1
|
Endo S, Yamamoto S, Miyoshi H. Development of label-free cell tracking for discrimination of the heterogeneous mesenchymal migration. PLoS One 2025; 20:e0320287. [PMID: 40163519 PMCID: PMC11957292 DOI: 10.1371/journal.pone.0320287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
Image-based cell phenotyping is fundamental in both cell biology and medicine. As cells are dynamic systems, phenotyping based on static data is complemented by dynamic data extracted from time-dependent cell characteristics. We developed a label-free automatic tracking method for phase contrast images. We examined the possibility of using cell motility-based discrimination to identify different types of mesenchymal migration in invasive malignant cancer and non-cancer cells. These cells were cultured in plastic tissue culture vessels, using motility parameters from cell trajectories extracted with label-free tracking. Correlation analysis with these motility parameters identified characteristic parameters for cancer HT1080 fibrosarcoma and non-cancer 3T3-Swiss fibroblast cell lines. The parameter "sum of turn angles," combined with the "frequency of turns" at shallow angles and "migration speed," proved effective in highlighting the migration characteristics of these cells. It revealed differences in their mechanisms for generating effective propulsive forces. The requirements to characterize these differences included the spatiotemporal resolution of segmentation and tracking, capable of detecting polarity changes associated with cell morphological alterations and cell body displacement. With the segmentation and tracking method proposed here, a discrimination curve computed using quadratic discrimination analysis from the "sum of turn angles" and "frequency of turns below 30°" gave the best performance with a 94% sensitivity. Cell migration is a process related not only to cancer but also to tissue healing and growth. The proposed methodology is easy to use, enabling anyone without professional skills in image analysis, large training datasets, or special devices. It has the potential for application not only in cancer cell discrimination but also in a broad range of applications and basic research. Validating the expandability of this method to characterize cell migration, including the scheme of propulsive force generation, is an important consideration for future study.
Collapse
Affiliation(s)
- Sota Endo
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Shotaro Yamamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Hiromi Miyoshi
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| |
Collapse
|
2
|
Weißenbruch K, Mayor R. Actomyosin forces in cell migration: Moving beyond cell body retraction. Bioessays 2024; 46:e2400055. [PMID: 39093597 DOI: 10.1002/bies.202400055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
In textbook illustrations of migrating cells, actomyosin contractility is typically depicted as the contraction force necessary for cell body retraction. This dogma has been transformed by the molecular clutch model, which acknowledges that actomyosin traction forces also generate and transmit biomechanical signals at the leading edge, enabling cells to sense and shape their migratory path in mechanically complex environments. To fulfill these complementary functions, the actomyosin system assembles a gradient of contractile energy along the front-rear axis of migratory cells. Here, we highlight the hierarchic assembly and self-regulatory network structure of the actomyosin system and explain how the kinetics of different nonmuscle myosin II (NM II) paralogs synergize during contractile force generation. Our aim is to emphasize how protrusion formation, cell adhesion, contraction, and retraction are spatiotemporally integrated during different modes of migration, including chemotaxis and durotaxis. Finally, we hypothesize how different NM II paralogs might tune aspects of migration in vivo, highlighting future research directions.
Collapse
Affiliation(s)
- Kai Weißenbruch
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
3
|
Feroz W, Park BS, Siripurapu M, Ntim N, Kilroy MK, Sheikh AMA, Mishra R, Garrett JT. Non-Muscle Myosin II A: Friend or Foe in Cancer? Int J Mol Sci 2024; 25:9435. [PMID: 39273383 PMCID: PMC11395477 DOI: 10.3390/ijms25179435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.
Collapse
Affiliation(s)
- Wasim Feroz
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Briley SoYoung Park
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
- Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meghna Siripurapu
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Nicole Ntim
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Mary Kate Kilroy
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | | | - Rosalin Mishra
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Joan T. Garrett
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| |
Collapse
|
4
|
Chinthalapudi K, Heissler SM. Structure, regulation, and mechanisms of nonmuscle myosin-2. Cell Mol Life Sci 2024; 81:263. [PMID: 38878079 PMCID: PMC11335295 DOI: 10.1007/s00018-024-05264-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 06/23/2024]
Abstract
Members of the myosin superfamily of molecular motors are large mechanochemical ATPases that are implicated in an ever-expanding array of cellular functions. This review focuses on mammalian nonmuscle myosin-2 (NM2) paralogs, ubiquitous members of the myosin-2 family of filament-forming motors. Through the conversion of chemical energy into mechanical work, NM2 paralogs remodel and shape cells and tissues. This process is tightly controlled in time and space by numerous synergetic regulation mechanisms to meet cellular demands. We review how recent advances in structural biology together with elegant biophysical and cell biological approaches have contributed to our understanding of the shared and unique mechanisms of NM2 paralogs as they relate to their kinetics, regulation, assembly, and cellular function.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
5
|
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
|
6
|
Garrido-Casado M, Asensio-Juárez G, Talayero VC, Vicente-Manzanares M. Engines of change: Nonmuscle myosin II in mechanobiology. Curr Opin Cell Biol 2024; 87:102344. [PMID: 38442667 DOI: 10.1016/j.ceb.2024.102344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024]
Abstract
The emergence of mechanobiology has unveiled complex mechanisms by which cells adjust intracellular force production to their needs. Most communicable intracellular forces are generated by myosin II, an actin-associated molecular motor that transforms adenosine triphosphate (ATP) hydrolysis into contraction in nonmuscle and muscle cells. Myosin II-dependent force generation is tightly regulated, and deregulation is associated with specific pathologies. Here, we focus on the role of myosin II (nonmuscle myosin II, NMII) in force generation and mechanobiology. We outline the regulation and molecular mechanism of force generation by NMII, focusing on the actual outcome of contraction, that is, force application to trigger mechanosensitive events or the building of dissipative structures. We describe how myosin II-generated forces drive two major types of events: modification of the cellular morphology and/or triggering of genetic programs, which enhance the ability of cells to adapt to, or modify, their microenvironment. Finally, we address whether targeting myosin II to impair or potentiate its activity at the motor level is a viable therapeutic strategy, as illustrated by recent examples aimed at modulating cardiac myosin II function in heart disease.
Collapse
Affiliation(s)
- Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Gloria Asensio-Juárez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Vanessa C Talayero
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
7
|
Quintanilla MA, Patel H, Wu H, Sochacki KA, Chandrasekar S, Akamatsu M, Rotty JD, Korobova F, Bear JE, Taraska JW, Oakes PW, Beach JR. Local monomer levels and established filaments potentiate non-muscle myosin 2 assembly. J Cell Biol 2024; 223:e202305023. [PMID: 38353656 PMCID: PMC10866686 DOI: 10.1083/jcb.202305023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/02/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
The ability to dynamically assemble contractile networks is required throughout cell physiology, yet direct biophysical mechanisms regulating non-muscle myosin 2 filament assembly in living cells are lacking. Here, we use a suite of dynamic, quantitative imaging approaches to identify deterministic factors that drive myosin filament appearance and amplification. We find that actin dynamics regulate myosin assembly, but that the static actin architecture plays a less clear role. Instead, remodeling of actin networks modulates the local myosin monomer levels and facilitates assembly through myosin:myosin-driven interactions. Using optogenetically controlled myosin, we demonstrate that locally concentrating myosin is sufficient to both form filaments and jump-start filament amplification and partitioning. By counting myosin monomers within filaments, we demonstrate a myosin-facilitated assembly process that establishes filament stacks prior to partitioning into clusters that feed higher-order networks. Together, these findings establish the biophysical mechanisms regulating the assembly of non-muscle contractile structures that are ubiquitous throughout cell biology.
Collapse
Affiliation(s)
- Melissa A. Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Hiral Patel
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Huini Wu
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Kem A. Sochacki
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shreya Chandrasekar
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Matthew Akamatsu
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Jeremy D. Rotty
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Farida Korobova
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - James E. Bear
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Justin W. Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patrick W. Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
8
|
Miller SG, Hoh M, Ebmeier CC, Tay JW, Ahn NG. Cooperative polarization of MCAM/CD146 and ERM family proteins in melanoma. Mol Biol Cell 2024; 35:ar31. [PMID: 38117590 PMCID: PMC10916866 DOI: 10.1091/mbc.e23-06-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/15/2023] [Indexed: 12/22/2023] Open
Abstract
The WRAMP structure is a protein network associated with tail-end actomyosin contractility, membrane retraction, and directional persistence during cell migration. A marker of WRAMP structures is melanoma cell adhesion molecule (MCAM) which dynamically polarizes to the cell rear. However, factors that mediate MCAM polarization are still unknown. In this study, BioID using MCAM as bait identifies the ERM family proteins, moesin, ezrin, and radixin, as WRAMP structure components. We also present a novel image analysis pipeline, Protein Polarity by Percentile ("3P"), which classifies protein polarization using machine learning and facilitates quantitative analysis. Using 3P, we find that depletion of moesin, and to a lesser extent ezrin, decreases the proportion of cells with polarized MCAM. Furthermore, although copolarized MCAM and ERM proteins show high spatial overlap, 3P identifies subpopulations with ERM proteins closer to the cell periphery. Live-cell imaging confirms that MCAM and ERM protein polarization is tightly coordinated, but ERM proteins enrich at the cell edge first. Finally, deletion of a juxtamembrane segment in MCAM previously shown to promote ERM protein interactions impedes MCAM polarization. Our findings highlight the requirement for ERM proteins in recruitment of MCAM to WRAMP structures and an advanced computational tool to characterize protein polarization.
Collapse
Affiliation(s)
- Suzannah G. Miller
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
| | - Maria Hoh
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
| | | | - Jian Wei Tay
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO 80303
| | - Natalie G. Ahn
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303
- BioFrontiers Institute, University of Colorado Boulder, Boulder CO 80303
| |
Collapse
|
9
|
Kim OV, Litvinov RI, Gagne AL, French DL, Brass LF, Weisel JW. Megakaryocyte-induced contraction of plasma clots: cellular mechanisms and structural mechanobiology. Blood 2024; 143:548-560. [PMID: 37944157 PMCID: PMC11033616 DOI: 10.1182/blood.2023021545] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbβ3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alyssa L. Gagne
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence F. Brass
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
10
|
Southern BD, Li H, Mao H, Crish JF, Grove LM, Scheraga RG, Mansoor S, Reinhardt A, Abraham S, Deshpande G, Loui A, Ivanov AI, Rosenfeld SS, Bresnick AR, Olman MA. A novel mechanoeffector role of fibroblast S100A4 in myofibroblast transdifferentiation and fibrosis. J Biol Chem 2024; 300:105530. [PMID: 38072048 PMCID: PMC10789633 DOI: 10.1016/j.jbc.2023.105530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/23/2023] Open
Abstract
Fibroblast to myofibroblast transdifferentiation mediates numerous fibrotic disorders, such as idiopathic pulmonary fibrosis (IPF). We have previously demonstrated that non-muscle myosin II (NMII) is activated in response to fibrotic lung extracellular matrix, thereby mediating myofibroblast transdifferentiation. NMII-A is known to interact with the calcium-binding protein S100A4, but the mechanism by which S100A4 regulates fibrotic disorders is unclear. In this study, we show that fibroblast S100A4 is a calcium-dependent, mechanoeffector protein that is uniquely sensitive to pathophysiologic-range lung stiffness (8-25 kPa) and thereby mediates myofibroblast transdifferentiation. Re-expression of endogenous fibroblast S100A4 rescues the myofibroblastic phenotype in S100A4 KO fibroblasts. Analysis of NMII-A/actin dynamics reveals that S100A4 mediates the unraveling and redistribution of peripheral actomyosin to a central location, resulting in a contractile myofibroblast. Furthermore, S100A4 loss protects against murine in vivo pulmonary fibrosis, and S100A4 expression is dysregulated in IPF. Our data reveal a novel mechanosensor/effector role for endogenous fibroblast S100A4 in inducing cytoskeletal redistribution in fibrotic disorders such as IPF.
Collapse
Affiliation(s)
- Brian D Southern
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Haiyan Li
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hongxia Mao
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - James F Crish
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lisa M Grove
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rachel G Scheraga
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sanaa Mansoor
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Amanda Reinhardt
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Susamma Abraham
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gauravi Deshpande
- Lerner Research Institute Imaging Core, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alicia Loui
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrei I Ivanov
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Steven S Rosenfeld
- Division of Hematology/Oncology, Mayo Clinic Jacksonville, Jacksonville, Florida, USA
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mitchell A Olman
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
11
|
Newman D, Young LE, Waring T, Brown L, Wolanska KI, MacDonald E, Charles-Orszag A, Goult BT, Caswell PT, Sakuma T, Yamamoto T, Machesky LM, Morgan MR, Zech T. 3D matrix adhesion feedback controls nuclear force coupling to drive invasive cell migration. Cell Rep 2023; 42:113554. [PMID: 38100355 DOI: 10.1016/j.celrep.2023.113554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 06/23/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Cell invasion is a multi-step process, initiated by the acquisition of a migratory phenotype and the ability to move through complex 3D extracellular environments. We determine the composition of cell-matrix adhesion complexes of invasive breast cancer cells in 3D matrices and identify an interaction complex required for invasive migration. βPix and myosin18A (Myo18A) drive polarized recruitment of non-muscle myosin 2A (NM2A) to adhesion complexes at the tips of protrusions. Actomyosin force engagement then displaces the Git1-βPix complex from paxillin, establishing a feedback loop for adhesion maturation. We observe active force transmission to the nucleus during invasive migration that is needed to pull the nucleus forward. The recruitment of NM2A to adhesions creates a non-muscle myosin isoform gradient, which extends from the protrusion to the nucleus. We postulate that this gradient facilitates coupling of cell-matrix interactions at the protrusive cell front with nuclear movement, enabling effective invasive migration and front-rear cell polarity.
Collapse
Affiliation(s)
- Daniel Newman
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lorna E Young
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Thomas Waring
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Louise Brown
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katarzyna I Wolanska
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Ewan MacDonald
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | | | | | - Patrick T Caswell
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Tetsushi Sakuma
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8526, Japan
| | - Takashi Yamamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8526, Japan
| | - Laura M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, UK; Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, UK
| | - Mark R Morgan
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Tobias Zech
- Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
12
|
Dang I, Brazzo JA, Bae Y, Assoian RK. Key role for Rac in the early transcriptional response to extracellular matrix stiffness and stiffness-dependent repression of ATF3. J Cell Sci 2023; 136:jcs260636. [PMID: 37737020 PMCID: PMC10617619 DOI: 10.1242/jcs.260636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
The Rho family GTPases Rac and Rho play critical roles in transmitting mechanical information contained within the extracellular matrix (ECM) to the cell. Rac and Rho have well-described roles in regulating stiffness-dependent actin remodeling, proliferation and motility. However, much less is known about the relative roles of these GTPases in stiffness-dependent transcription, particularly at the genome-wide level. Here, we selectively inhibited Rac and Rho in mouse embryonic fibroblasts cultured on deformable substrata and used RNA sequencing to elucidate and compare the contribution of these GTPases to the early transcriptional response to ECM stiffness. Surprisingly, we found that the stiffness-dependent activation of Rac was dominant over Rho in the initial transcriptional response to ECM stiffness. We also identified activating transcription factor 3 (ATF3) as a major target of stiffness- and Rac-mediated signaling and show that ATF3 repression by ECM stiffness helps to explain how the stiffness-dependent activation of Rac results in the induction of cyclin D1.
Collapse
Affiliation(s)
- Irène Dang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Richard K. Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Brito C, Pereira JM, Mesquita FS, Cabanes D, Sousa S. Src-Dependent NM2A Tyrosine Phosphorylation Regulates Actomyosin Remodeling. Cells 2023; 12:1871. [PMID: 37508535 PMCID: PMC10377941 DOI: 10.3390/cells12141871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Non-muscle myosin 2A (NM2A) is a key cytoskeletal enzyme that, along with actin, assembles into actomyosin filaments inside cells. NM2A is fundamental for cell adhesion and motility, playing important functions in different stages of development and during the progression of viral and bacterial infections. Phosphorylation events regulate the activity and the cellular localization of NM2A. We previously identified the tyrosine phosphorylation of residue 158 (pTyr158) in the motor domain of the NM2A heavy chain. This phosphorylation can be promoted by Listeria monocytogenes infection of epithelial cells and is dependent on Src kinase; however, its molecular role is unknown. Here, we show that the status of pTyr158 defines cytoskeletal organization, affects the assembly/disassembly of focal adhesions, and interferes with cell migration. Cells overexpressing a non-phosphorylatable NM2A variant or expressing reduced levels of Src kinase display increased stress fibers and larger focal adhesions, suggesting an altered contraction status consistent with the increased NM2A activity that we also observed. We propose NM2A pTyr158 as a novel layer of regulation of actomyosin cytoskeleton organization.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Joana M Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| |
Collapse
|
14
|
Quintanilla MA, Patel H, Wu H, Sochacki KA, Akamatsu M, Rotty JD, Korobova F, Bear JE, Taraska JW, Oakes PW, Beach JR. Local Monomer Levels and Established Filaments Potentiate Non-Muscle Myosin 2 Assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538303. [PMID: 37162845 PMCID: PMC10168331 DOI: 10.1101/2023.04.26.538303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The ability to dynamically assemble contractile networks is required throughout cell physiology, yet the biophysical mechanisms regulating non-muscle myosin 2 filament assembly in living cells are lacking. Here we use a suite of dynamic, quantitative imaging approaches to identify deterministic factors that drive myosin filament appearance and amplification. We find that actin dynamics regulate myosin assembly, but that the actin architecture plays a minimal direct role. Instead, remodeling of actin networks modulates the local myosin monomer levels and facilitates assembly through myosin:myosin driven interactions. Using optogenetically controlled myosin, we demonstrate that locally concentrating myosin is sufficient to both form filaments and jump-start filament amplification and partitioning. By counting myosin monomers within filaments, we demonstrate a myosin-facilitated assembly process that establishes sub-resolution filament stacks prior to partitioning into clusters that feed higher-order networks. Together these findings establish the biophysical mechanisms regulating the assembly of non-muscle contractile structures that are ubiquitous throughout cell biology.
Collapse
Affiliation(s)
- Melissa A Quintanilla
- 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
| | - Kem A Sochacki
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | | | - Jeremy D Rotty
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Farida Korobova
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Patrick W Oakes
- 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
|
15
|
Cowan JM, Duggan JJ, Hewitt BR, Petrie RJ. Non-muscle myosin II and the plasticity of 3D cell migration. Front Cell Dev Biol 2022; 10:1047256. [PMID: 36438570 PMCID: PMC9691290 DOI: 10.3389/fcell.2022.1047256] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/31/2022] [Indexed: 09/08/2024] Open
Abstract
Confined cells migrating through 3D environments are also constrained by the laws of physics, meaning for every action there must be an equal and opposite reaction for cells to achieve motion. Fascinatingly, there are several distinct molecular mechanisms that cells can use to move, and this is reflected in the diverse ways non-muscle myosin II (NMII) can generate the mechanical forces necessary to sustain 3D cell migration. This review summarizes the unique modes of 3D migration, as well as how NMII activity is regulated and localized within each of these different modes. In addition, we highlight tropomyosins and septins as two protein families that likely have more secrets to reveal about how NMII activity is governed during 3D cell migration. Together, this information suggests that investigating the mechanisms controlling NMII activity will be helpful in understanding how a single cell transitions between distinct modes of 3D migration in response to the physical environment.
Collapse
Affiliation(s)
| | | | | | - Ryan J. Petrie
- Department of Biology, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
16
|
Kokate SB, Ciuba K, Tran VD, Kumari R, Tojkander S, Engel U, Kogan K, Kumar S, Lappalainen P. Caldesmon controls stress fiber force-balance through dynamic cross-linking of myosin II and actin-tropomyosin filaments. Nat Commun 2022; 13:6032. [PMID: 36229430 PMCID: PMC9561149 DOI: 10.1038/s41467-022-33688-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Contractile actomyosin bundles are key force-producing and mechanosensing elements in muscle and non-muscle tissues. Whereas the organization of muscle myofibrils and mechanism regulating their contractility are relatively well-established, the principles by which myosin-II activity and force-balance are regulated in non-muscle cells have remained elusive. We show that Caldesmon, an important component of smooth muscle and non-muscle cell actomyosin bundles, is an elongated protein that functions as a dynamic cross-linker between myosin-II and tropomyosin-actin filaments. Depletion of Caldesmon results in aberrant lateral movement of myosin-II filaments along actin bundles, leading to irregular myosin distribution within stress fibers. This manifests as defects in stress fiber network organization and contractility, and accompanied problems in cell morphogenesis, migration, invasion, and mechanosensing. These results identify Caldesmon as critical factor that ensures regular myosin-II spacing within non-muscle cell actomyosin bundles, and reveal how stress fiber networks are controlled through dynamic cross-linking of tropomyosin-actin and myosin filaments.
Collapse
Affiliation(s)
- Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Katarzyna Ciuba
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.,Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Vivien D Tran
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sari Tojkander
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, E318, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Ulrike Engel
- Nikon Imaging Center at Heidelberg University and Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 267, Heidelberg, 69120, Germany
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|
17
|
Ivanov AI, Lechuga S, Marino‐Melendez A, Naydenov NG. Unique and redundant functions of cytoplasmic actins and nonmuscle myosin II isoforms at epithelial junctions. Ann N Y Acad Sci 2022; 1515:61-74. [PMID: 35673768 PMCID: PMC9489603 DOI: 10.1111/nyas.14808] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The integrity and functions of epithelial barriers depend on the formation of adherens junctions (AJs) and tight junctions (TJs). A characteristic feature of AJs and TJs is their association with the cortical cytoskeleton composed of actin filaments and nonmuscle myosin II (NM-II) motors. Mechanical forces generated by the actomyosin cytoskeleton are essential for junctional assembly, stability, and remodeling. Epithelial cells express two different actin proteins and three NM-II isoforms, all known to be associated with AJs and TJs. Despite their structural similarity, different actin and NM-II isoforms have distinct biochemical properties, cellular distribution, and functions. The diversity of epithelial actins and myosin motors could be essential for the regulation of different steps of junctional formation, maturation, and disassembly. This review focuses on the roles of actin and NM-II isoforms in controlling the integrity and barrier properties of various epithelia. We discuss the effects of the depletion of individual actin isoforms and NM-II motors on the assembly and barrier function of AJs and TJs in model epithelial monolayers in vitro. We also describe the functional consequences of either total or tissue-specific gene knockout of different actins and NM-II motors, with a focus on the development and integrity of different epithelia in vivo.
Collapse
Affiliation(s)
- Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Armando Marino‐Melendez
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| |
Collapse
|
18
|
Morris T, Sue E, Geniesse C, Brieher WM, Tang VW. Synaptopodin stress fiber and contractomere at the epithelial junction. J Cell Biol 2022; 221:e202011162. [PMID: 35416930 PMCID: PMC9011326 DOI: 10.1083/jcb.202011162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.
Collapse
Affiliation(s)
- Timothy Morris
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Eva Sue
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Caleb Geniesse
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
19
|
Kage F, Vicente-Manzanares M, McEwan BC, Kettenbach AN, Higgs HN. Myosin II proteins are required for organization of calcium-induced actin networks upstream of mitochondrial division. Mol Biol Cell 2022; 33:ar63. [PMID: 35427150 PMCID: PMC9561854 DOI: 10.1091/mbc.e22-01-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The formin INF2 polymerizes a calcium-activated cytoplasmic network of actin filaments, which we refer to as calcium-induced actin polymerization (CIA). CIA plays important roles in multiple cellular processes, including mitochondrial dynamics and vesicle transport. Here, we show that nonmuscle myosin II (NMII) is activated within 60 s of calcium stimulation and rapidly recruited to the CIA network. Knockout of any individual NMII in U2OS cells affects the organization of the CIA network, as well as three downstream effects: endoplasmic-reticulum-to-mitochondrial calcium transfer, mitochondrial Drp1 recruitment, and mitochondrial division. Interestingly, while NMIIC is the least abundant NMII in U2OS cells (>200-fold less than NMIIA and >10-fold less than NMIIB), its knockout is equally deleterious to CIA. On the basis of these results, we propose that myosin II filaments containing all three NMII heavy chains exert organizational and contractile roles in the CIA network. In addition, NMIIA knockout causes a significant decrease in myosin regulatory light chain levels, which might have additional effects.
Collapse
Affiliation(s)
- Frieda Kage
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
| | - Miguel Vicente-Manzanares
- Centro de Investigacion del Cancer/Instituto de Biologia Molecular y Celular del Cancer, Centro Mixto Universidad de Salamanca, 37007 Salamanca, Spain
| | - Brennan C. McEwan
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
- Program in Cancer Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
| | - Arminja N. Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
- Program in Cancer Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
| | - Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, USA
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Garrido-Casado M, Asensio-Juárez G, Vicente-Manzanares M. Nonmuscle Myosin II Regulation Directs Its Multiple Roles in Cell Migration and Division. Annu Rev Cell Dev Biol 2021; 37:285-310. [PMID: 34314591 DOI: 10.1146/annurev-cellbio-042721-105528] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nonmuscle myosin II (NMII) is a multimeric protein complex that generates most mechanical force in eukaryotic cells. NMII function is controlled at three main levels. The first level includes events that trigger conformational changes that extend the complex to enable its assembly into filaments. The second level controls the ATPase activity of the complex and its binding to microfilaments in extended NMII filaments. The third level includes events that modulate the stability and contractility of the filaments. They all work in concert to finely control force generation inside cells. NMII is a common endpoint of mechanochemical signaling pathways that control cellular responses to physical and chemical extracellular cues. Specific phosphorylations modulate NMII activation in a context-dependent manner. A few kinases control these phosphorylations in a spatially, temporally, and lineage-restricted fashion, enabling functional adaptability to the cellular microenvironment. Here, we review mechanisms that control NMII activity in the context of cell migration and division. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - Gloria Asensio-Juárez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas, University of Salamanca, 37007 Salamanca, Spain;
| | - 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, University of Salamanca, 37007 Salamanca, Spain;
| |
Collapse
|
23
|
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
|
24
|
von Kleeck R, Castagnino P, Roberts E, Talwar S, Ferrari G, Assoian RK. Decreased vascular smooth muscle contractility in Hutchinson-Gilford Progeria Syndrome linked to defective smooth muscle myosin heavy chain expression. Sci Rep 2021; 11:10625. [PMID: 34012019 PMCID: PMC8134495 DOI: 10.1038/s41598-021-90119-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/06/2021] [Indexed: 01/12/2023] Open
Abstract
Children with Hutchinson-Gilford Progeria Syndrome (HGPS) suffer from multiple cardiovascular pathologies due to the expression of progerin, a mutant form of the nuclear envelope protein Lamin A. Progerin expression has a dramatic effect on arterial smooth muscle cells (SMCs) and results in decreased viability and increased arterial stiffness. However, very little is known about how progerin affects SMC contractility. Here, we studied the LaminAG609G/G609G mouse model of HGPS and found reduced arterial contractility at an early age that correlates with a decrease in smooth muscle myosin heavy chain (SM-MHC) mRNA and protein expression. Traction force microscopy on isolated SMCs from these mice revealed reduced force generation compared to wild-type controls; this effect was phenocopied by depletion of SM-MHC in WT SMCs and overcome by ectopic expression of SM-MHC in HGPS SMCs. Arterial SM-MHC levels are also reduced with age in wild-type mice and humans, suggesting a common defect in arterial contractility in HGPS and normal aging.
Collapse
Affiliation(s)
- Ryan von Kleeck
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paola Castagnino
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emilia Roberts
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shefali Talwar
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Giovanni Ferrari
- Departments of Surgery and Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute of Translational Medicine and Therapeutics at University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Nishimura Y, Shi S, Zhang F, Liu R, Takagi Y, Bershadsky AD, Viasnoff V, Sellers JR. The formin inhibitor SMIFH2 inhibits members of the myosin superfamily. J Cell Sci 2021; 134:237818. [PMID: 33589498 PMCID: PMC8121067 DOI: 10.1242/jcs.253708] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/03/2021] [Indexed: 12/31/2022] Open
Abstract
The small molecular inhibitor of formin FH2 domains, SMIFH2, is widely used in cell biological studies. It inhibits formin-driven actin polymerization in vitro, but not polymerization of pure actin. It is active against several types of formin from different species. Here, we found that SMIFH2 inhibits retrograde flow of myosin 2 filaments and contraction of stress fibers. We further checked the effect of SMIFH2 on non-muscle myosin 2A and skeletal muscle myosin 2 in vitro, and found that SMIFH2 inhibits activity of myosin ATPase and the ability to translocate actin filaments in the gliding actin in vitro motility assay. Inhibition of non-muscle myosin 2A in vitro required a higher concentration of SMIFH2 compared with that needed to inhibit retrograde flow and stress fiber contraction in cells. We also found that SMIFH2 inhibits several other non-muscle myosin types, including bovine myosin 10, Drosophila myosin 7a and Drosophila myosin 5, more efficiently than it inhibits formins. These off-target inhibitions demand additional careful analysis in each case when solely SMIFH2 is used to probe formin functions. This article has an associated First Person interview with Yukako Nishimura, joint first author of the paper.
Collapse
Affiliation(s)
- Yukako Nishimura
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore
| | - Shidong Shi
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore
| | - Fang Zhang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rong Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yasuharu Takagi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander D Bershadsky
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore.,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Virgile Viasnoff
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore.,CNRS UMI 3639 BMC, Singapore 117411, Singapore.,Department of Biological Sciences, National university of Singapore, Singapore 117558, Singapore
| | - James R Sellers
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Talwar S, Kant A, Xu T, Shenoy VB, Assoian RK. Mechanosensitive smooth muscle cell phenotypic plasticity emerging from a null state and the balance between Rac and Rho. Cell Rep 2021; 35:109019. [PMID: 33882318 PMCID: PMC8142933 DOI: 10.1016/j.celrep.2021.109019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/07/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Reversible differentiation of vascular smooth muscle cells (VSMCs) plays a critical role in vascular biology and disease. Changes in VSMC differentiation correlate with stiffness of the arterial extracellular matrix (ECM), but causal relationships remain unclear. We show that VSMC plasticity is mechanosensitive and that both the de-differentiated and differentiated fates are promoted by the same ECM stiffness. Differential equations developed to model this behavior predicted that a null VSMC state generates the dual fates in response to ECM stiffness. Direct measurements of cellular forces, proliferation, and contractile gene expression validated these predictions and showed that fate outcome is mediated by Rac-Rho homeostasis. Rac, through distinct effects on YAP and TAZ, is required for both fates. Rho drives the contractile state alone, so its level of activity, relative to Rac, drives phenotypic choice. Our results show how the cellular response to a single ECM stiffness generates bi-stability and VSMC plasticity. Reversible differentiation/de-differentiation of smooth muscle cells plays a critical role in vascular biology and disease. Talwar et al. show that these differentiated and de-differentiated phenotypes emerge from a null state that is regulated by ECM stiffness and bidirectional effects of Rac on YAP and TAZ transcriptional coregulators.
Collapse
Affiliation(s)
- Shefali Talwar
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Departments of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aayush Kant
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tina Xu
- Departments of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivek B Shenoy
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Richard K Assoian
- Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Departments of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Guo S, Meng L, Liu H, Yuan L, Zhao N, Ni J, Zhang Y, Ben J, Li YP, Ma J. Trio cooperates with Myh9 to regulate neural crest-derived craniofacial development. Am J Cancer Res 2021; 11:4316-4334. [PMID: 33754063 PMCID: PMC7977452 DOI: 10.7150/thno.51745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
Trio is a unique member of the Rho-GEF family that has three catalytic domains and is vital for various cellular processes in both physiological and developmental settings. TRIO mutations in humans are involved in craniofacial abnormalities, in which patients present with mandibular retrusion. However, little is known about the molecular mechanisms of Trio in neural crest cell (NCC)-derived craniofacial development, and there is still a lack of direct evidence to assign a functional role to Trio in NCC-induced craniofacial abnormalities. Methods: In vivo, we used zebrafish and NCC-specific knockout mouse models to investigate the phenotype and dynamics of NCC development in Trio morphants. In vitro, iTRAQ, GST pull-down assays, and proximity ligation assay (PLA) were used to explore the role of Trio and its potential downstream mediators in NCC migration and differentiation. Results: In zebrafish and mouse models, disruption of Trio elicited a migration deficit and impaired the differentiation of NCC derivatives, leading to craniofacial growth deficiency and mandibular retrusion. Moreover, Trio positively regulated Myh9 expression and directly interacted with Myh9 to coregulate downstream cellular signaling in NCCs. We further demonstrated that disruption of Trio or Myh9 inhibited Rac1 and Cdc42 activity, specifically affecting the nuclear export of β-catenin and NCC polarization. Remarkably, craniofacial abnormalities caused by trio deficiency in zebrafish could be partially rescued by the injection of mRNA encoding myh9, ca-Rac1, or ca-Cdc42. Conclusions: Here, we identified that Trio, interacting mostly with Myh9, acts as a key regulator of NCC migration and differentiation during craniofacial development. Our results indicate that trio morphant zebrafish and Wnt1-cre;Triofl/fl mice offer potential model systems to facilitate the study of the pathogenic mechanisms of Trio mutations causing craniofacial abnormalities.
Collapse
|
28
|
Grewe J, Schwarz US. Mechanosensitive self-assembly of myosin II minifilaments. Phys Rev E 2021; 101:022402. [PMID: 32168598 DOI: 10.1103/physreve.101.022402] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/15/2020] [Indexed: 01/23/2023]
Abstract
Self-assembly and force generation are two central processes in biological systems that usually are considered in separation. However, the signals that activate nonmuscle myosin II molecular motors simultaneously lead to self-assembly into myosin II minifilaments as well as progression of the motor heads through the cross-bridge cycle. Here we investigate theoretically the possible effects of coupling these two processes. Our assembly model, which builds on a consensus architecture of the minifilament, predicts a critical aggregation concentration at which the assembly kinetics slows down dramatically. The combined model predicts that increasing actin filament concentration and force both lead to a decrease in the critical aggregation concentration. We suggest that due to these effects, myosin II minifilaments in a filamentous context might be in a critical state that reacts faster to varying conditions than in solution. We finally compare our model to experiments by simulating fluorescence recovery after photobleaching.
Collapse
Affiliation(s)
- Justin Grewe
- Institute for Theoretical Physics and Bioquant, Heidelberg University, Heidelberg, Germany
| | - Ulrich S Schwarz
- Institute for Theoretical Physics and Bioquant, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
29
|
Hippler M, Weißenbruch K, Richler K, Lemma ED, Nakahata M, Richter B, Barner-Kowollik C, Takashima Y, Harada A, Blasco E, Wegener M, Tanaka M, Bastmeyer M. Mechanical stimulation of single cells by reversible host-guest interactions in 3D microscaffolds. SCIENCE ADVANCES 2020; 6:6/39/eabc2648. [PMID: 32967835 PMCID: PMC7531888 DOI: 10.1126/sciadv.abc2648] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/07/2020] [Indexed: 05/19/2023]
Abstract
Many essential cellular processes are regulated by mechanical properties of their microenvironment. Here, we introduce stimuli-responsive composite scaffolds fabricated by three-dimensional (3D) laser lithography to simultaneously stretch large numbers of single cells in tailored 3D microenvironments. The key material is a stimuli-responsive photoresist containing cross-links formed by noncovalent, directional interactions between β-cyclodextrin (host) and adamantane (guest). This allows reversible actuation under physiological conditions by application of soluble competitive guests. Cells adhering in these scaffolds build up initial traction forces of ~80 nN. After application of an equibiaxial stretch of up to 25%, cells remodel their actin cytoskeleton, double their traction forces, and equilibrate at a new dynamic set point within 30 min. When the stretch is released, traction forces gradually decrease until the initial set point is retrieved. Pharmacological inhibition or knockout of nonmuscle myosin 2A prevents these adjustments, suggesting that cellular tensional homeostasis strongly depends on functional myosin motors.
Collapse
Affiliation(s)
- Marc Hippler
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany.
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Kai Weißenbruch
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Kai Richler
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Enrico D Lemma
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Masaki Nakahata
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Benjamin Richter
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Christopher Barner-Kowollik
- Centre for Materials Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia
- School of Chemistry and Physics, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia
- Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Yoshinori Takashima
- Department of Macromolecular Science, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Akira Harada
- Department of Macromolecular Science, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Eva Blasco
- Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
- Institute for Chemical Technology and Polymer Chemistry, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Martin Wegener
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany.
- Institute for Chemical Technology and Polymer Chemistry, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| | - Motomu Tanaka
- Institute of Physical Chemistry, Heidelberg University, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany.
- Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan
| | - Martin Bastmeyer
- Zoological Institute, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany.
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), 76128 Karlsruhe, Germany
| |
Collapse
|
30
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
31
|
Rao TC, Ma VPY, Blanchard A, Urner TM, Grandhi S, Salaita K, Mattheyses AL. EGFR activation attenuates the mechanical threshold for integrin tension and focal adhesion formation. J Cell Sci 2020; 133:jcs238840. [PMID: 32546532 PMCID: PMC7358133 DOI: 10.1242/jcs.238840] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Mechanical forces, growth factors and the extracellular matrix all play crucial roles in cell adhesion. To understand how epidermal growth factor receptor (EGFR) impacts the mechanics of adhesion, we employed tension gauge tether (TGT) probes displaying the integrin ligand cRGDfK and quantified integrin tension. EGF exposure significantly increased spread area, cell circularity, integrated integrin tension, mechanical rupture density, radial organization and size of focal adhesions in Cos-7 cells on TGT surfaces. These findings suggest that EGFR regulates integrin tension and the spatial organization of focal adhesions. Additionally, we found that the mechanical tension threshold for outside-in integrin activation is tunable by EGFR. Parallel genetic and pharmacologic strategies demonstrated that these phenotypes are driven by ligand-dependent EGFR signaling. Our results establish a novel mechanism whereby EGFR regulates integrin activation and cell adhesion, providing control over cellular responses to the environment.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tejeshwar C Rao
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Aaron Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Tara M Urner
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Shreya Grandhi
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Alexa L Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
32
|
Non-Muscle Myosin 2A (NM2A): Structure, Regulation and Function. Cells 2020; 9:cells9071590. [PMID: 32630196 PMCID: PMC7408548 DOI: 10.3390/cells9071590] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022] Open
Abstract
Non-muscle myosin 2A (NM2A) is a motor cytoskeletal enzyme with crucial importance from the early stages of development until adulthood. Due to its capacity to convert chemical energy into force, NM2A powers the contraction of the actomyosin cytoskeleton, required for proper cell division, adhesion and migration, among other cellular functions. Although NM2A has been extensively studied, new findings revealed that a lot remains to be discovered concerning its spatiotemporal regulation in the intracellular environment. In recent years, new functions were attributed to NM2A and its activity was associated to a plethora of illnesses, including neurological disorders and infectious diseases. Here, we provide a concise overview on the current knowledge regarding the structure, the function and the regulation of NM2A. In addition, we recapitulate NM2A-associated diseases and discuss its potential as a therapeutic target.
Collapse
|
33
|
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
|
34
|
Replicative senescence in MSCWJ-1 human umbilical cord mesenchymal stem cells is marked by characteristic changes in motility, cytoskeletal organization, and RhoA localization. Mol Biol Rep 2020; 47:3867-3883. [PMID: 32372170 DOI: 10.1007/s11033-020-05476-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
Here, we document changes in cell motility and organization of the contractile apparatus of human umbilical cord Wharton's jelly mesenchymal stem cells (MSCWJ-1) in the process of replicative senescence. Colocalization dynamics of F-actin and actin-binding proteins (myosin-9, α-actinin-4, RhoA) were examined in the MSCWJ-1 cell line. The results show that nuclear-cytoplasmic redistribution of RhoA occurs during replicative senescence, with maximal RhoA/nucleus colocalization evident at passage 15. At that time point, decreases in colocalization, namely myosin-9/F-actin and α-actinin-4/F-actin, were seen and myosin-9 was found in cytosolic extracts in the assembly-incompetent form. Using an automated intravital confocal cytometry system and quantitative analysis of MSCWJ-1 movements, we found that changes in cytoskeletal organization correlate with cell motility characteristics over a time period from passages 9 to 38. The factors examined (cytoskeleton structure, cell motility) indicate that the process by which cells transition to replicative senescence is best represented as three stages. The first stage lasts from cell culture isolation to passage 15 and is characterized by: accumulation of actin-binding proteins in assembly-incompetent forms; nuclear RhoA accumulation; and an increase in movement tortuosity. The second stage extends from passages 15 to 28 and is characterized by: an increase in the structural integrity of the actin cytoskeleton; exit of RhoA and alpha-actinin-4 from the nucleus; and a decrease in path tortuosity. The third stage extends from passage 28 to 38 and is marked by: a plateau in actin cytoskeleton structural integrity; significant decreases in nuclear RhoA levels; and decreases in cell speed.
Collapse
|
35
|
Mosby LS, Hundt N, Young G, Fineberg A, Polin M, Mayor S, Kukura P, Köster DV. Myosin II Filament Dynamics in Actin Networks Revealed with Interferometric Scattering Microscopy. Biophys J 2020; 118:1946-1957. [PMID: 32191863 PMCID: PMC7175421 DOI: 10.1016/j.bpj.2020.02.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 11/30/2022] Open
Abstract
The plasma membrane and the underlying cytoskeletal cortex constitute active platforms for a variety of cellular processes. Recent work has shown that the remodeling acto-myosin network modifies local membrane organization, but the molecular details are only partly understood because of difficulties with experimentally accessing the relevant time and length scales. Here, we use interferometric scattering microscopy to investigate a minimal acto-myosin network linked to a supported lipid bilayer membrane. Using the magnitude of the interferometric contrast, which is proportional to molecular mass, and fast acquisition rates, we detect and image individual membrane-attached actin filaments diffusing within the acto-myosin network and follow individual myosin II filament dynamics. We quantify myosin II filament dwell times and processivity as functions of ATP concentration, providing experimental evidence for the predicted ensemble behavior of myosin head domains. Our results show how decreasing ATP concentrations lead to both increasing dwell times of individual myosin II filaments and a global change from a remodeling to a contractile state of the acto-myosin network.
Collapse
Affiliation(s)
- Lewis S Mosby
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, United Kingdom; Physics Department, University of Warwick, Coventry, United Kingdom
| | - Nikolas Hundt
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Gavin Young
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Adam Fineberg
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Marco Polin
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, United Kingdom; Physics Department, University of Warwick, Coventry, United Kingdom
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, India; Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.
| | - Philipp Kukura
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom.
| | - Darius V Köster
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, United Kingdom; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
36
|
Yang B, Wolfenson H, Chung VY, Nakazawa N, Liu S, Hu J, Huang RYJ, Sheetz MP. Stopping transformed cancer cell growth by rigidity sensing. NATURE MATERIALS 2020; 19:239-250. [PMID: 31659296 PMCID: PMC7477912 DOI: 10.1038/s41563-019-0507-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 09/11/2019] [Indexed: 05/16/2023]
Abstract
A common feature of cancer cells is the alteration of kinases and biochemical signalling pathways enabling transformed growth on soft matrices, whereas cytoskeletal protein alterations are thought to be a secondary issue. However, we report here that cancer cells from different tissues can be toggled between transformed and rigidity-dependent growth states by the absence or presence of mechanosensory modules, respectively. In various cancer lines from different tissues, cells had over tenfold fewer rigidity-sensing contractions compared with normal cells from the same tissues. Restoring normal levels of cytoskeletal proteins, including tropomyosins, restored rigidity sensing and rigidity-dependent growth. Further depletion of other rigidity sensor proteins, including myosin IIA, restored transformed growth and blocked sensing. In addition, restoration of rigidity sensing to cancer cells inhibited tumour formation and changed expression patterns. Thus, the depletion of rigidity-sensing modules through alterations in cytoskeletal protein levels enables cancer cell growth on soft surfaces, which is an enabling factor for cancer progression.
Collapse
Affiliation(s)
- Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel of Technology, Haifa, Israel
| | - Vin Yee Chung
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Naotaka Nakazawa
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Shuaimin Liu
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Junqiang Hu
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
37
|
Sato Y, Kamijo K, Tsutsumi M, Murakami Y, Takahashi M. Nonmuscle myosin IIA and IIB differently suppress microtubule growth to stabilize cell morphology. J Biochem 2019; 167:25-39. [DOI: 10.1093/jb/mvz082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/22/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Precise regulation of cytoskeletal dynamics is important in many fundamental cellular processes such as cell shape determination. Actin and microtubule (MT) cytoskeletons mutually regulate their stability and dynamics. Nonmuscle myosin II (NMII) is a candidate protein that mediates the actin–MT crosstalk. NMII regulates the stability and dynamics of actin filaments to control cell morphology. Additionally, previous reports suggest that NMII-dependent cellular contractility regulates MT dynamics, and MTs also control cell morphology; however, the detailed mechanism whereby NMII regulates MT dynamics and the relationship among actin dynamics, MT dynamics and cell morphology remain unclear. The present study explores the roles of two well-characterized NMII isoforms, NMIIA and NMIIB, on the regulation of MT growth dynamics and cell morphology. We performed RNAi and drug experiments and demonstrated the NMII isoform-specific mechanisms—NMIIA-dependent cellular contractility upregulates the expression of some mammalian diaphanous-related formin (mDia) proteins that suppress MT dynamics; NMIIB-dependent inhibition of actin depolymerization suppresses MT growth independently of cellular contractility. The depletion of either NMIIA or NMIIB resulted in the increase in cellular morphological dynamicity, which was alleviated by the perturbation of MT dynamics. Thus, the NMII-dependent control of cell morphology significantly relies on MT dynamics.
Collapse
Affiliation(s)
- Yuta Sato
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Kita 13, Nishi 8, Kita-ku, Sapporo Hokkaido, Japan
| | - Keiju Kamijo
- Division of Anatomy and Cell Biology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai Miyagi, Japan
| | - Motosuke Tsutsumi
- Research Institute for Electronic Science, Hokkaido University, Kita 20, Nishi 10, Kita-ku, Sapporo Hokkaido, Japan
| | - Yota Murakami
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Kita 13, Nishi 8, