their length and mass to cell size. This scaling capacity is particularly remarkable during early embryo cleavage when cells divide rapidly in the absence of cell growth, thus leading to a reduction of cell volume at each division. Although mitotic spindle size scaling can occur over an order of magnitude in early embryos, in many species the duration of mitosis is relatively short, constant throughout early development and independent of cell size. Therefore, a key challenge for cells during embryo cleavage is not only to assemble a spindle of proper size, but also to do it in an appropriate time window which is compatible with embryo development. How spatial and temporal scaling of the mitotic spindle is achieved and coordinated with the duration of mitosis remains elusive. In this review, we will focus on the mechanisms that support mitotic spindle spatial and temporal scaling over a wide range of cell sizes and cellular contexts. We will present current models and propose alternative mechanisms allowing cells to spatially and temporally coordinate microtubule and mitotic spindle assembly.
Collapse
Affiliation(s)
- Benjamin Lacroix
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, Université de Montpellier, 1919 Route de Mende, CEDEX 5, 34293 Montpellier, France
- Correspondence:
| | - Julien Dumont
- Université de Paris, CNRS, Institut Jacques Monod, F-75013 Paris, France;
| |
Collapse
|
14
|
Collins MA, Coon LA, Thomas R, Mandigo TR, Wynn E, Folker ES. Ensconsin-dependent changes in microtubule organization and LINC complex-dependent changes in nucleus-nucleus interactions result in quantitatively distinct myonuclear positioning defects. Mol Biol Cell 2021; 32:ar27. [PMID: 34524872 PMCID: PMC8693964 DOI: 10.1091/mbc.e21-06-0324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Nuclear movement is a fundamental process of eukaryotic cell biology. Skeletal muscle presents an intriguing model to study nuclear movement because its development requires the precise positioning of multiple nuclei within a single cytoplasm. Furthermore, there is a high correlation between aberrant nuclear positioning and poor muscle function. Although many genes that regulate nuclear movement have been identified, the mechanisms by which these genes act are not known. Using Drosophila melanogaster muscle development as a model system and a combination of live-embryo microscopy and laser ablation of nuclei, we have found that clustered nuclei encompass at least two phenotypes that are caused by distinct mechanisms. Specifically, Ensconsin is necessary for productive force production to drive any movement of nuclei, whereas Bocksbeutel and Klarsicht are necessary to form distinct populations of nuclei that move to different cellular locations. Mechanistically, Ensconsin regulates the number of growing microtubules that are used to move nuclei, whereas Bocksbeutel and Klarsicht regulate interactions between nuclei.
Collapse
Affiliation(s)
| | - L Alexis Coon
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Riya Thomas
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | | | - Elizabeth Wynn
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| |
Collapse
|
15
|
Thomas A, Gallaud E, Pascal A, Serre L, Arnal I, Richard-Parpaillon L, Savoian MS, Giet R. Peripheral astral microtubules ensure asymmetric furrow positioning in neural stem cells. Cell Rep 2021; 37:109895. [PMID: 34706235 DOI: 10.1016/j.celrep.2021.109895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/26/2021] [Accepted: 10/06/2021] [Indexed: 11/18/2022] Open
Abstract
Neuroblast division is characterized by asymmetric positioning of the cleavage furrow, resulting in a large difference in size between the future daughter cells. In animal cells, furrow placement and assembly are governed by centralspindlin that accumulates at the equatorial cell cortex of the future cleavage site and at the spindle midzone. In neuroblasts, these two centralspindlin populations are spatially and temporally separated. A leading pool is located at the basal cleavage site and a second pool accumulates at the midzone before traveling to the cleavage site. The cortical centralspindlin population requires peripheral astral microtubules and the chromosome passenger complex for efficient recruitment. Loss of this pool does not prevent cytokinesis but enhances centralspindlin signaling at the midzone, leading to equatorial furrow repositioning and decreased size asymmetry. These data show that basal furrow positioning in neuroblasts results from a competition between different centralspindlin pools in which the cortical pool is dominant.
Collapse
Affiliation(s)
- Alexandre Thomas
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR17 6290, 35000 Rennes, France
| | - Emmanuel Gallaud
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR17 6290, 35000 Rennes, France
| | - Aude Pascal
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR17 6290, 35000 Rennes, France
| | - Laurence Serre
- Inserm U1216, CEA, CNRS, Grenoble Institut Neurosciences (GIN), Université Grenoble Alpes, 38000 Grenoble, France
| | - Isabelle Arnal
- Inserm U1216, CEA, CNRS, Grenoble Institut Neurosciences (GIN), Université Grenoble Alpes, 38000 Grenoble, France
| | - Laurent Richard-Parpaillon
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR17 6290, 35000 Rennes, France
| | - Matthew Scott Savoian
- School of Fundamental Sciences, Massey University, 4410 Palmerston North, New Zealand
| | - Régis Giet
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR17 6290, 35000 Rennes, France.
| |
Collapse
|
16
|
A release-and-capture mechanism generates an essential non-centrosomal microtubule array during tube budding. Nat Commun 2021; 12:4096. [PMID: 34215746 PMCID: PMC8253823 DOI: 10.1038/s41467-021-24332-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/27/2021] [Indexed: 11/08/2022] Open
Abstract
Non-centrosomal microtubule arrays serve crucial functions in cells, yet the mechanisms of their generation are poorly understood. During budding of the epithelial tubes of the salivary glands in the Drosophila embryo, we previously demonstrated that the activity of pulsatile apical-medial actomyosin depends on a longitudinal non-centrosomal microtubule array. Here we uncover that the exit from the last embryonic division cycle of the epidermal cells of the salivary gland placode leads to one centrosome in the cells losing all microtubule-nucleation capacity. This restriction of nucleation activity to the second, Centrobin-enriched, centrosome is key for proper morphogenesis. Furthermore, the microtubule-severing protein Katanin and the minus-end-binding protein Patronin accumulate in an apical-medial position only in placodal cells. Loss of either in the placode prevents formation of the longitudinal microtubule array and leads to loss of apical-medial actomyosin and impaired apical constriction. We thus propose a mechanism whereby Katanin-severing at the single active centrosome releases microtubule minus-ends that are then anchored by apical-medial Patronin to promote formation of the longitudinal microtubule array crucial for apical constriction and tube formation.
Collapse
|
17
|
Métivier M, Gallaud E, Thomas A, Pascal A, Gagné JP, Poirier GG, Chrétien D, Gibeaux R, Richard-Parpaillon L, Benaud C, Giet R. Drosophila Tubulin-Specific Chaperone E Recruits Tubulin around Chromatin to Promote Mitotic Spindle Assembly. Curr Biol 2021; 31:684-695.e6. [PMID: 33259793 DOI: 10.1016/j.cub.2020.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 09/29/2020] [Accepted: 11/03/2020] [Indexed: 12/31/2022]
Abstract
Proper assembly of mitotic spindles requires microtubule nucleation not only at the centrosomes but also around chromatin. In this study, we found that the Drosophila tubulin-specific chaperone dTBCE is required for the enrichment of tubulin in the nuclear space after nuclear envelope breakdown and for subsequent promotion of spindle microtubule nucleation. These events depend on the CAP-Gly motif found in dTBCE and are regulated by Ran and lamin proteins. Our data suggest that during early mitosis, dTBCE and nuclear pore proteins become enriched in the nucleus, where they interact with the Ran GTPase to promote dynamic tubulin enrichment. We propose that this novel mechanism enhances microtubule nucleation around chromatin, thereby facilitating mitotic spindle assembly.
