1
|
Tidei JJ, Oakes PW, Beach JR. Myosin 2 - A general contractor for the cytoskeleton. Curr Opin Cell Biol 2025; 94:102522. [PMID: 40319507 DOI: 10.1016/j.ceb.2025.102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
Cells derive their shape, and in turn much of their behavior, from the organization of the cytoskeleton. While a myriad of proteins contribute to the regulation and organization of this dynamic structure, two of the principal components are actin filaments, which provide the structure, and myosin motors, which generate the majority of the forces. Here we review recent results on the assembly and kinetics of non-muscle myosin 2, and highlight how the cellular environment modulates local myosin behavior and signaling.
Collapse
Affiliation(s)
- Joseph J Tidei
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W Oakes
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA.
| | - Jordan R Beach
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
2
|
Rolfs LA, Falat EJ, Gutzman JH. myh9b is a critical non-muscle myosin II encoding gene that interacts with myh9a and myh10 during zebrafish development in both compensatory and redundant pathways. G3 (BETHESDA, MD.) 2025; 15:jkae260. [PMID: 39503257 PMCID: PMC11708221 DOI: 10.1093/g3journal/jkae260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Non-muscle myosin (NMII) motor proteins have diverse developmental functions due to their roles in cell shape changes, cell migration, and cell adhesion. Zebrafish are an ideal vertebrate model system to study the NMII encoding myh genes and proteins due to high sequence homology, established gene editing tools, and rapid ex utero development. In humans, mutations in the NMII encoding MYH genes can lead to abnormal developmental processes and disease. This study utilized zebrafish myh9a, myh9b, and myh10 null mutants to examine potential genetic interactions and roles for each gene in development. It was determined that the myh9b gene is the most critical NMII encoding gene, as myh9b mutants develop pericardial edema and have a partially penetrant lethal phenotype, which was not observed in the other myh mutants. This study also established that genetic interactions occur between the zebrafish myh9a, myh9b, and myh10 genes where myh9b is required for the expression of both myh9a and myh10, and myh10 is required for the expression of myh9b. Additionally, protein analyses suggested that enhanced NMII protein stability in some mutant backgrounds may play a role in compensation. Finally, double mutant studies revealed different and more severe phenotypes at earlier time points than single mutants, suggesting roles for tissue specific genetic redundancy, and in some genotypes, haploinsufficiency. These mutants are the first in vivo models allowing for the study of complete loss of the NMIIA and NMIIB proteins, establishing them as valuable tools to elucidate the role of NMII encoding myh genes in development and disease.
Collapse
Affiliation(s)
- Laura A Rolfs
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Elizabeth J Falat
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Jennifer H Gutzman
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| |
Collapse
|
3
|
Wu K, Patel H, Wu H, Quintanilla MA, Bennett MA, Sala S, Beach JR. Non-muscle myosin 2 can incorporate into established filaments in cells without an assembly competence domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602405. [PMID: 39026829 PMCID: PMC11257479 DOI: 10.1101/2024.07.07.602405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Myosin 2 dynamically assembles into filaments that exert force on the actin cytoskeleton. To form filaments, myosin 2 monomers transition between folded and unfolded states. Monomer unfolding exposes an extended coiled-coil that interacts with other monomers in parallel and antiparallel fashions, enabling bipolar filament formation. A C-terminal domain of the coiled-coil, termed assembly competence domain (ACD), has been repeatedly identified as necessary for filament assembly. Here, we revisit ACD contribution when full-length filaments are present. Non-muscle myosin 2A lacking the ACD (ΔACD) initially appears diffuse, but triton extraction of cytosolic fraction reveals cytoskeletal association. Disruption of the folded monomer enhances the cytoskeletal fraction, while inhibition of endogenous filament assembly appears to reduce it. Finally, high resolution imaging of endogenous and exogenous bipolar filamentous structures reveals highly coincident signal, suggesting ΔACD constructs co-assemble with endogenous myosin 2A filaments. Our data demonstrate that while the ACD is required for de novo filament assembly, it is not required for monomers to recognize and associate with established filaments in cells. More broadly, this highlights the existence of distinct mechanisms governing myosin 2 monomer assembly into nascent filaments, and monomer recognition and association with established filaments to maintain steady-state contractile networks.
Collapse
Affiliation(s)
- Kehan Wu
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Hiral Patel
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Huini Wu
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Melissa A Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Margaret A Bennett
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Stefano Sala
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| |
Collapse
|
4
|
Wang L, Tsang HY, Yan Z, Tojkander S, Ciuba K, Kogan K, Liu X, Zhao H. LUZP1 regulates the maturation of contractile actomyosin bundles. Cell Mol Life Sci 2024; 81:248. [PMID: 38832964 PMCID: PMC11335285 DOI: 10.1007/s00018-024-05294-0] [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: 02/15/2024] [Revised: 05/07/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
Contractile actomyosin bundles play crucial roles in various physiological processes, including cell migration, morphogenesis, and muscle contraction. The intricate assembly of actomyosin bundles involves the precise alignment and fusion of myosin II filaments, yet the underlying mechanisms and factors involved in these processes remain elusive. Our study reveals that LUZP1 plays a central role in orchestrating the maturation of thick actomyosin bundles. Loss of LUZP1 caused abnormal cell morphogenesis, migration, and the ability to exert forces on the environment. Importantly, knockout of LUZP1 results in significant defects in the concatenation and persistent association of myosin II filaments, severely impairing the assembly of myosin II stacks. The disruption of these processes in LUZP1 knockout cells provides mechanistic insights into the defective assembly of thick ventral stress fibers and the associated cellular contractility abnormalities. Overall, these results significantly contribute to our understanding of the molecular mechanism involved in actomyosin bundle formation and highlight the essential role of LUZP1 in this process.
Collapse
Affiliation(s)
- Liang Wang
- Faculty of Biological and Environmental Sciences, University of Helsinki, FI-00014, Helsinki, Finland
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hoi Ying Tsang
- Faculty of Biological and Environmental Sciences, University of Helsinki, FI-00014, Helsinki, Finland
| | - Ziyi Yan
- Faculty of Biological and Environmental Sciences, University of Helsinki, FI-00014, Helsinki, Finland
| | - Sari Tojkander
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Katarzyna Ciuba
- Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Konstantin Kogan
- Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Xiaonan Liu
- Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Hongxia Zhao
- Faculty of Biological and Environmental Sciences, University of Helsinki, FI-00014, Helsinki, Finland.
| |
Collapse
|
5
|
Abstract
Non-muscle myosin 2 (NM2) motors are the major contractile machines in most cell types. Unsurprisingly, these ubiquitously expressed actin-based motors power a plethora of subcellular, cellular and multicellular processes. In this Cell Science at a Glance article and the accompanying poster, we review the biochemical properties and mechanisms of regulation of this myosin. We highlight the central role of NM2 in multiple fundamental cellular processes, which include cell migration, cytokinesis, epithelial barrier function and tissue morphogenesis. In addition, we highlight recent studies using advanced imaging technologies that have revealed aspects of NM2 assembly hitherto inaccessible. This article will hopefully appeal to both cytoskeletal enthusiasts and investigators from outside the cytoskeleton field who have interests in one of the many basic cellular processes requiring actomyosin force production.
Collapse
Affiliation(s)
- Melissa A. Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| | - John A. Hammer
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| |
Collapse
|
6
|
Weißenbruch K, Fladung M, Grewe J, Baulesch L, Schwarz US, Bastmeyer M. Nonmuscle myosin IIA dynamically guides regulatory light chain phosphorylation and assembly of nonmuscle myosin IIB. Eur J Cell Biol 2022; 101:151213. [DOI: 10.1016/j.ejcb.2022.151213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
|
7
|
Abstract
Background: Myosin 7a is an actin-binding motor protein involved in the formation of hair-cell stereocilia both in the cochlea and in the vestibular system. Mutations in myosin 7a are linked to congenital hearing loss and are present in 50% of Type-1 Usher syndrome patients who suffer from progressive hearing loss and vestibular system dysfunction. Methods: Myosin 7a is often used to visualise sensory hair cells due to its well characterised and localised expression profile. We thus conducted myosin-7a immunostaining across all three turns of the adult rat organ of Corti to visualise hair cells. Results: As expected, we observed myosin 7a staining in both inner and outer hair cells. Unexpectedly, we also observed strong myosin 7a staining in the medial olivocochlear efferent synaptic boutons contacting the outer hair cells. Efferent bouton myosin-7a staining was present across all three turns of the cochlea. We verified this localisation by co-staining with a known efferent bouton marker, the vesicular acetylcholine transporter. Conclusions: In addition to its role in stereocilia formation and maintenance, myosin 7a or certain myosin-7a expression variants might play a role in efferent synaptic transmission in the cochlea and thus ultimately influence cochlear gain regulation. Our immunohistochemistry results should be validated with other methods to confirm these serendipitous findings.
Collapse
Affiliation(s)
- Piotr Sirko
- Laboratory of Auditory Neuroscience and Biophysics, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Andrei S. Kozlov
- Laboratory of Auditory Neuroscience and Biophysics, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
8
|
Sirko P, Kozlov AS. Immunohistochemistry localises myosin-7a to cochlear efferent boutons. Wellcome Open Res 2022; 7:1. [PMID: 35224213 PMCID: PMC8866907 DOI: 10.12688/wellcomeopenres.17428.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 08/11/2023] Open
Abstract
Background: Myosin 7a is an actin-binding motor protein involved in the formation of hair-cell stereocilia both in the cochlea and in the vestibular system. Mutations in myosin 7a are linked to congenital hearing loss and are present in 50% of Type-1 Usher syndrome patients who suffer from progressive hearing loss and vestibular system dysfunction. Methods: Myosin 7a is often used to visualise sensory hair cells due to its well characterised and localised expression profile. We thus conducted myosin-7a immunostaining across all three turns of the adult rat organ of Corti to visualise hair cells. Results: As expected, we observed myosin 7a staining in both inner and outer hair cells. Unexpectedly, we also observed strong myosin 7a staining in the medial olivocochlear efferent synaptic boutons contacting the outer hair cells. Efferent bouton myosin-7a staining was present across all three turns of the cochlea. We verified this localisation by co-staining with a known efferent bouton marker, the vesicular acetylcholine transporter. Conclusions: In addition to its role in stereocilia formation and maintenance, myosin 7a or certain myosin-7a expression variants might play a role in efferent synaptic transmission in the cochlea and thus ultimately influence cochlear gain regulation. Our immunohistochemistry results should be validated with other methods to confirm these serendipitous findings.
Collapse
Affiliation(s)
- Piotr Sirko
- Laboratory of Auditory Neuroscience and Biophysics, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Andrei S. Kozlov
- Laboratory of Auditory Neuroscience and Biophysics, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
9
|
Yu-Kemp HC, Szymanski RA, Cortes DB, Gadda NC, Lillich ML, Maddox AS, Peifer M. Micron-scale supramolecular myosin arrays help mediate cytoskeletal assembly at mature adherens junctions. J Cell Biol 2022; 221:212872. [PMID: 34812842 PMCID: PMC8614156 DOI: 10.1083/jcb.202103074] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 01/19/2023] Open
Abstract
Epithelial cells assemble specialized actomyosin structures at E-Cadherin–based cell–cell junctions, and the force exerted drives cell shape change during morphogenesis. The mechanisms that build this supramolecular actomyosin structure remain unclear. We used ZO-knockdown MDCK cells, which assemble a robust, polarized, and highly organized actomyosin cytoskeleton at the zonula adherens, combining genetic and pharmacologic approaches with superresolution microscopy to define molecular machines required. To our surprise, inhibiting individual actin assembly pathways (Arp2/3, formins, or Ena/VASP) did not prevent or delay assembly of this polarized actomyosin structure. Instead, as junctions matured, micron-scale supramolecular myosin arrays assembled, with aligned stacks of myosin filaments adjacent to the apical membrane, overlying disorganized actin filaments. This suggested that myosin arrays might bundle actin at mature junctions. Consistent with this idea, inhibiting ROCK or myosin ATPase disrupted myosin localization/organization and prevented actin bundling and polarization. We obtained similar results in Caco-2 cells. These results suggest a novel role for myosin self-assembly, helping drive actin organization to facilitate cell shape change.
Collapse
Affiliation(s)
- Hui-Chia Yu-Kemp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rachel A Szymanski
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Daniel B Cortes
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nicole C Gadda
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Madeline L Lillich
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Amy S Maddox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mark Peifer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
10
|
Weißenbruch K, Grewe J, Hippler M, Fladung M, Tremmel M, Stricker K, Schwarz US, Bastmeyer M. Distinct roles of nonmuscle myosin II isoforms for establishing tension and elasticity during cell morphodynamics. eLife 2021; 10:71888. [PMID: 34374341 PMCID: PMC8391736 DOI: 10.7554/elife.71888] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022] Open
Abstract
Nonmuscle myosin II (NM II) is an integral part of essential cellular processes, including adhesion and migration. Mammalian cells express up to three isoforms termed NM IIA, B, and C. We used U2OS cells to create CRISPR/Cas9-based knockouts of all three isoforms and analyzed the phenotypes on homogenously coated surfaces, in collagen gels, and on micropatterned substrates. In contrast to homogenously coated surfaces, a structured environment supports a cellular phenotype with invaginated actin arcs even in the absence of NM IIA-induced contractility. A quantitative shape analysis of cells on micropatterns combined with a scale-bridging mathematical model reveals that NM IIA is essential to build up cellular tension during initial stages of force generation, while NM IIB is necessary to elastically stabilize NM IIA-generated tension. A dynamic cell stretch/release experiment in a three-dimensional scaffold confirms these conclusions and in addition reveals a novel role for NM IIC, namely the ability to establish tensional homeostasis.
Collapse
Affiliation(s)
- Kai Weißenbruch
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Justin Grewe
- Institute for Theoretical Physics, University of Heidelberg, Heidelberg, Germany.,BioQuant-Center for Quantitative Biology, University of Heidelberg, Heidelberg, Germany
| | - Marc Hippler
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Magdalena Fladung
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Moritz Tremmel
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Kathrin Stricker
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Ulrich Sebastian Schwarz
- Institute for Theoretical Physics, University of Heidelberg, Heidelberg, Germany.,BioQuant-Center for Quantitative Biology, University of Heidelberg, Heidelberg, Germany
| | - Martin Bastmeyer
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.,Institute for Biological and Chemical Systems - Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
11
|
Barvitenko N, Aslam M, Lawen A, Saldanha C, Skverchinskaya E, Uras G, Manca A, Pantaleo A. Two Motors and One Spring: Hypothetic Roles of Non-Muscle Myosin II and Submembrane Actin-Based Cytoskeleton in Cell Volume Sensing. Int J Mol Sci 2021; 22:7967. [PMID: 34360739 PMCID: PMC8347689 DOI: 10.3390/ijms22157967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Changes in plasma membrane curvature and intracellular ionic strength are two key features of cell volume perturbations. In this hypothesis we present a model of the responsible molecular apparatus which is assembled of two molecular motors [non-muscle myosin II (NMMII) and protrusive actin polymerization], a spring [a complex between the plasma membrane (PM) and the submembrane actin-based cytoskeleton (smACSK) which behaves like a viscoelastic solid] and the associated signaling proteins. We hypothesize that this apparatus senses changes in both the plasma membrane curvature and the ionic strength and in turn activates signaling pathways responsible for regulatory volume increase (RVI) and regulatory volume decrease (RVD). During cell volume changes hydrostatic pressure (HP) changes drive alterations in the cell membrane curvature. HP difference has opposite directions in swelling versus shrinkage, thus allowing distinction between them. By analogy with actomyosin contractility that appears to sense stiffness of the extracellular matrix we propose that NMMII and actin polymerization can actively probe the transmembrane gradient in HP. Furthermore, NMMII and protein-protein interactions in the actin cortex are sensitive to ionic strength. Emerging data on direct binding to and regulating activities of transmembrane mechanosensors by NMMII and actin cortex provide routes for signal transduction from transmembrane mechanosensors to cell volume regulatory mechanisms.
Collapse
Affiliation(s)
| | - Muhammad Aslam
- Department of Internal Medicine I, Experimental Cardiology, Justus Liebig University, 35392 Giessen, Germany;
| | - Alfons Lawen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia;
| | - Carlota Saldanha
- Institute of Biochemistry, Institute of Molecular Medicine, Faculty of Medicine University of Lisbon, 1649-028 Lisboa, Portugal;
| | | | - Giuseppe Uras
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK;
| | - Alessia Manca
- Department of Biomedical Science, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy;
| | - Antonella Pantaleo
- Department of Biomedical Science, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy;
| |
Collapse
|
12
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
13
|
Rogerson C, Wotherspoon DJ, Tommasi C, Button RW, O'Shaughnessy RFL. Akt1-associated actomyosin remodelling is required for nuclear lamina dispersal and nuclear shrinkage in epidermal terminal differentiation. Cell Death Differ 2021; 28:1849-1864. [PMID: 33462407 PMCID: PMC8184862 DOI: 10.1038/s41418-020-00712-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/30/2023] Open
Abstract
Keratinocyte cornification and epidermal barrier formation are tightly controlled processes, which require complete degradation of intracellular organelles, including removal of keratinocyte nuclei. Keratinocyte nuclear destruction requires Akt1-dependent phosphorylation and degradation of the nuclear lamina protein, Lamin A/C, essential for nuclear integrity. However, the molecular mechanisms that result in complete nuclear removal and their regulation are not well defined. Post-confluent cultures of rat epidermal keratinocytes (REKs) undergo spontaneous and complete differentiation, allowing visualisation and perturbation of the differentiation process in vitro. We demonstrate that there is dispersal of phosphorylated Lamin A/C to structures throughout the cytoplasm in differentiating keratinocytes. We show that the dispersal of phosphorylated Lamin A/C is Akt1-dependent and these structures are specific for the removal of Lamin A/C from the nuclear lamina; nuclear contents and Lamin B were not present in these structures. Immunoprecipitation identified a group of functionally related Akt1 target proteins involved in Lamin A/C dispersal, including actin, which forms cytoskeletal microfilaments, Arp3, required for actin filament nucleation, and Myh9, a component of myosin IIa, a molecular motor that can translocate along actin filaments. Disruption of actin filament polymerisation, nucleation or myosin IIa activity prevented formation and dispersal of cytoplasmic Lamin A/C structures. Live imaging of keratinocytes expressing fluorescently tagged nuclear proteins showed a nuclear volume reduction step taking less than 40 min precedes final nuclear destruction. Preventing Akt1-dependent Lamin A/C phosphorylation and disrupting cytoskeletal Akt1-associated proteins prevented nuclear volume reduction. We propose keratinocyte nuclear destruction and differentiation requires myosin II activity and the actin cytoskeleton for two intermediate processes: Lamin A/C dispersal and rapid nuclear volume reduction.
Collapse
Affiliation(s)
- Clare Rogerson
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Duncan J Wotherspoon
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cristina Tommasi
- Immunobiology and Dermatology, UCL Great Ormond Street Institute of Child Health, London, UK
- School of Cellular & Molecular Medicine, University of Bristol, Bristol, UK
| | - Robert W Button
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ryan F L O'Shaughnessy
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
14
|
Taneja N, Bersi MR, Baillargeon SM, Fenix AM, Cooper JA, Ohi R, Gama V, Merryman WD, Burnette DT. Precise Tuning of Cortical Contractility Regulates Cell Shape during Cytokinesis. Cell Rep 2021; 31:107477. [PMID: 32268086 DOI: 10.1016/j.celrep.2020.03.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/27/2020] [Accepted: 03/13/2020] [Indexed: 01/30/2023] Open
Abstract
The mechanical properties of the actin cortex regulate shape changes during cell division, cell migration, and tissue morphogenesis. We show that modulation of myosin II (MII) filament composition allows tuning of surface tension at the cortex to maintain cell shape during cytokinesis. Our results reveal that MIIA generates cortex tension, while MIIB acts as a stabilizing motor and its inclusion in MII hetero-filaments reduces cortex tension. Tension generation by MIIA drives faster cleavage furrow ingression and bleb formation. We also show distinct roles for the motor and tail domains of MIIB in maintaining cytokinetic fidelity. Maintenance of cortical stability by the motor domain of MIIB safeguards against shape instability-induced chromosome missegregation, while its tail domain mediates cortical localization at the terminal stages of cytokinesis to mediate cell abscission. Because most non-muscle contractile systems are cortical, this tuning mechanism will likely be applicable to numerous processes driven by myosin-II contractility.
Collapse
Affiliation(s)
- Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Sophie M Baillargeon
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - James A Cooper
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
15
|
Abstract
In this report, we describe how endothelial cells, the cells lining the interior of blood vessels, invade into tissues to form new vessels through sprouting angiogenesis. We found that endothelial cells use a specific lamellipodia-related membrane protrusion for invasion, which we termed dactylopodia. These protrusions have a special morphology, originate from filopodia, are linked to membrane-ruffling activity, and are specialized in invading into avascular extracellular matrix. Our work lays the foundations for drug discovery targeting sprouting angiogenesis. Sprouting angiogenesis is fundamental for development and contributes to cancer, diabetic retinopathy, and cardiovascular diseases. Sprouting angiogenesis depends on the invasive properties of endothelial tip cells. However, there is very limited knowledge on how tip cells invade into tissues. Here, we show that endothelial tip cells use dactylopodia as the main cellular protrusion for invasion into nonvascular extracellular matrix. We show that dactylopodia and filopodia protrusions are balanced by myosin IIA (NMIIA) and actin-related protein 2/3 (Arp2/3) activity. Endothelial cell-autonomous ablation of NMIIA promotes excessive dactylopodia formation in detriment of filopodia. Conversely, endothelial cell-autonomous ablation of Arp2/3 prevents dactylopodia development and leads to excessive filopodia formation. We further show that NMIIA inhibits Rac1-dependent activation of Arp2/3 by regulating the maturation state of focal adhesions. Our discoveries establish a comprehensive model of how endothelial tip cells regulate its protrusive activity and will pave the way toward strategies to block invasive tip cells during sprouting angiogenesis.
Collapse
|
16
|
Wang K, Okada H, Bi E. Comparative Analysis of the Roles of Non-muscle Myosin-IIs in Cytokinesis in Budding Yeast, Fission Yeast, and Mammalian Cells. Front Cell Dev Biol 2020; 8:593400. [PMID: 33330476 PMCID: PMC7710916 DOI: 10.3389/fcell.2020.593400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/30/2020] [Indexed: 12/31/2022] Open
Abstract
The contractile ring, which plays critical roles in cytokinesis in fungal and animal cells, has fascinated biologists for decades. However, the basic question of how the non-muscle myosin-II and actin filaments are assembled into a ring structure to drive cytokinesis remains poorly understood. It is even more mysterious why and how the budding yeast Saccharomyces cerevisiae, the fission yeast Schizosaccharomyces pombe, and humans construct the ring structure with one, two, and three myosin-II isoforms, respectively. Here, we provide a comparative analysis of the roles of the non-muscle myosin-IIs in cytokinesis in these three model systems, with the goal of defining the common and unique features and highlighting the major questions regarding this family of proteins.
Collapse
Affiliation(s)
- Kangji Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hiroki Okada
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
17
|
Costa AR, Sousa MM. Non-Muscle Myosin II in Axonal Cell Biology: From the Growth Cone to the Axon Initial Segment. Cells 2020; 9:cells9091961. [PMID: 32858875 PMCID: PMC7563147 DOI: 10.3390/cells9091961] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
By binding to actin filaments, non-muscle myosin II (NMII) generates actomyosin networks that hold unique contractile properties. Their dynamic nature is essential for neuronal biology including the establishment of polarity, growth cone formation and motility, axon growth during development (and axon regeneration in the adult), radial and longitudinal axonal tension, and synapse formation and function. In this review, we discuss the current knowledge on the spatial distribution and function of the actomyosin cytoskeleton in different axonal compartments. We highlight some of the apparent contradictions and open questions in the field, including the role of NMII in the regulation of axon growth and regeneration, the possibility that NMII structural arrangement along the axon shaft may control both radial and longitudinal contractility, and the mechanism and functional purpose underlying NMII enrichment in the axon initial segment. With the advances in live cell imaging and super resolution microscopy, it is expected that in the near future the spatial distribution of NMII in the axon, and the mechanisms by which it participates in axonal biology will be further untangled.
Collapse
|
18
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
19
|
Tang VW. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol Biol Cell 2020; 31:1823-1834. [PMID: 32730166 PMCID: PMC7525820 DOI: 10.1091/mbc.e19-12-0709] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The emergence of collagen I in vertebrates resulted in a dramatic increase in the stiffness of the extracellular environment, supporting long-range force propagation and the development of low-compliant tissues necessary for the development of vertebrate traits including pressurized circulation and renal filtration. Vertebrates have also evolved integrins that can bind to collagens, resulting in the generation of higher tension and more efficient force transmission in the extracellular matrix. The stiffer environment provides an opportunity for the vertebrates to create new structures such as the stress fibers, new cell types such as endothelial cells, new developmental processes such as neural crest delamination, and new tissue organizations such as the blood-brain barrier. Molecular players found only in vertebrates allow the modification of conserved mechanisms as well as the design of novel strategies that can better serve the physiological needs of the vertebrates. These innovations collectively contribute to novel morphogenetic behaviors and unprecedented increases in the complexities of tissue mechanics and functions.
Collapse
Affiliation(s)
- Vivian W. Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana–Champaign, Urbana, IL 61801
| |
Collapse
|
20
|
Asensio-Juárez G, Llorente-González C, Vicente-Manzanares M. Linking the Landscape of MYH9-Related Diseases to the Molecular Mechanisms that Control Non-Muscle Myosin II-A Function in Cells. Cells 2020; 9:E1458. [PMID: 32545517 PMCID: PMC7348894 DOI: 10.3390/cells9061458] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
The MYH9 gene encodes the heavy chain (MHCII) of non-muscle myosin II A (NMII-A). This is an actin-binding molecular motor essential for development that participates in many crucial cellular processes such as adhesion, cell migration, cytokinesis and polarization, maintenance of cell shape and signal transduction. Several types of mutations in the MYH9 gene cause an array of autosomal dominant disorders, globally known as MYH9-related diseases (MYH9-RD). These include May-Hegglin anomaly (MHA), Epstein syndrome (EPS), Fechtner syndrome (FTS) and Sebastian platelet syndrome (SPS). Although caused by different MYH9 mutations, all patients present macrothrombocytopenia, but may later display other pathologies, including loss of hearing, renal failure and presenile cataracts. The correlation between the molecular and cellular effects of the different mutations and clinical presentation are beginning to be established. In this review, we correlate the defects that MYH9 mutations cause at a molecular and cellular level (for example, deficient filament formation, altered ATPase activity or actin-binding) with the clinical presentation of the syndromes in human patients. We address why these syndromes are tissue restricted, and the existence of possible compensatory mechanisms, including residual activity of mutant NMII-A and/ or the formation of heteropolymers or co-polymers with other NMII isoforms.
Collapse
Affiliation(s)
| | | | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain; (G.A.-J.); (C.L.-G.)
| |
Collapse
|
21
|
Diverse roles of non-muscle myosin II contractility in 3D cell migration. Essays Biochem 2020; 63:497-508. [PMID: 31551323 DOI: 10.1042/ebc20190026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/15/2019] [Accepted: 08/27/2019] [Indexed: 01/13/2023]
Abstract
All is flux, nothing stays still. Heraclitus of Ephesus' characterization of the universe holds true for cells within animals and for proteins within cells. In this review, we examine the dynamics of actin and non-muscle myosin II within cells, and how their dynamics power the movement of cells within tissues. The 3D environment that migrating cells encounter along their path also changes over time, and cells can adopt various mechanisms of motility, depending on the topography, mechanics and chemical composition of their surroundings. We describe the differential spatio-temporal regulation of actin and myosin II-mediated contractility in mesenchymal, lobopodial, amoeboid, and swimming modes of cell migration. After briefly reviewing the biochemistry of myosin II, we discuss the role actomyosin contractility plays in the switch between modes of 3D migration that cells use to adapt to changing environments.
Collapse
|
22
|
Wang J, Fan Y, Sanger JM, Sanger JW. Nonmuscle myosin II in cardiac and skeletal muscle cells. Cytoskeleton (Hoboken) 2019; 75:339-351. [PMID: 29781105 DOI: 10.1002/cm.21454] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/27/2018] [Accepted: 05/05/2018] [Indexed: 11/08/2022]
Abstract
De novo assembly of contractile myofibrils begins with the formation of premyofibrils where filaments of non-muscle myosin (NM II), and actin organize in sarcomeric patterns with Z-Bodies containing muscle-specific alpha-actinin. Interactions of muscle specific myosin (MM II) with NM II occur in a nascent myofibril stage that precedes the assembly of mature myofibrils. By the final stage of myofibrillogenesis, the only myosin II present in the mature myofibrils is MM II. In this current study of myofibril assembly, the three vertebrate isoforms of NM II (A, B, and C) and sarcomeric alpha-actinin, ligated to GFP family proteins, were coexpressed in avian embryonic skeletal and cardiac muscle cells. Each isoform of NM II localized only in the mini-A-Bands of premyofibrils and nascent myofibrils. There was no evidence of localization of NM II in Z-Bodies of premyofibrils and nascent myofibrils or in Z-Bands of mature myofibrils. Fluorescence Recovery After Photobleaching (FRAP) experiments indicated similar exchange rates in premyofibrils for NM II isoforms A and B, whereas the IIC isoform was significantly less dynamic. Fluorescence Resonance Energy Transfer (FRET) measurements of colocalized fluorescent pairs of different NM II isoforms yielded signals similar to identical pairs, indicating copolymerization of the different NM II pairs. The role of NM II may reside in establishing the future sarcomere pattern in mature myofibrils by binding to the oppositely polarized actin filaments that extend between pairs of Z-Bodies along premyofibrils prior to their transformation into mature myofibrils.
Collapse
Affiliation(s)
- Jushuo Wang
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Yingli Fan
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Jean M Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Joseph W Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| |
Collapse
|
23
|
Heuzé ML, Sankara Narayana GHN, D'Alessandro J, Cellerin V, Dang T, Williams DS, Van Hest JC, Marcq P, Mège RM, Ladoux B. Myosin II isoforms play distinct roles in adherens junction biogenesis. eLife 2019; 8:46599. [PMID: 31486768 PMCID: PMC6756789 DOI: 10.7554/elife.46599] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022] Open
Abstract
Adherens junction (AJ) assembly under force is essential for many biological processes like epithelial monolayer bending, collective cell migration, cell extrusion and wound healing. The acto-myosin cytoskeleton acts as a major force-generator during the de novo formation and remodeling of AJ. Here, we investigated the role of non-muscle myosin II isoforms (NMIIA and NMIIB) in epithelial junction assembly. NMIIA and NMIIB differentially regulate biogenesis of AJ through association with distinct actin networks. Analysis of junction dynamics, actin organization, and mechanical forces of control and knockdown cells for myosins revealed that NMIIA provides the mechanical tugging force necessary for cell-cell junction reinforcement and maintenance. NMIIB is involved in E-cadherin clustering, maintenance of a branched actin layer connecting E-cadherin complexes and perijunctional actin fibres leading to the building-up of anisotropic stress. These data reveal unanticipated complementary functions of NMIIA and NMIIB in the biogenesis and integrity of AJ.
Collapse
Affiliation(s)
- Mélina L Heuzé
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | | | - Joseph D'Alessandro
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Victor Cellerin
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Tien Dang
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - David S Williams
- Department of Chemistry, College of Science, Swansea University, Swansea, United Kingdom
| | - Jan Cm Van Hest
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Philippe Marcq
- Laboratoire Physique et Mécanique des Milieux Hétérogènes, Sorbonne Université and CNRS UMR 7636, Paris, France
| | - René-Marc Mège
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| | - Benoit Ladoux
- Institut Jacques Monod, Université de Paris and CNRS UMR 7592, Paris, France
| |
Collapse
|
24
|
Shutova MS, Svitkina TM. Common and Specific Functions of Nonmuscle Myosin II Paralogs in Cells. BIOCHEMISTRY (MOSCOW) 2019; 83:1459-1468. [PMID: 30878021 DOI: 10.1134/s0006297918120040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Various forms of cell motility critically depend on pushing, pulling, and resistance forces generated by the actin cytoskeleton. Whereas pushing forces largely depend on actin polymerization, pulling forces responsible for cell contractility and resistance forces maintaining the cell shape require interaction of actin filaments with the multivalent molecular motor myosin II. In contrast to muscle-specific myosin II paralogs, nonmuscle myosin II (NMII) functions in virtually all mammalian cells, where it executes numerous mechanical tasks. NMII is expressed in mammalian cells as a tissue-specific combination of three paralogs, NMIIA, NMIIB, and NMIIC. Despite overall similarity, these paralogs differ in their molecular properties, which allow them to play both unique and common roles. Importantly, the three paralogs can also cooperate with each other by mixing and matching their unique capabilities. Through specialization and cooperation, NMII paralogs together execute a great variety of tasks in many different cell types. Here, we focus on mammalian NMII paralogs and review novel aspects of their kinetics, regulation, and functions in cells from the perspective of how distinct features of the three myosin II paralogs adapt them to perform specialized and joint tasks in the cells.
Collapse
Affiliation(s)
- M S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - T M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
25
|
Wang K, Wloka C, Bi E. Non-muscle Myosin-II Is Required for the Generation of a Constriction Site for Subsequent Abscission. iScience 2019; 13:69-81. [PMID: 30825839 PMCID: PMC6396101 DOI: 10.1016/j.isci.2019.02.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 01/07/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
It remains unknown when, where, and how the site of abscission is generated during cytokinesis. Here, we show that the sites of constriction, i.e., the sites of future abscission, are initially formed at the ends of the intercellular bridge during early midbody stage, and that these sites are associated with the non-muscle myosin-IIB (not myosin-IIA), actin filaments, and septin 9 until abscission. The ESCRT-III component CHMP4B localizes to the midbody and "spreads" to the site of abscission only during late midbody stage. Strikingly, inhibition of myosin-II motor activity by a low dose of Blebbistatin completely abolishes the formation of the constriction sites, resulting in the localization of all the above-mentioned components to the midbody region. These data strongly suggest that a secondary actomyosin ring provides the primary driving force for the thinning of the intercellular bridge to allow ESCRT-mediated membrane fission.
Collapse
Affiliation(s)
- Kangji Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Carsten Wloka
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA; Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AE Groningen, the Netherlands.
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA.
| |
Collapse
|
26
|
Yamamoto K, Otomo K, Nemoto T, Ishihara S, Haga H, Nagasaki A, Murakami Y, Takahashi M. Differential contributions of nonmuscle myosin IIA and IIB to cytokinesis in human immortalized fibroblasts. Exp Cell Res 2019; 376:67-76. [PMID: 30711568 DOI: 10.1016/j.yexcr.2019.01.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 01/28/2023]
Abstract
Nonmuscle myosin II (NMII) plays an important role in cytokinesis by constricting a contractile ring. However, it is poorly understood how NMII isoforms contribute to cytokinesis in mammalian cells. Here, we investigated the roles of the two major NMII isoforms, NMIIA and NMIIB, in cytokinesis using a WI-38 VA13 cell line (human immortalized fibroblast). In this cell line, NMIIB tended to localize to the contractile ring more than NMIIA. The expression level of NMIIA affected the localization of NMIIB. Most NMIIB accumulated at the cleavage furrow in NMIIA-knockout (KO) cells, and most NMIIA was displaced from this location in exogenous NMIIB-expressing cells, indicating that NMIIB preferentially localizes to the contractile ring. Specific KO of each isoform elicited opposite effects. The rate of furrow ingression was decreased and increased in NMIIA-KO and NMIIB-KO cells, respectively. Meanwhile, the length of NMII-filament stacks in the contractile ring was increased and decreased in NMIIA-KO and NMIIB-KO cells, respectively. Moreover, NMIIA helped to maintain cortical stiffness during cytokinesis. These findings suggest that appropriate ratio of NMIIA and NMIIB in the contractile ring is important for proper cytokinesis in specific cell types. In addition, two-photon excitation spinning-disk confocal microscopy enabled us to image constriction of the contractile ring in live cells in a three-dimensional manner.
Collapse
Affiliation(s)
- Kei Yamamoto
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Kohei Otomo
- Research Institute for Electronic Science, Hokkaido University, Sapporo 001-0020, Japan
| | - Tomomi Nemoto
- Research Institute for Electronic Science, Hokkaido University, Sapporo 001-0020, Japan
| | - Seiichiro Ishihara
- Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Hisashi Haga
- Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Akira Nagasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8562, Japan
| | - Yota Murakami
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan; Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Masayuki Takahashi
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan; Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan.
| |
Collapse
|
27
|
Pecci A, Ma X, Savoia A, Adelstein RS. MYH9: Structure, functions and role of non-muscle myosin IIA in human disease. Gene 2018; 664:152-167. [PMID: 29679756 PMCID: PMC5970098 DOI: 10.1016/j.gene.2018.04.048] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
The MYH9 gene encodes the heavy chain of non-muscle myosin IIA, a widely expressed cytoplasmic myosin that participates in a variety of processes requiring the generation of intracellular chemomechanical force and translocation of the actin cytoskeleton. Non-muscle myosin IIA functions are regulated by phosphorylation of its 20 kDa light chain, of the heavy chain, and by interactions with other proteins. Variants of MYH9 cause an autosomal-dominant disorder, termed MYH9-related disease, and may be involved in other conditions, such as chronic kidney disease, non-syndromic deafness, and cancer. This review discusses the structure of the MYH9 gene and its protein, as well as the regulation and physiologic functions of non-muscle myosin IIA with particular reference to embryonic development. Moreover, the review focuses on current knowledge about the role of MYH9 variants in human disease.
Collapse
Affiliation(s)
- Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Piazzale Golgi, 27100 Pavia, Italy.
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10 Room 6C-103B, 10 Center Drive, Bethesda, MD 20892-1583, USA.
| | - Anna Savoia
- Department of Medical Sciences, University of Trieste, via Dell'Istria, 65/1, I-34137 Trieste, Italy; IRCCS Burlo Garofolo, via Dell'Istria, 65/1, I-34137 Trieste, Italy.
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg. 10 Room 6C-103B, 10 Center Drive, Bethesda, MD 20892-1583, USA.
| |
Collapse
|
28
|
Abstract
( S)-Blebbistatin, a chiral tetrahydropyrroloquinolinone, is a widely used and well-characterized ATPase inhibitor selective for myosin II. The central role of myosin II in many normal and pathological biological processes has been revealed with the aid of this small molecule. The first part of this manuscript provides a summary of myosin II and ( S)-blebbistatin literature from a medicinal chemist's perspective. The second part of this perspective deals with the physicochemical deficiencies that trouble the use of ( S)-blebbistatin in advanced biological settings: low potency and solubility, fluorescence interference, (photo)toxicity, and stability issues. A large toolbox of analogues has been developed in which particular shortcomings have been addressed. This perspective provides a necessary overview of these developments and presents guidelines for selecting the best available analogue for a given application. As the unmet need for high-potency analogues remains, we also propose starting points for medicinal chemists in search of nanomolar myosin II inhibitors.
Collapse
|
29
|
Kuragano M, Uyeda TQP, Kamijo K, Murakami Y, Takahashi M. Different contributions of nonmuscle myosin IIA and IIB to the organization of stress fiber subtypes in fibroblasts. Mol Biol Cell 2018; 29:911-922. [PMID: 29467250 PMCID: PMC5896930 DOI: 10.1091/mbc.e17-04-0215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 12/25/2022] Open
Abstract
We demonstrated that myosin IIA and IIB are essential for the formation of transverse arcs and ventral stress fibers, respectively. Furthermore, we illustrated the roles of both isoforms in lamellar flattening and also raised the possibility that actin filaments in ventral stress fibers are in a stretched conformation.
Collapse
Affiliation(s)
- Masahiro Kuragano
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
| | - Taro Q. P. Uyeda
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Keiju Kamijo
- Department of Anatomy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Yota Murakami
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Masayuki Takahashi
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| |
Collapse
|
30
|
Shutova MS, Svitkina TM. Mammalian nonmuscle myosin II comes in three flavors. Biochem Biophys Res Commun 2018; 506:394-402. [PMID: 29550471 DOI: 10.1016/j.bbrc.2018.03.103] [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: 03/05/2018] [Accepted: 03/13/2018] [Indexed: 12/16/2022]
Abstract
Nonmuscle myosin II is an actin-based motor that executes numerous mechanical tasks in cells including spatiotemporal organization of the actin cytoskeleton, adhesion, migration, cytokinesis, tissue remodeling, and membrane trafficking. Nonmuscle myosin II is ubiquitously expressed in mammalian cells as a tissue-specific combination of three paralogs. Recent studies reveal novel specific aspects of their kinetics, intracellular regulation and functions. On the other hand, the three paralogs also can copolymerize and cooperate in cells. Here we review the recent advances from the prospective of how distinct features of the three myosin II paralogs adapt them to perform specialized and joint tasks in the cell.
Collapse
Affiliation(s)
- Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
31
|
Nonmuscle myosin IIA and IIB differentially contribute to intrinsic and directed migration of human embryonic lung fibroblasts. Biochem Biophys Res Commun 2018; 498:25-31. [DOI: 10.1016/j.bbrc.2018.02.171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 02/22/2018] [Indexed: 11/19/2022]
|
32
|
Abstract
The actin cytoskeleton-a collection of actin filaments with their accessory and regulatory proteins-is the primary force-generating machinery in the cell. It can produce pushing (protrusive) forces through coordinated polymerization of multiple actin filaments or pulling (contractile) forces through sliding actin filaments along bipolar filaments of myosin II. Both force types are particularly important for whole-cell migration, but they also define and change the cell shape and mechanical properties of the cell surface, drive the intracellular motility and morphogenesis of membrane organelles, and allow cells to form adhesions with each other and with the extracellular matrix.
Collapse
Affiliation(s)
- Tatyana Svitkina
- Department of Biology, University of Pennsylvania, 221 Leidy Labs, Philadelphia, Pennsylvania 19104
| |
Collapse
|
33
|
Lehtimäki JI, Fenix AM, Kotila TM, Balistreri G, Paavolainen L, Varjosalo M, Burnette DT, Lappalainen P. UNC-45a promotes myosin folding and stress fiber assembly. J Cell Biol 2017; 216:4053-4072. [PMID: 29055011 PMCID: PMC5716280 DOI: 10.1083/jcb.201703107] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/28/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022] Open
Abstract
Contractile actomyosin bundles, stress fibers, are crucial for adhesion, morphogenesis, and mechanosensing in nonmuscle cells. However, the mechanisms by which nonmuscle myosin II (NM-II) is recruited to those structures and assembled into functional bipolar filaments have remained elusive. We report that UNC-45a is a dynamic component of actin stress fibers and functions as a myosin chaperone in vivo. UNC-45a knockout cells display severe defects in stress fiber assembly and consequent abnormalities in cell morphogenesis, polarity, and migration. Experiments combining structured-illumination microscopy, gradient centrifugation, and proteasome inhibition approaches revealed that a large fraction of NM-II and myosin-1c molecules fail to fold in the absence of UNC-45a. The remaining properly folded NM-II molecules display defects in forming functional bipolar filaments. The C-terminal UNC-45/Cro1/She4p domain of UNC-45a is critical for NM-II folding, whereas the N-terminal tetratricopeptide repeat domain contributes to the assembly of functional stress fibers. Thus, UNC-45a promotes generation of contractile actomyosin bundles through synchronized NM-II folding and filament-assembly activities.
Collapse
Affiliation(s)
| | - Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Tommi M Kotila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Giuseppe Balistreri
- Department of Biosciences, Division of General Microbiology, University of Helsinki, Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Zhu Z, Peng L, Chen G, Jiang W, Shen Z, Du C, Zang R, Su Y, Xie H, Li H, Xia Y, Tang W. Mutations of MYH14 are associated to anorectal malformations with recto-perineal fistulas in a small subset of Chinese population. Clin Genet 2017; 92:503-509. [PMID: 28191911 DOI: 10.1111/cge.12993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/18/2017] [Accepted: 02/05/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND Anorectal malformations (ARMs) are among the most commonly congenital abnormalities of distal hindgut development, ranging from anal stenosis to anal atresia with or without fistulas and persistent cloaca. The etiology remains elusive for most ARM cases and the majority of genetic studies on ARMs were based on a candidate gene approach. MATERIALS AND METHODS In all eight family members of a non-consanguineous Chinese family, we performed whole-exome sequencing. Subsequently, exome sequencing of MYH14 in 72 unrelated probands with ARMs was performed. The accurate distribution of non-muscle myosin II heavy chain (NMHC II) was investigated by immunohistochemistry in serial sagittal sections of E11.5-13.5 mouse cloacal regions. RESULTS A homozygous mutation in MYH14 was identified in the two siblings of family 1. Compound heterozygous MYH14 changes were identified in an unrelated individual. Immunohistochemical analysis suggest stronger NMHC IIC localization in the epithelium of the murine embryonic cloaca, urorectal septum and hindgut compared with another two NMHC II isoforms. CONCLUSION This is the first identification of mutations in MYH14 as a cause of ARMs. The stronger localization of NMHC IIC in E11.5-13.5 mouse cloacal regions further supports the role of MYH14 in anorectal development.
Collapse
Affiliation(s)
- Zhongxian Zhu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Peng
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Guanglin Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Jiang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ziyang Shen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Chunxia Du
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rujin Zang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Su
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology (Nanjing Medical University), Ministry of Education, Nanjing, China
| | - Hua Xie
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongxing Li
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Modern Toxicology (Nanjing Medical University), Ministry of Education, Nanjing, China
| | - Weibing Tang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Soriano-Castell D, Chavero A, Rentero C, Bosch M, Vidal-Quadras M, Pol A, Enrich C, Tebar F. ROCK1 is a novel Rac1 effector to regulate tubular endocytic membrane formation during clathrin-independent endocytosis. Sci Rep 2017; 7:6866. [PMID: 28761175 PMCID: PMC5537229 DOI: 10.1038/s41598-017-07130-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 06/23/2017] [Indexed: 01/10/2023] Open
Abstract
Clathrin-dependent and -independent pathways contribute for β1-integrin endocytosis. This study defines a tubular membrane clathrin-independent endocytic network, induced with the calmodulin inhibitor W13, for β1-integrin internalization. This pathway is dependent on increased phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) levels and dynamin activity at the plasma membrane. Exogenous addition of PI(4,5)P2 or phosphatidylinositol-4-phosphate 5-kinase (PIP5K) expression mimicked W13-generated-tubules which are inhibited by active Rac1. Therefore, the molecular mechanisms downstream of Rac1, that controls this plasma membrane tubulation, were analyzed biochemically and by the expression of different Rac1 mutants. The results indicate that phospholipase C and ROCK1 are the main Rac1 effectors that impair plasma membrane invagination and tubule formation, essentially by decreasing PI(4,5)P2 levels and promoting cortical actomyosin assembly respectively. Interestingly, among the plethora of proteins that participate in membrane remodeling, this study revealed that ROCK1, the well-known downstream RhoA effector, has an important role in Rac1 regulation of actomyosin at the cell cortex. This study provides new insights into Rac1 functioning on plasma membrane dynamics combining phosphatidylinositides and cytoskeleton regulation.
Collapse
Affiliation(s)
- David Soriano-Castell
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Albert Chavero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Marta Bosch
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Maite Vidal-Quadras
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Albert Pol
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
| |
Collapse
|
36
|
Shutova MS, Asokan SB, Talwar S, Assoian RK, Bear JE, Svitkina TM. Self-sorting of nonmuscle myosins IIA and IIB polarizes the cytoskeleton and modulates cell motility. J Cell Biol 2017; 216:2877-2889. [PMID: 28701425 PMCID: PMC5584186 DOI: 10.1083/jcb.201705167] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/21/2023] Open
Abstract
Nonmuscle myosin II (NMII) is uniquely responsible for cell contractility and thus defines multiple aspects of cell behavior. To generate contraction, NMII molecules polymerize into bipolar minifilaments. Different NMII paralogs are often coexpressed in cells and can copolymerize, suggesting that they may cooperate to facilitate cell motility. However, whether such cooperation exists and how it may work remain unknown. We show that copolymerization of NMIIA and NMIIB followed by their differential turnover leads to self-sorting of NMIIA and NMIIB along the front-rear axis, thus producing a polarized actin-NMII cytoskeleton. Stress fibers newly formed near the leading edge are enriched in NMIIA, but over time, they become progressively enriched with NMIIB because of faster NMIIA turnover. In combination with retrograde flow, this process results in posterior accumulation of more stable NMIIB-rich stress fibers, thus strengthening cell polarity. By copolymerizing with NMIIB, NMIIA accelerates the intrinsically slow NMIIB dynamics, thus increasing cell motility and traction and enabling chemotaxis.
Collapse
Affiliation(s)
- Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Sreeja B Asokan
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shefali Talwar
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - James E Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | |
Collapse
|
37
|
Chi Q, Shan J, Ding X, Yin T, Wang Y, Jia D, Wang G. Smart mechanosensing machineries enable migration of vascular smooth muscle cells in atherosclerosis-relevant 3D matrices. Cell Biol Int 2017; 41:586-598. [PMID: 28328100 DOI: 10.1002/cbin.10764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/19/2017] [Indexed: 11/05/2022]
Abstract
At the early stage of atherosclerosis, neointima is formed due to the migration of vascular smooth muscle cells (VSMCs) from the media to the intima. VSMCs are surrounded by highly adhesive 3D matrices. They take specific strategies to cross various 3D matrices in the media, including heterogeneous collagen and mechanically strong basement membrane. Migration of VSMCs is potentially caused by biomechanical mechanism. Most in vitro studies focus on cell migration on 2D substrates in response to biochemical factors. How the cells move through 3D matrices under the action of mechanosensing machineries remains unexplored. In this review, we propose that several interesting tension-dependent machineries act as "tractor"-posterior myosin II accumulation, and "wrecker"-anterior podosome maintaining, to power VSMCs ahead. VSMCs embedded in 3D matrices may accumulate a minor myosin II isoform, myosin IIB, at the cell rear. Anisotropic myosin IIB distribution creates cell rear, polarizes cell body, pushes the nucleus and reshapes the cell body, and cooperates with a uniformly distributed myosin IIA to propel the cell forward. On the other hand, matrix digestion by podosome further promote the migration when the matrix becomes denser. Actomyosin tension activates Src to induce podosome in soft 3D matrices and retain the podosome integrity to steadily digest the matrix.
Collapse
Affiliation(s)
- Qingjia Chi
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jieling Shan
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiaorong Ding
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Yazhou Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Dongyu Jia
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
38
|
Lin YH, Zhen YY, Chien KY, Lee IC, Lin WC, Chen MY, Pai LM. LIMCH1 regulates nonmuscle myosin-II activity and suppresses cell migration. Mol Biol Cell 2017; 28:1054-1065. [PMID: 28228547 PMCID: PMC5391182 DOI: 10.1091/mbc.e15-04-0218] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 02/13/2017] [Accepted: 02/17/2017] [Indexed: 11/18/2022] Open
Abstract
LIMCH1 specifically associates with contractile stress fibers. The N-terminus of LIMCH1 directly interacts with the head of NM-IIA. LIMCH1 depletion modulates the myosin regulatory light chain and FAK phosphorylation. Actin stress fiber and focal adhesion are decreased in LIMCH1-depleted cells, leading to increased cell migration. Nonmuscle myosin II (NM-II) is an important motor protein involved in cell migration. Incorporation of NM-II into actin stress fiber provides a traction force to promote actin retrograde flow and focal adhesion assembly. However, the components involved in regulation of NM-II activity are not well understood. Here we identified a novel actin stress fiber–associated protein, LIM and calponin-homology domains 1 (LIMCH1), which regulates NM-II activity. The recruitment of LIMCH1 into contractile stress fibers revealed its localization complementary to actinin-1. LIMCH1 interacted with NM-IIA, but not NM-IIB, independent of the inhibition of myosin ATPase activity with blebbistatin. Moreover, the N-terminus of LIMCH1 binds to the head region of NM-IIA. Depletion of LIMCH1 attenuated myosin regulatory light chain (MRLC) diphosphorylation in HeLa cells, which was restored by reexpression of small interfering RNA–resistant LIMCH1. In addition, LIMCH1-depleted HeLa cells exhibited a decrease in the number of actin stress fibers and focal adhesions, leading to enhanced cell migration. Collectively, our data suggest that LIMCH1 plays a positive role in regulation of NM-II activity through effects on MRLC during cell migration.
Collapse
Affiliation(s)
- Yu-Hung Lin
- Graduate Institute of Biomedical Sciences, Department of Biochemistry, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Yen-Yi Zhen
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Kun-Yi Chien
- Graduate Institute of Biomedical Sciences, Department of Biochemistry, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Tao-Yuan 333, Taiwan
| | - I-Ching Lee
- Graduate Institute of Biomedical Sciences, Department of Biochemistry, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Wei-Chi Lin
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 11221, Taiwan
| | - Mei-Yu Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming University, Taipei 11221, Taiwan.,Genome Research Center, National Yang Ming University, Taipei 11221, Taiwan
| | - Li-Mei Pai
- Graduate Institute of Biomedical Sciences, Department of Biochemistry, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan .,Molecular Medicine Research Center, Chang Gung University, Tao-Yuan 333, Taiwan.,Liver Research Center, Chang Gung Memorial Hospital, Tao-Yuan 333, Taiwan
| |
Collapse
|
39
|
Hatakeyama H, Nakahata Y, Yarimizu H, Kanzaki M. Live-cell single-molecule labeling and analysis of myosin motors with quantum dots. Mol Biol Cell 2016; 28:173-181. [PMID: 28035048 PMCID: PMC5221621 DOI: 10.1091/mbc.e16-06-0413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 01/07/2023] Open
Abstract
Quantum dots (QDs) are a powerful tool for quantitative biology, but two challenges are associated with using them to track intracellular molecules in live cells. A simple and convenient method is presented for labeling intracellular molecules by using HaloTag technology and electroporation and is used to successfully track myosins within live cells. Quantum dots (QDs) are a powerful tool for quantitatively analyzing dynamic cellular processes by single-particle tracking. However, tracking of intracellular molecules with QDs is limited by their inability to penetrate the plasma membrane and bind to specific molecules of interest. Although several techniques for overcoming these problems have been proposed, they are either complicated or inconvenient. To address this issue, in this study, we developed a simple, convenient, and nontoxic method for labeling intracellular molecules in cells using HaloTag technology and electroporation. We labeled intracellular myosin motors with this approach and tracked their movement within cells. By simultaneously imaging myosin movement and F-actin architecture, we observed that F-actin serves not only as a rail but also as a barrier for myosin movement. We analyzed the effect of insulin on the movement of several myosin motors, which have been suggested to regulate intracellular trafficking of the insulin-responsive glucose transporter GLUT4, but found no significant enhancement in myosin motor motility as a result of insulin treatment. Our approach expands the repertoire of proteins for which intracellular dynamics can be analyzed at the single-molecule level.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8579, Japan .,Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan
| | - Yoshihito Nakahata
- Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan
| | - Hirokazu Yarimizu
- Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan
| | - Makoto Kanzaki
- Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8579, Japan.,Department of Information and Intelligent Systems, Tohoku University, Sendai 980-8579, Japan
| |
Collapse
|
40
|
Activity of nonmuscle myosin II isoforms determines localization at the cleavage furrow of megakaryocytes. Blood 2016; 128:3137-3145. [PMID: 27737892 DOI: 10.1182/blood-2016-04-711630] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 10/07/2016] [Indexed: 01/03/2023] Open
Abstract
Megakaryocyte polyploidy is characterized by cytokinesis failure resulting from defects in contractile forces at the cleavage furrow. Although immature megakaryocytes express 2 nonmuscle myosin II isoforms (MYH9 [NMIIA] and MYH10 [NMIIB]), only NMIIB localizes at the cleavage furrow, and its subsequent absence contributes to polyploidy. In this study, we tried to understand why the abundant NMIIA does not localize at the furrow by focusing on the RhoA/ROCK pathway that has a low activity in polyploid megakaryocytes. We observed that under low RhoA activity, NMII isoforms presented different activity that determined their localization. Inhibition of RhoA/ROCK signaling abolished the localization of NMIIB, whereas constitutively active RhoA induced NMIIA at the cleavage furrow. Thus, although high RhoA activity favored the localization of both the isoforms, only NMIIB could localize at the furrow at low RhoA activity. This was further confirmed in erythroblasts that have a higher basal RhoA activity than megakaryocytes and express both NMIIA and NMIIB at the cleavage furrow. Decreased RhoA activity in erythroblasts abolished localization of NMIIA but not of NMIIB from the furrow. This differential localization was related to differences in actin turnover. Megakaryocytes had a higher actin turnover compared with erythroblasts. Strikingly, inhibition of actin polymerization was found to be sufficient to recapitulate the effects of inhibition of RhoA/ROCK pathway on NMII isoform localization; thus, cytokinesis failure in megakaryocytes is the consequence of both the absence of NMIIB and a low RhoA activity that impairs NMIIA localization at the cleavage furrow through increased actin turnover.
Collapse
|
41
|
Abstract
Mechanotransduction is the process through which cells survey the mechanical properties of their environment, convert these mechanical inputs into biochemical signals, and modulate their phenotype in response. These mechanical inputs, which may be encoded in the form of extracellular matrix stiffness, dimensionality, and adhesion, all strongly influence cell morphology, migration, and fate decisions. One mechanism through which cells on planar or pseudo-planar matrices exert tensile forces and interrogate microenvironmental mechanics is through stress fibers, which are bundles composed of actin filaments and, in most cases, non-muscle myosin II filaments. Stress fibers form a continuous structural network that is mechanically coupled to the extracellular matrix through focal adhesions. Furthermore, myosin-driven contractility plays a central role in the ability of stress fibers to sense matrix mechanics and generate tension. Here, we review the distinct roles that non-muscle myosin II plays in driving mechanosensing and focus specifically on motility. In a closely related discussion, we also describe stress fiber classification schemes and the differing roles of various myosin isoforms in each category. Finally, we briefly highlight recent studies exploring mechanosensing in three-dimensional environments, in which matrix content, structure, and mechanics are often tightly interrelated. Stress fibers and the myosin motors therein represent an intriguing and functionally important biological system in which mechanics, biochemistry, and architecture all converge.
Collapse
Affiliation(s)
- Stacey Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
42
|
A highly soluble, non-phototoxic, non-fluorescent blebbistatin derivative. Sci Rep 2016; 6:26141. [PMID: 27241904 PMCID: PMC4886532 DOI: 10.1038/srep26141] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/20/2016] [Indexed: 12/15/2022] Open
Abstract
Blebbistatin is a commonly used molecular tool for the specific inhibition of various myosin II isoforms both in vitro and in vivo. Despite its popularity, the use of blebbistatin is hindered by its poor water-solubility (below 10 micromolar in aqueous buffer) and blue-light sensitivity, resulting in the photoconversion of the molecule, causing severe cellular phototoxicity in addition to its cytotoxicity. Furthermore, blebbistatin forms insoluble aggregates in water-based media above 10 micromolar with extremely high fluorescence and also high adherence to different types of surfaces, which biases its experimental usage. Here, we report a highly soluble (440 micromolar in aqueous buffer), non-fluorescent and photostable C15 amino-substituted derivative of blebbistatin, called para-aminoblebbistatin. Importantly, it is neither photo- nor cytotoxic, as demonstrated on HeLa cells and zebrafish embryos. Additionally, para-aminoblebbistatin bears similar myosin II inhibitory properties to blebbistatin or para-nitroblebbistatin (not to be confused with the C7 substituted nitroblebbistatin), tested on rabbit skeletal muscle myosin S1 and on M2 and HeLa cells. Due to its drastically improved solubility and photochemical feature, as well as lack of photo- or cytotoxicity, para-aminoblebbistatin may become a feasible replacement for blebbistatin, especially at applications when high concentrations of the inhibitor or blue light irradiation is required.
Collapse
|
43
|
Kiss B, Kalmár L, Nyitray L, Pál G. Structural determinants governing S100A4-induced isoform-selective disassembly of nonmuscle myosin II filaments. FEBS J 2016; 283:2164-80. [PMID: 27029887 DOI: 10.1111/febs.13728] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/21/2016] [Accepted: 03/30/2016] [Indexed: 12/30/2022]
Abstract
The Ca(2+) -binding protein S100A4 interacts with the C terminus of nonmuscle myosin IIA (NMIIA) causing filament disassembly, which is correlated with an increased metastatic potential of tumor cells. Despite high sequence similarity of the three NMII isoforms, S100A4 discriminates against binding to NMIIB. We searched for structural determinants of this selectivity. Based on paralog scanning using phage display, we identified a single position as major determinant of isoform selectivity. Reciprocal single amino acid replacements showed that at position 1907 (NMIIA numbering), the NMIIA/NMIIC-specific alanine provides about 60-fold higher affinity than the NMIIB-specific asparagine. The structural background of this can be explained in part by a communication between the two consecutive α-helical binding segments. This communication is completely abolished by the Ala-to-Asn substitution. Mutual swapping of the disordered tailpieces only slightly affects the affinity of the NMII chimeras. Interestingly, we found that the tailpiece and position 1907 act in a nonadditive fashion. Finally, we also found that the higher stability of the C-terminal coiled-coil region of NMIIB also discriminates against interaction with S100A4. Our results clearly show that the isoform-selective binding of S100A4 is determined at multiple levels in the structure of the three NMII isoforms and the corresponding functional elements of NMII act synergistically with one another resulting in a complex interaction network. The experimental and in silico results suggest two divergent evolutionary pathways: NMIIA and NMIIB evolved to possess S100A4-dependent and -independent regulations, respectively.
Collapse
Affiliation(s)
- Bence Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Lajos Kalmár
- Institute of Enzymology, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Budapest, Hungary.,Department of Veterinary Medicine, University of Cambridge, Cambridgeshire, UK
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
44
|
Lehtimäki J, Hakala M, Lappalainen P. Actin Filament Structures in Migrating Cells. Handb Exp Pharmacol 2016; 235:123-152. [PMID: 27469496 DOI: 10.1007/164_2016_28] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell migration is necessary for several developmental processes in multicellular organisms. Furthermore, many physiological processes such as wound healing and immunological events in adult animals are dependent on cell migration. Consequently, defects in cell migration are linked to various diseases including immunological disorders as well as cancer progression and metastasis formation. Cell migration is driven by specific protrusive and contractile actin filament structures, but the types and relative contributions of these actin filament arrays vary depending on the cell type and the environment of the cell. In this chapter, we introduce the most important actin filament structures that contribute to mesenchymal and amoeboid cell migration modes and discuss the mechanisms by which the assembly and turnover of these structures are controlled by various actin-binding proteins.
Collapse
Affiliation(s)
- Jaakko Lehtimäki
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Markku Hakala
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland.
| |
Collapse
|
45
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
46
|
Differential Contributions of Nonmuscle Myosin II Isoforms and Functional Domains to Stress Fiber Mechanics. Sci Rep 2015; 5:13736. [PMID: 26336830 PMCID: PMC4559901 DOI: 10.1038/srep13736] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/04/2015] [Indexed: 01/14/2023] Open
Abstract
While is widely acknowledged that nonmuscle myosin II (NMMII) enables stress fibers (SFs) to generate traction forces against the extracellular matrix, little is known about how specific NMMII isoforms and functional domains contribute to SF mechanics. Here we combine biophotonic and genetic approaches to address these open questions. First, we suppress the NMMII isoforms MIIA and MIIB and apply femtosecond laser nanosurgery to ablate and investigate the viscoelastic retraction of individual SFs. SF retraction dynamics associated with MIIA and MIIB suppression qualitatively phenocopy our earlier measurements in the setting of Rho kinase (ROCK) and myosin light chain kinase (MLCK) inhibition, respectively. Furthermore, fluorescence imaging and photobleaching recovery reveal that MIIA and MIIB are enriched in and more stably localize to ROCK- and MLCK-controlled central and peripheral SFs, respectively. Additional domain-mapping studies surprisingly reveal that deletion of the head domain speeds SF retraction, which we ascribe to reduced drag from actomyosin crosslinking and frictional losses. We propose a model in which ROCK/MIIA and MLCK/MIIB functionally regulate common pools of SFs, with MIIA crosslinking and motor functions jointly contributing to SF retraction dynamics and cellular traction forces.
Collapse
|
47
|
Juanes-Garcia A, Chapman JR, Aguilar-Cuenca R, Delgado-Arevalo C, Hodges J, Whitmore LA, Shabanowitz J, Hunt DF, Horwitz AR, Vicente-Manzanares M. A regulatory motif in nonmuscle myosin II-B regulates its role in migratory front-back polarity. ACTA ACUST UNITED AC 2015; 209:23-32. [PMID: 25869664 PMCID: PMC4395487 DOI: 10.1083/jcb.201407059] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In this study, we show that the role of nonmuscle myosin II (NMII)-B in front-back migratory cell polarity is controlled by a short stretch of amino acids containing five serines (1935-1941). This motif resides near the junction between the C terminus helical and nonhelical tail domains. Removal of this motif inhibited NMII-B assembly, whereas its insertion into NMII-A endowed an NMII-B-like ability to generate large actomyosin bundles that determine the rear of the cell. Phosphomimetic mutation of the five serines also inhibited NMII-B assembly, rendering it unable to support front-back polarization. Mass spectrometric analysis showed that several of these serines are phosphorylated in live cells. Single-site mutagenesis showed that serine 1935 is a major regulatory site of NMII-B function. These data reveal a novel regulatory mechanism of NMII in polarized migrating cells by identifying a key molecular determinant that confers NMII isoform functional specificity.
Collapse
Affiliation(s)
- Alba Juanes-Garcia
- Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain
| | - Jessica R Chapman
- Department of Chemistry and Department of Pathology, University of Virginia, Charlottesville, VA 22901
| | - Rocio Aguilar-Cuenca
- Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain
| | - Cristina Delgado-Arevalo
- Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain
| | - Jennifer Hodges
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Leanna A Whitmore
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Jeffrey Shabanowitz
- Department of Chemistry and Department of Pathology, University of Virginia, Charlottesville, VA 22901
| | - Donald F Hunt
- Department of Chemistry and Department of Pathology, University of Virginia, Charlottesville, VA 22901 Department of Chemistry and Department of Pathology, University of Virginia, Charlottesville, VA 22901
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Miguel Vicente-Manzanares
- Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain Instituto de Investigacion Sanitaria-Hospital Universitario de la Princesa and Universidad Autonoma de Madrid School of Medicine, 28006 Madrid, Spain
| |
Collapse
|
48
|
Badirou I, Pan J, Souquere S, Legrand C, Pierron G, Wang A, Eckly A, Roy A, Gachet C, Vainchenker W, Chang Y, Léon C. Distinct localizations and roles of non-muscle myosin II during proplatelet formation and platelet release. J Thromb Haemost 2015; 13:851-9. [PMID: 25736522 DOI: 10.1111/jth.12887] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/24/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND At the end of maturation, megakaryocytes (MKs) form long cytoplasmic extensions called proplatelets (PPT). Enormous changes in cytoskeletal structures cause PPT to extend further, to re-localize organelles such as mitochondria and to fragment, leading to platelet release. Two non-muscle myosin IIs (NMIIs) are expressed in MKs; however, only NMII-A (MYH9), but not NMII-B (MYH10), is expressed in mature MKs and is implicated in PPT formation. OBJECTIVES To provide in vivo evidence on the specific role of NMII-A and IIB in MK PPT formation. METHODS We studied two transgenic mouse models in which non-muscle myosin heavy chain (NMHC) II-A was genetically replaced either by II-B or by a chimeric NMHCII that combined the head domain of II-A with the rod and tail domains of II-B. RESULTS AND CONCLUSIONS This work demonstrates that the kinetic properties of NM-IIA, depending on the N-terminal domain, render NMII-A the better NMII candidate to control PPT formation. Furthermore, the carboxyl-terminal domain determines myosin II localization in the constriction region of PPT and is responsible for the specific role of NMII in platelet release.
Collapse
Affiliation(s)
- I Badirou
- Institut National de la Santé et de la Recherche Médicale, Villejuif, France; Université Paris-Sud, Le Kremlin-Bicêtre, France; Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Moehle MS, Daher JPL, Hull TD, Boddu R, Abdelmotilib HA, Mobley J, Kannarkat GT, Tansey MG, West AB. The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins. Hum Mol Genet 2015; 24:4250-67. [PMID: 25926623 DOI: 10.1093/hmg/ddv157] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022] Open
Abstract
The Leucine rich repeat kinase 2 (LRRK2) gene is genetically and biochemically linked to several diseases that involve innate immunity. LRRK2 protein is highly expressed in phagocytic cells of the innate immune system, most notably in myeloid cells capable of mounting potent pro-inflammatory responses. Knockdown of LRRK2 protein in these cells reduces pro-inflammatory responses. However, the effect of LRRK2 pathogenic mutations that cause Parkinson's disease on myeloid cell function is not clear but could provide insight into LRRK2-linked disease. Here, we find that rats expressing G2019S LRRK2 have exaggerated pro-inflammatory responses and subsequent neurodegeneration after lipopolysaccharide injections in the substantia nigra, with a marked increase in the recruitment of CD68 myeloid cells to the site of injection. While G2019S LRRK2 expression did not affect immunological homeostasis, myeloid cells expressing G2019S LRRK2 show enhanced chemotaxis both in vitro in two-chamber assays and in vivo in response to thioglycollate injections in the peritoneum. The G2019S mutation enhanced the association between LRRK2 and actin-regulatory proteins that control chemotaxis. The interaction between G2019S LRRK2 and actin-regulatory proteins can be blocked by LRRK2 kinase inhibitors, although we did not find evidence that LRRK2 phosphorylated these interacting proteins. These results suggest that the primary mechanism of G2019S LRRK2 with respect to myeloid cell function in disease may be related to exaggerated chemotactic responses.
Collapse
Affiliation(s)
- Mark S Moehle
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics
| | | | | | - Ravindra Boddu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA and
| | | | | | - George T Kannarkat
- Department of Physiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Malú G Tansey
- Department of Physiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Andrew B West
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics,
| |
Collapse
|
50
|
Beach JR, Hammer JA. Myosin II isoform co-assembly and differential regulation in mammalian systems. Exp Cell Res 2015; 334:2-9. [PMID: 25655283 DOI: 10.1016/j.yexcr.2015.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 10/24/2022]
Abstract
Non-muscle myosin 2 (NM2) is a major force-producing, actin-based motor in mammalian non-muscle cells, where it plays important roles in a broad range of fundamental biological processes, including cytokinesis, cell migration, and epithelial barrier function. This breadth of function at the tissue and cellular levels suggests extensive diversity and differential regulation of NM2 bipolar filaments, the major, if not sole, functional form of NM2s in vivo. Previous in vitro, cellular and animal studies indicate that some of this diversity is supported by the existence of multiple NM2 isoforms. Moreover, two recent studies have shown that these isoforms can co-assemble to form heterotypic filaments, further expanding functional diversity. In addition to isoform co-assembly, cells may differentially regulate NM2 function via isoform-specific expression, RLC phosphorylation, MHC phosphorylation or regulation via binding partners. Here, we provide a brief summary of NM2 filament assembly, summarize the recent findings regarding NM2 isoform co-assembly, consider the mechanisms cells might utilize to differentially regulate NM2 isoforms, and review the data available to support these mechanisms.
Collapse
Affiliation(s)
- Jordan R Beach
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|