Collapse
Affiliation(s)
- Mathieu Métivier
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Emmanuel Gallaud
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Alexandre Thomas
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Aude Pascal
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Jean-Philippe Gagné
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Pavillon CHUL, Université Laval, Québec City, QC, Canada
| | - Guy G Poirier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Pavillon CHUL, Université Laval, Québec City, QC, Canada
| | - Denis Chrétien
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Romain Gibeaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Laurent Richard-Parpaillon
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Christelle Benaud
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Régis Giet
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France.
| |
Collapse
|
18
|
Chippalkatti R, Egger B, Suter B. Mms19 promotes spindle microtubule assembly in Drosophila neural stem cells. PLoS Genet 2020; 16:e1008913. [PMID: 33211700 PMCID: PMC7714366 DOI: 10.1371/journal.pgen.1008913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/03/2020] [Accepted: 10/13/2020] [Indexed: 01/27/2023] Open
Abstract
Mitotic divisions depend on the timely assembly and proper orientation of the mitotic spindle. Malfunctioning of these processes can considerably delay mitosis, thereby compromising tissue growth and homeostasis, and leading to chromosomal instability. Loss of functional Mms19 drastically affects the growth and development of mitotic tissues in Drosophila larvae and we now demonstrate that Mms19 is an important factor that promotes spindle and astral microtubule (MT) growth, and MT stability and bundling. Mms19 function is needed for the coordination of mitotic events and for the rapid progression through mitosis that is characteristic of neural stem cells. Surprisingly, Mms19 performs its mitotic activities through two different pathways. By stimulating the mitotic kinase cascade, it triggers the localization of the MT regulatory complex TACC/Msps (Transforming Acidic Coiled Coil/Minispindles, the homolog of human ch-TOG) to the centrosome. This activity of Mms19 can be rescued by stimulating the mitotic kinase cascade. However, other aspects of the Mms19 phenotypes cannot be rescued in this way, pointing to an additional mechanism of Mms19 action. We provide evidence that Mms19 binds directly to MTs and that this stimulates MT stability and bundling.
Collapse
Affiliation(s)
- Rohan Chippalkatti
- Cell Biology, University of Bern, Berne, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Berne, Switzerland
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Beat Suter
- Cell Biology, University of Bern, Berne, Switzerland
| |
Collapse
|
19
|
Liang Y, Meyer A, Kratochwil CF. Neural innervation as a potential trigger of morphological color change and sexual dimorphism in cichlid fish. Sci Rep 2020; 10:12329. [PMID: 32704058 PMCID: PMC7378239 DOI: 10.1038/s41598-020-69239-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
Many species change their coloration during ontogeny or even as adults. Color change hereby often serves as sexual or status signal. The cellular and subcellular changes that drive color change and how they are orchestrated have been barely understood, but a deeper knowledge of the underlying processes is important to our understanding of how such plastic changes develop and evolve. Here we studied the color change of the Malawi golden cichlid (Melanchromis auratus). Females and subordinate males of this species are yellow and white with two prominent black stripes (yellow morph; female and non-breeding male coloration), while dominant males change their color and completely invert this pattern with the yellow and white regions becoming black, and the black stripes becoming white to iridescent blue (dark morph; male breeding coloration). A comparison of the two morphs reveals that substantial changes across multiple levels of biological organization underlie this polyphenism. These include changes in pigment cell (chromatophore) number, intracellular dispersal of pigments, and tilting of reflective platelets (iridosomes) within iridophores. At the transcriptional level, we find differences in pigmentation gene expression between these two color morphs but, surprisingly, 80% of the genes overexpressed in the dark morph relate to neuronal processes including synapse formation. Nerve fiber staining confirms that scales of the dark morph are indeed innervated by 1.3 to 2 times more axonal fibers. Our results might suggest an instructive role of nervous innervation orchestrating the complex cellular and ultrastructural changes that drive the morphological color change of this cichlid species.
Collapse
Affiliation(s)
- Yipeng Liang
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany
| | - Axel Meyer
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany.
| | - Claudius F Kratochwil
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany.
| |
Collapse
|
20
|
Zhang R, Li L, Chen L, Suo Y, Fan J, Zhang S, Wang Y, Gao S, Wang Y. MAP7 interacts with RC3H1 and cooperatively regulate cell-cycle progression of cervical cancer cells via activating the NF-κB signaling. Biochem Biophys Res Commun 2020; 527:56-63. [PMID: 32446391 DOI: 10.1016/j.bbrc.2020.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/04/2020] [Indexed: 12/14/2022]
Abstract
Ensconsin is encoded by the MAP7 gene and belongs to the microtubule-associated proteins. This study aimed to explore its functional roles and partners in cell-cycle progression in cervical cancer. Data from the Cancer Genome Atlas-Cervical & Endocervical Cancer (TCGA-CESC) and the Genotype-Tissue Expression project were used for bioinformatic analysis. SiHa cells were used for in-vitro and in-vivo analysis. Co-immunoprecipitation (Co-IP) assay was conducted to explore the proteins interacted with MAP7. Results showed that MAP7 mRNA expression might serve as an independent biomarker of shorter survival. MAP7 overexpression elevated cyclin D1/cyclin B1 expression, facilitated cell-cycle progression and promoted SiHa cell growth in a xenograft tumor model. Co-IP experiments confirmed a novel interaction between MAP7 and RC3H1. Knockdown of either RC3H1 or MAP7 significantly attenuated cyclin D1/cyclin B1 upregulation, and cell-cycle progression induced by the other partner. MAP7 overexpression led to increased expression of P-IKK (Ser176/177) and P-p65 (Ser536). RC3H1 inhibition abrogated MAP7 induced upregulation of P-IKK and P-p65. Data in TCGA-CESC showed that MAP7 expression was positively correlated with its copy number segments, but was negatively correlated with the methylation level of three CpG sites within the gene locus. Demethylation treatment by 5-Aza-dC elevated both MAP7 mRNA and protein expression in a dose-dependent manner. In conclusion, this study revealed a novel interaction between MAP7 and RC3H1 in cervical cancer cells, which cooperatively enhanced cyclin D1/cyclin B1 expression and facilitated cell-cycle progression. These effects were at least partly mediated by activated canonical IKK/NF-kB signaling.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China; Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Lei Li
- Department of Radiotherapy, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Lingli Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yuping Suo
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Jingjing Fan
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Suyu Zhang
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Yulan Wang
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Shenxiang Gao
- Department of Gynecology and Obstetrics, People's Hospital of Shanxi Province, Taiyuan, 030012, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
21
|
MAP7D2 Localizes to the Proximal Axon and Locally Promotes Kinesin-1-Mediated Cargo Transport into the Axon. Cell Rep 2020; 26:1988-1999.e6. [PMID: 30784582 PMCID: PMC6381606 DOI: 10.1016/j.celrep.2019.01.084] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/20/2018] [Accepted: 01/24/2019] [Indexed: 11/20/2022] Open
Abstract
The motor protein kinesin-1 plays an important role in polarized sorting of transport vesicles to the axon. However, the mechanism by which the axonal entry of kinesin-1-dependent cargo transport is regulated remains unclear. Microtubule-associated protein MAP7 (ensconsin in Drosophila) is an essential kinesin-1 cofactor and promotes kinesin-1 recruitment to microtubules. Here, we found that MAP7 family member MAP7D2 concentrates at the proximal axon, where it overlaps with the axon initial segment and interacts with kinesin-1. Depletion of MAP7D2 results in reduced axonal cargo entry and defects in axon development and neuronal migration. We propose a model in which MAP7D2 in the proximal axon locally promotes kinesin-1-mediated cargo entry into the axon.
Collapse
|
22
|
Goupil A, Nano M, Letort G, Gemble S, Edwards F, Goundiam O, Gogendeau D, Pennetier C, Basto R. Chromosomes function as a barrier to mitotic spindle bipolarity in polyploid cells. J Cell Biol 2020; 219:133854. [PMID: 32328633 PMCID: PMC7147111 DOI: 10.1083/jcb.201908006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/13/2019] [Accepted: 01/24/2020] [Indexed: 01/22/2023] Open
Abstract
Ploidy variations such as genome doubling are frequent in human tumors and have been associated with genetic instability favoring tumor progression. How polyploid cells deal with increased centrosome numbers and DNA content remains unknown. Using Drosophila neuroblasts and human cancer cells to study mitotic spindle assembly in polyploid cells, we found that most polyploid cells divide in a multipolar manner. We show that even if an initial centrosome clustering step can occur at mitotic entry, the establishment of kinetochore-microtubule attachments leads to spatial chromosome configurations, whereby the final coalescence of supernumerary poles into a bipolar array is inhibited. Using in silico approaches and various spindle and DNA perturbations, we show that chromosomes act as a physical barrier blocking spindle pole coalescence and bipolarity. Importantly, microtubule stabilization suppressed multipolarity by improving both centrosome clustering and pole coalescence. This work identifies inhibitors of bipolar division in polyploid cells and provides a rationale to understand chromosome instability typical of polyploid cancer cells.
Collapse
Affiliation(s)
- Alix Goupil
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Maddalena Nano
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Gaëlle Letort
- Center for Interdisciplinary Research in Biology, Collège de France, UMR7241/U1050, Paris, France
| | - Simon Gemble
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Frances Edwards
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Oumou Goundiam
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France.,Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Delphine Gogendeau
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Carole Pennetier
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Renata Basto
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| |
Collapse
|
23
|
Pirani V, Métivier M, Gallaud E, Thomas A, Ku S, Chretien D, Ettari R, Giet R, Corsi L, Benaud C. A novel benzodiazepine derivative that suppresses microtubule dynamics and impairs mitotic progression. J Cell Sci 2020; 133:jcs239244. [PMID: 32094264 DOI: 10.1242/jcs.239244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/17/2020] [Indexed: 11/20/2022] Open
Abstract
A novel 2,3-benzodiazepine-4 derivative, named 1g, has recently been shown to function as an anti-proliferative compound. We now show that it perturbs the formation of a functional mitotic spindle, inducing a spindle assembly checkpoint (SAC)-dependent arrest in human cells. Live analysis of individual microtubules indicates that 1g promotes a rapid and reversible reduction in microtubule growth. Unlike most anti-mitotic compounds, we found that 1g does not interfere directly with tubulin or perturb microtubule assembly in vitro The observation that 1g also triggers a SAC-dependent mitotic delay associated with chromosome segregation in Drosophila neural stem cells, suggests that it targets a conserved microtubule regulation module in humans and flies. Altogether, our results indicate that 1g is a novel promising anti-mitotic drug with the unique properties of altering microtubule growth and mitotic spindle organization.
Collapse
Affiliation(s)
- Vittoria Pirani
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Mathieu Métivier
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Emmanuel Gallaud
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Alexandre Thomas
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Siou Ku
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Denis Chretien
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Regis Giet
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Christelle Benaud
- University of Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, 35000 Rennes, France
| |
Collapse
|
24
|
Rohrberg J, Van de Mark D, Amouzgar M, Lee JV, Taileb M, Corella A, Kilinc S, Williams J, Jokisch ML, Camarda R, Balakrishnan S, Shankar R, Zhou A, Chang AN, Chen B, Rugo HS, Dumont S, Goga A. MYC Dysregulates Mitosis, Revealing Cancer Vulnerabilities. Cell Rep 2020; 30:3368-3382.e7. [PMID: 32160543 PMCID: PMC7085414 DOI: 10.1016/j.celrep.2020.02.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/18/2019] [Accepted: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Tumors that overexpress the MYC oncogene are frequently aneuploid, a state associated with highly aggressive cancers and tumor evolution. However, how MYC causes aneuploidy is not well understood. Here, we show that MYC overexpression induces mitotic spindle assembly defects and chromosomal instability (CIN) through effects on microtubule nucleation and organization. Attenuating MYC expression reverses mitotic defects, even in established tumor cell lines, indicating an ongoing role for MYC in CIN. MYC reprograms mitotic gene expression, and we identify TPX2 to be permissive for spindle assembly in MYC-high cells. TPX2 depletion blocks mitotic progression, induces cell death, and prevents tumor growth. Further elevating TPX2 expression reduces mitotic defects in MYC-high cells. MYC and TPX2 expression may be useful biomarkers to stratify patients for anti-mitotic therapies. Our studies implicate MYC as a regulator of mitosis and suggest that blocking MYC activity can attenuate the emergence of CIN and tumor evolution.
Collapse
Affiliation(s)
- Julia Rohrberg
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA.
| | - Daniel Van de Mark
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Meelad Amouzgar
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Joyce V Lee
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Moufida Taileb
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Corella
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Seda Kilinc
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy Williams
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Marie-Lena Jokisch
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Roman Camarda
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Sanjeev Balakrishnan
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Rama Shankar
- Department of Pediatrics and Human Development and Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Alicia Zhou
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Bin Chen
- Department of Pediatrics and Human Development and Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Hope S Rugo
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Sophie Dumont
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrei Goga
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
25
|
Monroy BY, Tan TC, Oclaman JM, Han JS, Simó S, Niwa S, Nowakowski DW, McKenney RJ, Ori-McKenney KM. A Combinatorial MAP Code Dictates Polarized Microtubule Transport. Dev Cell 2020; 53:60-72.e4. [PMID: 32109385 DOI: 10.1016/j.devcel.2020.01.029] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/19/2019] [Accepted: 01/27/2020] [Indexed: 01/14/2023]
Abstract
Many eukaryotic cells distribute their intracellular components asymmetrically through regulated active transport driven by molecular motors along microtubule tracks. While intrinsic and extrinsic regulation of motor activity exists, what governs the overall distribution of activated motor-cargo complexes within cells remains unclear. Here, we utilize in vitro reconstitution of purified motor proteins and non-enzymatic microtubule-associated proteins (MAPs) to demonstrate that MAPs exhibit distinct influences on the motility of the three main classes of transport motors: kinesin-1, kinesin-3, and cytoplasmic dynein. Further, we dissect how combinations of MAPs affect motors and unveil MAP9 as a positive modulator of kinesin-3 motility. From these data, we propose a general "MAP code" that has the capacity to strongly bias directed movement along microtubules and helps elucidate the intricate intracellular sorting observed in highly polarized cells such as neurons.
Collapse
Affiliation(s)
- Brigette Y Monroy
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Tracy C Tan
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Janah May Oclaman
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Jisoo S Han
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| | | | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
26
|
Zhang L, Liu X, Song L, Zhai H, Chang C. MAP7 promotes migration and invasion and progression of human cervical cancer through modulating the autophagy. Cancer Cell Int 2020; 20:17. [PMID: 31956295 PMCID: PMC6958635 DOI: 10.1186/s12935-020-1095-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Microtubule-associated proteins 7(MAP7) was reported to be engaged into the function of neuronal function. The function of MAP7 in human cervical cancer (CC) was unknown. We aimed to uncover the function and mechanism of MAP7 on CC. Methods We applied qRT-PCR, western blot and immunochemistry to detect the expression difference between normal tissue and CC. In vitro, we establish MAP7 stable knocking down and overexpression cell lines and investigated the function and underlying mechanism of MAP7 in CC. Results Both mRNA and protein of MAP7 were upregulated in CC compared with the normal tissue. MAP7 was correlated with the clinical stage and tumor size and lymph node metastasis. MAP7 promotes the invasion and migration of CC cell lines. We next detected EMT pathway and autophagy associated pathway. MAP7 promotes the EMT through modulating the autophagy. Conclusion Taken above, our results showed that MAP7 promotes the migration and invasion and EMT through modulating the autophagy.
Collapse
Affiliation(s)
- Li Zhang
- 1Department of Gynaecology and Obstetrics, Jinan Women and Children Health Hospital, No. 2 Jianguo Xiaojing 3rd Road Center Area, Jinan, Shandong 250001 China
| | - Xudong Liu
- 2Department of Pain, Qilu Hospital of Shandong University, Jinan, China
| | - Lina Song
- 1Department of Gynaecology and Obstetrics, Jinan Women and Children Health Hospital, No. 2 Jianguo Xiaojing 3rd Road Center Area, Jinan, Shandong 250001 China
| | - Hui Zhai
- 1Department of Gynaecology and Obstetrics, Jinan Women and Children Health Hospital, No. 2 Jianguo Xiaojing 3rd Road Center Area, Jinan, Shandong 250001 China
| | - Chaohua Chang
- 3Department of Anethesia, Jinan Women and Children Health Hospital, No. 2 Jianguo Xiaojing 3rd Road Center Area, Jinan, Shandong 250001 China
| |
Collapse
|
27
|
Colgan W, Leanza A, Hwang A, DeBiasse MB, Llosa I, Rodrigues D, Adhikari H, Barreto Corona G, Bock S, Carillo-Perez A, Currie M, Darkoa-Larbi S, Dellal D, Gutow H, Hokama P, Kibby E, Linhart N, Moody S, Naganuma A, Nguyen D, Stanton R, Stark S, Tumey C, Velleca A, Ryan JF, Davidson B. Variable levels of drift in tunicate cardiopharyngeal gene regulatory elements. EvoDevo 2019; 10:24. [PMID: 31632631 PMCID: PMC6790052 DOI: 10.1186/s13227-019-0137-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/13/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Mutations in gene regulatory networks often lead to genetic divergence without impacting gene expression or developmental patterning. The rules governing this process of developmental systems drift, including the variable impact of selective constraints on different nodes in a gene regulatory network, remain poorly delineated. RESULTS Here we examine developmental systems drift within the cardiopharyngeal gene regulatory networks of two tunicate species, Corella inflata and Ciona robusta. Cross-species analysis of regulatory elements suggests that trans-regulatory architecture is largely conserved between these highly divergent species. In contrast, cis-regulatory elements within this network exhibit distinct levels of conservation. In particular, while most of the regulatory elements we analyzed showed extensive rearrangements of functional binding sites, the enhancer for the cardiopharyngeal transcription factor FoxF is remarkably well-conserved. Even minor alterations in spacing between binding sites lead to loss of FoxF enhancer function, suggesting that bound trans-factors form position-dependent complexes. CONCLUSIONS Our findings reveal heterogeneous levels of divergence across cardiopharyngeal cis-regulatory elements. These distinct levels of divergence presumably reflect constraints that are not clearly associated with gene function or position within the regulatory network. Thus, levels of cis-regulatory divergence or drift appear to be governed by distinct structural constraints that will be difficult to predict based on network architecture.
Collapse
Affiliation(s)
| | - Alexis Leanza
- Thomas Jefferson University Sidney Kimmel Medical College, Philadelphia, USA
| | - Ariel Hwang
- University of North Carolina, Chapel Hill, USA
| | | | | | | | | | | | | | | | | | | | - Daniel Dellal
- Icahn School of Medicine at Mount Sinai, New York, USA
| | | | | | - Emily Kibby
- University of Colorado Boulder, Boulder, USA
| | | | | | | | | | | | - Sierra Stark
- University of California San Francisco, San Francisco, USA
| | | | | | - Joseph F. Ryan
- Whitney Laboratory for Marine Bioscience, St. Augustine, USA
| | | |
Collapse
|
28
|
Chaudhary AR, Lu H, Krementsova EB, Bookwalter CS, Trybus KM, Hendricks AG. MAP7 regulates organelle transport by recruiting kinesin-1 to microtubules. J Biol Chem 2019; 294:10160-10171. [PMID: 31085585 PMCID: PMC6664170 DOI: 10.1074/jbc.ra119.008052] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/07/2019] [Indexed: 12/14/2022] Open
Abstract
Microtubule-associated proteins (MAPs) regulate microtubule polymerization, dynamics, and organization. In addition, MAPs alter the motility of kinesin and dynein to control trafficking along microtubules. MAP7 (ensconsin, E-MAP-115) is a ubiquitous MAP that organizes the microtubule cytoskeleton in mitosis and neuronal branching. MAP7 also recruits kinesin-1 to microtubules. To understand how the activation of kinesin-1 by MAP7 regulates the motility of organelles transported by ensembles of kinesin and dynein, we isolated organelles and reconstituted their motility in vitro In the absence of MAP7, isolated phagosomes exhibit approximately equal fractions of plus- and minus-end-directed motility along microtubules. MAP7 causes a pronounced shift in motility; phagosomes move toward the plus-end ∼80% of the time, and kinesin teams generate more force. To dissect MAP7-mediated regulation of kinesin-driven transport, we examined its effects on the motility and force generation of single and teams of full-length kinesin-1 motors. We find that MAP7 does not alter the force exerted by a single kinesin-1 motor, but instead increases its binding rate to the microtubule. For ensembles of kinesin, a greater number of kinesin motors are simultaneously engaged and generating force to preferentially target organelles toward the microtubule plus-end.
Collapse
Affiliation(s)
- Abdullah R Chaudhary
- From the Department of Bioengineering, McGill University, Montréal, Quebec H3A 0C3, Canada and
| | - Hailong Lu
- the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405-0075
| | - Elena B Krementsova
- the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405-0075
| | - Carol S Bookwalter
- the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405-0075
| | - Kathleen M Trybus
- the Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont 05405-0075
| | - Adam G Hendricks
- From the Department of Bioengineering, McGill University, Montréal, Quebec H3A 0C3, Canada and
| |
Collapse
|
29
|
Métivier M, Monroy BY, Gallaud E, Caous R, Pascal A, Richard-Parpaillon L, Guichet A, Ori-McKenney KM, Giet R. Dual control of Kinesin-1 recruitment to microtubules by Ensconsin in Drosophila neuroblasts and oocytes. Development 2019; 146:dev.171579. [PMID: 30936181 DOI: 10.1242/dev.171579] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/25/2019] [Indexed: 01/02/2023]
Abstract
Drosophila Ensconsin (also known as MAP7) controls spindle length, centrosome separation in brain neuroblasts (NBs) and asymmetric transport in oocytes. The control of spindle length by Ensconsin is Kinesin-1 independent but centrosome separation and oocyte transport require targeting of Kinesin-1 to microtubules by Ensconsin. However, the molecular mechanism used for this targeting remains unclear. Ensconsin contains a microtubule (MT)-binding domain (MBD) and a Kinesin-binding domain (KBD). Rescue experiments show that only full-length Ensconsin restores the spindle length phenotype. KBD expression rescues ensc centrosome separation defects in NBs, but not the fast oocyte streaming and the localization of Staufen and Gurken. Interestingly, the KBD can stimulate Kinesin-1 targeting to MTs in vivo and in vitro We propose that a KBD and Kinesin-1 complex is a minimal activation module that increases Kinesin-1 affinity for MTs. Addition of the MBD present in full-length Ensconsin allows this process to occur directly on the MT and triggers higher Kinesin-1 targeting. This dual regulation by Ensconsin is essential for optimal Kinesin-1 targeting to MTs in oocytes, but not in NBs, illustrating the importance of adapting Kinesin-1 recruitment to different biological contexts.
Collapse
Affiliation(s)
- Mathieu Métivier
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Brigette Y Monroy
- University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Emmanuel Gallaud
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Renaud Caous
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Aude Pascal
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Laurent Richard-Parpaillon
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Antoine Guichet
- Institut Jacques Monod-Université Paris Diderot-Paris 7, 15 rue Hélène Brion, 75205 Paris Cedex 13, France
| | | | - Régis Giet
- Univ. Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| |
Collapse
|
30
|
Hooikaas PJ, Martin M, Mühlethaler T, Kuijntjes GJ, Peeters CAE, Katrukha EA, Ferrari L, Stucchi R, Verhagen DGF, van Riel WE, Grigoriev I, Altelaar AFM, Hoogenraad CC, Rüdiger SGD, Steinmetz MO, Kapitein LC, Akhmanova A. MAP7 family proteins regulate kinesin-1 recruitment and activation. J Cell Biol 2019; 218:1298-1318. [PMID: 30770434 PMCID: PMC6446838 DOI: 10.1083/jcb.201808065] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/22/2018] [Accepted: 01/22/2019] [Indexed: 02/08/2023] Open
Abstract
Kinesin-1 is responsible for microtubule-based transport of numerous cellular cargoes. Here, we explored the regulation of kinesin-1 by MAP7 proteins. We found that all four mammalian MAP7 family members bind to kinesin-1. In HeLa cells, MAP7, MAP7D1, and MAP7D3 act redundantly to enable kinesin-1-dependent transport and microtubule recruitment of the truncated kinesin-1 KIF5B-560, which contains the stalk but not the cargo-binding and autoregulatory regions. In vitro, purified MAP7 and MAP7D3 increase microtubule landing rate and processivity of kinesin-1 through transient association with the motor. MAP7 proteins promote binding of kinesin-1 to microtubules both directly, through the N-terminal microtubule-binding domain and unstructured linker region, and indirectly, through an allosteric effect exerted by the kinesin-binding C-terminal domain. Compared with MAP7, MAP7D3 has a higher affinity for kinesin-1 and a lower affinity for microtubules and, unlike MAP7, can be cotransported with the motor. We propose that MAP7 proteins are microtubule-tethered kinesin-1 activators, with which the motor transiently interacts as it moves along microtubules.
Collapse
Affiliation(s)
- Peter Jan Hooikaas
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Maud Martin
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Tobias Mühlethaler
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Gert-Jan Kuijntjes
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Cathelijn A E Peeters
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Luca Ferrari
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Riccardo Stucchi
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Daan G F Verhagen
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Wilhelmina E van Riel
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ilya Grigoriev
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - A F Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences and The Netherlands Proteomics Centre, Utrecht University, Utrecht, Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Stefan G D Rüdiger
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
31
|
Rosen JN, Azevedo M, Soffar DB, Boyko VP, Brendel MB, Schulman VK, Baylies MK. The Drosophila Ninein homologue Bsg25D cooperates with Ensconsin in myonuclear positioning. J Cell Biol 2019; 218:524-540. [PMID: 30626718 PMCID: PMC6363458 DOI: 10.1083/jcb.201808176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023] Open
Abstract
Rosen et al. identify a role for the centrosomal protein Bsg25D/Ninein in nuclear positioning and microtubule organization in Drosophila muscle fibers. Genetic, cell biological, and atomic force microscopy analyses demonstrate that complex interactions between Bsg25D and the microtubule-associated protein Ensconsin govern myonuclear positioning in Drosophila. Skeletal muscle consists of multinucleated cells in which the myonuclei are evenly spaced throughout the cell. In Drosophila, this pattern is established in embryonic myotubes, where myonuclei move via microtubules (MTs) and the MT-associated protein Ensconsin (Ens)/MAP7, to achieve their distribution. Ens regulates multiple aspects of MT biology, but little is known about how Ens itself is regulated. We find that Ens physically interacts and colocalizes with Bsg25D, the Drosophila homologue of the centrosomal protein Ninein. Bsg25D loss enhances myonuclear positioning defects in embryos sensitized by partial Ens loss. Bsg25D overexpression causes severe positioning defects in immature myotubes and fully differentiated myofibers, where it forms ectopic MT organizing centers, disrupts perinuclear MT arrays, reduces muscle stiffness, and decreases larval crawling velocity. These studies define a novel relationship between Ens and Bsg25D. At endogenous levels, Bsg25D positively regulates Ens activity during myonuclear positioning, but excess Bsg25D disrupts Ens localization and MT organization, with disastrous consequences for myonuclear positioning and muscle function.
Collapse
Affiliation(s)
- Jonathan N Rosen
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mafalda Azevedo
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Graduate Program in Areas of Basic and Applied Biology, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - David B Soffar
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vitaly P Boyko
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Molecular Cytology Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Matthew B Brendel
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Molecular Cytology Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Victoria K Schulman
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY.,Cell and Developmental Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY
| | - Mary K Baylies
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY .,Cell and Developmental Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY
| |
Collapse
|
32
|
Tymanskyj SR, Yang BH, Verhey KJ, Ma L. MAP7 regulates axon morphogenesis by recruiting kinesin-1 to microtubules and modulating organelle transport. eLife 2018; 7:36374. [PMID: 30132755 PMCID: PMC6133550 DOI: 10.7554/elife.36374] [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: 03/05/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
Neuronal cell morphogenesis depends on proper regulation of microtubule-based transport, but the underlying mechanisms are not well understood. Here, we report our study of MAP7, a unique microtubule-associated protein that interacts with both microtubules and the motor protein kinesin-1. Structure-function analysis in rat embryonic sensory neurons shows that the kinesin-1 interacting domain in MAP7 is required for axon and branch growth but not for branch formation. Also, two unique microtubule binding sites are found in MAP7 that have distinct dissociation kinetics and are both required for branch formation. Furthermore, MAP7 recruits kinesin-1 dynamically to microtubules, leading to alterations in organelle transport behaviors, particularly pause/speed switching. As MAP7 is localized to branch sites, our results suggest a novel mechanism mediated by the dual interactions of MAP7 with microtubules and kinesin-1 in the precise control of microtubule-based transport during axon morphogenesis.
Collapse
Affiliation(s)
- Stephen R Tymanskyj
- Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, United States.,Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, United States
| | - Benjamin H Yang
- Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, United States.,Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, United States
| | - Kristen J Verhey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Le Ma
- Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, United States.,Department of Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, United States
| |
Collapse
|
33
|
Monroy BY, Sawyer DL, Ackermann BE, Borden MM, Tan TC, Ori-McKenney KM. Competition between microtubule-associated proteins directs motor transport. Nat Commun 2018; 9:1487. [PMID: 29662074 PMCID: PMC5902456 DOI: 10.1038/s41467-018-03909-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/21/2018] [Indexed: 11/09/2022] Open
Abstract
Within cells, motor and non-motor microtubule-associated proteins (MAPs) simultaneously converge on the microtubule. How the binding activities of non-motor MAPs are coordinated and how they contribute to the balance and distribution of motor transport is unknown. Here, we examine the relationship between MAP7 and tau owing to their antagonistic roles in vivo. We find that MAP7 and tau compete for binding to microtubules, and determine a mechanism by which MAP7 displaces tau from the lattice. MAP7 promotes kinesin-based transport in vivo and strongly recruits kinesin-1 to the microtubule in vitro, providing evidence for direct enhancement of motor motility by a MAP. Both MAP7 and tau strongly inhibit kinesin-3 and have no effect on cytoplasmic dynein, demonstrating that MAPs differentially control distinct classes of motors. Overall, these results reveal a general principle for how MAP competition dictates access to the microtubule to determine the correct distribution and balance of motor activity.
Collapse
Affiliation(s)
- Brigette Y Monroy
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Danielle L Sawyer
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Bryce E Ackermann
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Melissa M Borden
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Tracy C Tan
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
34
|
Nag RN, Niggli S, Sousa-Guimarães S, Vazquez-Pianzola P, Suter B. Mms19 is a mitotic gene that permits Cdk7 to be fully active as a Cdk-activating kinase. Development 2018; 145:dev.156802. [PMID: 29361561 PMCID: PMC5825849 DOI: 10.1242/dev.156802] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 12/18/2017] [Indexed: 11/20/2022]
Abstract
Mms19 encodes a cytosolic iron-sulphur assembly component. We found that Drosophila Mms19 is also essential for mitotic divisions and for the proliferation of diploid cells. Reduced Mms19 activity causes severe mitotic defects in spindle dynamics and chromosome segregation, and loss of zygotic Mms19 prevents the formation of imaginal discs. The lack of mitotic tissue in Mms19P/P larvae can be rescued by overexpression of the Cdk-activating kinase (CAK) complex, an activator of mitotic Cdk1, suggesting that Mms19 functions in mitosis to allow CAK (Cdk7/Cyclin H/Mat1) to become fully active as a Cdk1-activating kinase. When bound to Xpd and TFIIH, the CAK subunit Cdk7 phosphorylates transcriptional targets and not cell cycle Cdks. In contrast, free CAK phosphorylates and activates Cdk1. Physical and genetic interaction studies between Mms19 and Xpd suggest that their interaction prevents Xpd from binding to the CAK complex. Xpd bound to Mms19 therefore frees CAK complexes, allowing them to phosphorylate Cdk1 and facilitating progression to metaphase. The structural basis for the competitive interaction with Xpd seems to be the binding of Mms19, core TFIIH and CAK to neighbouring or overlapping regions of Xpd. Summary: Interaction studies demonstrate that Mms19 forms complexes with Xpd, thereby preventing Xpd-mediated repression of the mitotic kinase activity of the CAK complex and facilitating progression through mitosis.
Collapse
Affiliation(s)
- Rishita Narendra Nag
- Institute of Cell Biology, Department of Biology, University of Bern, 3012 Bern, Switzerland
| | - Selina Niggli
- Institute of Cell Biology, Department of Biology, University of Bern, 3012 Bern, Switzerland
| | - Sofia Sousa-Guimarães
- Institute of Cell Biology, Department of Biology, University of Bern, 3012 Bern, Switzerland
| | - Paula Vazquez-Pianzola
- Institute of Cell Biology, Department of Biology, University of Bern, 3012 Bern, Switzerland
| | - Beat Suter
- Institute of Cell Biology, Department of Biology, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
35
|
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: 4.0] [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
|
36
|
Ramkumar A, Jong BY, Ori-McKenney KM. ReMAPping the microtubule landscape: How phosphorylation dictates the activities of microtubule-associated proteins. Dev Dyn 2017; 247:138-155. [PMID: 28980356 DOI: 10.1002/dvdy.24599] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Classical microtubule-associated proteins (MAPs) were originally identified based on their co-purification with microtubules assembled from mammalian brain lysate. They have since been found to perform a range of functions involved in regulating the dynamics of the microtubule cytoskeleton. Most of these MAPs play integral roles in microtubule organization during neuronal development, microtubule remodeling during neuronal activity, and microtubule stabilization during neuronal maintenance. As a result, mutations in MAPs contribute to neurodevelopmental disorders, psychiatric conditions, and neurodegenerative diseases. MAPs are post-translationally regulated by phosphorylation depending on developmental time point and cellular context. Phosphorylation can affect the microtubule affinity, cellular localization, or overall function of a particular MAP and can thus have profound implications for neuronal health. Here we review MAP1, MAP2, MAP4, MAP6, MAP7, MAP9, tau, and DCX, and how each is regulated by phosphorylation in neuronal physiology and disease. Developmental Dynamics 247:138-155, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amrita Ramkumar
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | - Brigette Y Jong
- Department of Molecular and Cellular Biology, University of California, Davis, CA
| | | |
Collapse
|
37
|
MAP7 Regulates Axon Collateral Branch Development in Dorsal Root Ganglion Neurons. J Neurosci 2017; 37:1648-1661. [PMID: 28069923 DOI: 10.1523/jneurosci.3260-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/27/2016] [Accepted: 01/04/2017] [Indexed: 01/26/2023] Open
Abstract
Collateral branches from axons are key components of functional neural circuits that allow neurons to connect with multiple synaptic targets. Like axon growth and guidance, formation of collateral branches depends on the regulation of microtubules, but how such regulation is coordinated to ensure proper circuit development is not known. Based on microarray analysis, we have identified a role for microtubule-associated protein 7 (MAP7) during collateral branch development of dorsal root ganglion (DRG) sensory neurons. We show that MAP7 is expressed at the onset of collateral branch formation. Perturbation of its expression by overexpression or shRNA knockdown alters axon branching in cultured DRG neurons. Localization and time-lapse imaging analysis reveals that MAP7 is enriched at branch points and colocalizes with stable microtubules, but enters the new branch with a delay, suggesting a role in branch maturation. We have also investigated a spontaneous mutant mouse that expresses a truncated MAP7 and found a gain-of-function phenotype both in vitro and in vivo Further domain analysis suggests that the amino half of MAP7 is responsible for branch formation, suggesting a mechanism that is independent of its known interaction with kinesin. Moreover, this mouse exhibits increased pain sensitivity, a phenotype that is consistent with increased collateral branch formation. Therefore, our study not only uncovers the first neuronal function of MAP7, but also demonstrates the importance of proper microtubule regulation in neural circuit development. Furthermore, our data provide new insights into microtubule regulation during axonal morphogenesis and may shed light on MAP7 function in neurological disorders.SIGNIFICANCE STATEMENT Neurons communicate with multiple targets by forming axonal branches. In search of intrinsic factors that control collateral branch development, we identified a role for microtubule-associated protein 7 (MAP7) in dorsal root ganglion sensory neurons. We show that MAP7 expression is developmentally regulated and perturbation of this expression alters branch formation. Cell biological analysis indicates that MAP7 promotes branch maturation. Analysis of a spontaneous mouse mutant suggests a molecular mechanism for branch regulation and the potential influence of collateral branches on pain sensitivity. Our studies thus establish the first neuronal function of MAP7 and demonstrate its role in branch morphogenesis and neural circuit function. These findings may help in our understanding of the contribution of MAP7 to neurological disorders and nerve regeneration.
Collapse
|
38
|
Drosophila melanogaster Neuroblasts: A Model for Asymmetric Stem Cell Divisions. Results Probl Cell Differ 2017; 61:183-210. [PMID: 28409305 DOI: 10.1007/978-3-319-53150-2_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Asymmetric cell division (ACD) is a fundamental mechanism to generate cell diversity, giving rise to daughter cells with different developmental potentials. ACD is manifested in the asymmetric segregation of proteins or mRNAs, when the two daughter cells differ in size or are endowed with different potentials to differentiate into a particular cell type (Horvitz and Herskowitz, Cell 68:237-255, 1992). Drosophila neuroblasts, the neural stem cells of the developing fly brain, are an ideal system to study ACD since this system encompasses all of these characteristics. Neuroblasts are intrinsically polarized cells, utilizing polarity cues to orient the mitotic spindle, segregate cell fate determinants asymmetrically, and regulate spindle geometry and physical asymmetry. The neuroblast system has contributed significantly to the elucidation of the basic molecular mechanisms underlying ACD. Recent findings also highlight its usefulness to study basic aspects of stem cell biology and tumor formation. In this review, we will focus on what has been learned about the basic mechanisms underlying ACD in fly neuroblasts.
Collapse
|
39
|
Ravidà A, Cwiklinski K, Aldridge AM, Clarke P, Thompson R, Gerlach JQ, Kilcoyne M, Hokke CH, Dalton JP, O'Neill SM. Fasciola hepatica Surface Tegument: Glycoproteins at the Interface of Parasite and Host. Mol Cell Proteomics 2016; 15:3139-3153. [PMID: 27466253 PMCID: PMC5054340 DOI: 10.1074/mcp.m116.059774] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Indexed: 11/20/2022] Open
Abstract
Fasciola hepatica, commonly known as liver fluke, is a trematode that causes Fasciolosis in ruminants and humans. The outer tegumental coat of F. hepatica (FhTeg) is a complex metabolically active biological matrix that is continually exposed to the host immune system and therefore makes a good vaccine target. F. hepatica tegumental coat is highly glycosylated and helminth-derived immunogenic oligosaccharide motifs and glycoproteins are currently being investigated as novel vaccine candidates. This report presents the first systematic characterization of FhTeg glycosylation using lectin microarrays to characterize carbohydrates motifs present, and lectin histochemistry to localize these on the F. hepatica tegument. We discovered that FhTeg glycoproteins are predominantly oligomannose oligosaccharides that are expressed on the spines, suckers and tegumental coat of F. hepatica and lectin blot analysis confirmed the abundance of N- glycosylated proteins. Although some oligosaccharides are widely distributed on the fluke surface other subsets are restricted to distinct anatomical regions. We selectively enriched for FhTeg mannosylated glycoprotein subsets using lectin affinity chromatography and identified 369 proteins by mass spectrometric analysis. Among these proteins are a number of potential vaccine candidates with known immune modulatory properties including proteases, protease inhibitors, paramyosin, Venom Allergen-like II, Enolase and two proteins, nardilysin and TRIL, that have not been previously associated with F. hepatica. Furthermore, we provide a comprehensive insight regarding the putative glycosylation of FhTeg components that could highlight the importance of further studies examining glycoconjugates in host-parasite interactions in the context of F. hepatica infection and the development of an effective vaccine.
Collapse
Affiliation(s)
- Alessandra Ravidà
- From the ‡Fundamental and Translational Immunology, School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Krystyna Cwiklinski
- §School of Biological Sciences, Medical Biology Centre (MBC), Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Allison M Aldridge
- From the ‡Fundamental and Translational Immunology, School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Paul Clarke
- ¶Glycoselect, Dublin City University, Glasnevin, Dublin 9
| | | | - Jared Q Gerlach
- ‖Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland; **Regenerative Medicine Institute, NUI Galway, Ireland
| | - Michelle Kilcoyne
- ‖Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland; ‡‡Carbohydrate Signalling Group, Microbiology, NUI Galway, Ireland
| | - Cornelis H Hokke
- §§Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - John P Dalton
- §School of Biological Sciences, Medical Biology Centre (MBC), Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sandra M O'Neill
- From the ‡Fundamental and Translational Immunology, School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin, Dublin 9, Ireland;
| |
Collapse
|
40
|
Oncogenic KRAS triggers MAPK-dependent errors in mitosis and MYC-dependent sensitivity to anti-mitotic agents. Sci Rep 2016; 6:29741. [PMID: 27412232 PMCID: PMC4944194 DOI: 10.1038/srep29741] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/23/2016] [Indexed: 12/21/2022] Open
Abstract
Oncogenic KRAS induces cell proliferation and transformation, but little is known about its effects on cell division. Functional genetic screens have recently revealed that cancer cell lines expressing oncogenic KRAS are sensitive to interference with mitosis, but neither the mechanism nor the uniformity of anti-mitotic drug sensitivity connected with mutant KRAS expression are yet clear. Here, we report that acute expression of oncogenic KRAS in HeLa cells induces mitotic delay and defects in chromosome segregation through mitogen-activated protein kinase (MAPK) pathway activation and de-regulated expression of several mitosis-related genes. These anomalies are accompanied by increased sensitivity to anti-mitotic agents, a phenotype dependent on the transcription factor MYC and its downstream target anti-apoptotic protein BCL-XL. Unexpectedly, we find no correlation between KRAS mutational status or MYC expression levels and anti-mitotic drug sensitivity when surveying a large database of anti-cancer drug responses. However, we report that the co-existence of KRAS mutations and high MYC expression predicts anti-mitotic drug sensitivity. Our findings reveal a novel function of oncogenic KRAS in regulating accurate mitotic progression and suggest new avenues to therapeutically target KRAS-mutant tumours and stratify patients in ongoing clinical trials of anti-mitotic drugs.
Collapse
|
41
|
Tegument Glycoproteins and Cathepsins of Newly Excysted Juvenile Fasciola hepatica Carry Mannosidic and Paucimannosidic N-glycans. PLoS Negl Trop Dis 2016; 10:e0004688. [PMID: 27139907 PMCID: PMC4854454 DOI: 10.1371/journal.pntd.0004688] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/14/2016] [Indexed: 11/19/2022] Open
Abstract
Recently, the prevalence of Fasciola hepatica in some areas has increased considerably and the availability of a vaccine to protect livestock from infection would represent a major advance in tools available for controlling this disease. To date, most vaccine-target discovery research on this parasite has concentrated on proteomic and transcriptomic approaches whereas little work has been carried out on glycosylation. As the F. hepatica tegument (Teg) may contain glycans potentially relevant to vaccine development and the Newly Excysted Juvenile (NEJ) is the first lifecycle stage in contact with the definitive host, our work has focused on assessing the glycosylation of the NEJTeg and identifying the NEJTeg glycoprotein repertoire. After in vitro excystation, NEJ were fixed and NEJTeg was extracted. Matrix-assisted laser desorption ionisation-time of flight-mass spectrometry (MALDI-TOF-MS) analysis of released N-glycans revealed that oligomannose and core-fucosylated truncated N-glycans were the most dominant glycan types. By lectin binding studies these glycans were identified mainly on the NEJ surface, together with the oral and ventral suckers. NEJTeg glycoproteins were affinity purified after targeted biotinylation of the glycans and identified using liquid chromatography and tandem mass spectrometry (LC-MS/MS). From the total set of proteins previously identified in NEJTeg, eighteen were also detected in the glycosylated fraction, including the F. hepatica Cathepsin B3 (FhCB3) and two of the Cathepsin L3 (FhCL3) proteins, among others. To confirm glycosylation of cathepsins, analysis at the glycopeptide level by LC-ESI-ion-trap-MS/MS with collision-induced dissociation (CID) and electron-transfer dissociation (ETD) was carried out. We established that cathepsin B1 (FhCB1) on position N80, and FhCL3 (BN1106_s10139B000014, scaffold10139) on position N153, carry unusual paucimannosidic Man2GlcNAc2 glycans. To our knowledge, this is the first description of F. hepatica NEJ glycosylation and the first report of N-glycosylation of F. hepatica cathepsins. The significance of these findings for immunological studies and vaccine development is discussed.
Collapse
|
42
|
Chen K, Koe CT, Xing ZB, Tian X, Rossi F, Wang C, Tang Q, Zong W, Hong WJ, Taneja R, Yu F, Gonzalez C, Wu C, Endow S, Wang H. Arl2- and Msps-dependent microtubule growth governs asymmetric division. J Cell Biol 2016; 212:661-76. [PMID: 26953351 PMCID: PMC4792071 DOI: 10.1083/jcb.201503047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 02/10/2016] [Indexed: 12/18/2022] Open
Abstract
Drosophila Arl2 governs neuroblast asymmetric cell division through regulation of microtubule growth and localization of Msps to centrosomes. Asymmetric division of neural stem cells is a fundamental strategy to balance their self-renewal and differentiation. It is long thought that microtubules are not essential for cell polarity in asymmetrically dividing Drosophila melanogaster neuroblasts (NBs; neural stem cells). Here, we show that Drosophila ADP ribosylation factor like-2 (Arl2) and Msps, a known microtubule-binding protein, control cell polarity and spindle orientation of NBs. Upon arl2 RNA intereference, Arl2-GDP expression, or arl2 deletions, microtubule abnormalities and asymmetric division defects were observed. Conversely, overactivation of Arl2 leads to microtubule overgrowth and depletion of NBs. Arl2 regulates microtubule growth and asymmetric division through localizing Msps to the centrosomes in NBs. Moreover, Arl2 regulates dynein function and in turn centrosomal localization of D-TACC and Msps. Arl2 physically associates with tubulin cofactors C, D, and E. Arl2 functions together with tubulin-binding cofactor D to control microtubule growth, Msps localization, and NB self-renewal. Therefore, Arl2- and Msps-dependent microtubule growth is a new paradigm regulating asymmetric division of neural stem cells.
Collapse
Affiliation(s)
- Keng Chen
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456
| | - Chwee Tat Koe
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456
| | - Zhanyuan Benny Xing
- Department of Cell Biology, Duke University, Duke University Medical Center, Durham, NC 27710
| | - Xiaolin Tian
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Fabrizio Rossi
- Institute for Research in Biomedicine Barcelona, 08028 Barcelona, Spain
| | - Cheng Wang
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857
| | - Quan Tang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Wenhui Zong
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Wan Jin Hong
- Institute of Molecular and Cell Biology, Singapore 138673
| | - Reshma Taneja
- National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Fengwei Yu
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Cayetano Gonzalez
- Institute for Research in Biomedicine Barcelona, 08028 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Chunlai Wu
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Sharyn Endow
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 Department of Cell Biology, Duke University, Duke University Medical Center, Durham, NC 27710
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
43
|
Caous R, Pascal A, Romé P, Richard-Parpaillon L, Karess R, Giet R. Spindle assembly checkpoint inactivation fails to suppress neuroblast tumour formation in aurA mutant Drosophila. Nat Commun 2015; 6:8879. [PMID: 26568519 PMCID: PMC4660220 DOI: 10.1038/ncomms9879] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/10/2015] [Indexed: 12/18/2022] Open
Abstract
Tissue homeostasis requires accurate control of cell proliferation, differentiation and chromosome segregation. Drosophila sas-4 and aurA mutants present brain tumours with extra neuroblasts (NBs), defective mitotic spindle assembly and delayed mitosis due to activation of the spindle assembly checkpoint (SAC). Here we inactivate the SAC in aurA and sas-4 mutants to determine whether the generation of aneuploidy compromises NB proliferation. Inactivation of the SAC in the sas-4 mutant impairs NB proliferation and disrupts euploidy. By contrast, disrupting the SAC in the aurA mutant does not prevent NB amplification, tumour formation or chromosome segregation. The monitoring of Mad2 and cyclin B dynamics in live aurA NBs reveals that SAC satisfaction is not coupled to cyclin B degradation. Thus, the NBs of aurA mutants present delayed mitosis, with accurate chromosome segregation occurring in a SAC-independent manner. We report here the existence of an Aurora A-dependent mechanism promoting efficient, timed cyclin B degradation.
Collapse
Affiliation(s)
- Renaud Caous
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Aude Pascal
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Pierre Romé
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Laurent Richard-Parpaillon
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Roger Karess
- Institut Jacques Monod-Université Paris Diderot-Paris 7, 15 rue Hélène Brion, 75205 Paris, France
| | - Régis Giet
- Institut de Génétique et Développement de Rennes-Université de Rennes I-CNRS- UMR 6290, 2 avenue du Pr Léon Bernard, 35043 Rennes, France
| |
Collapse
|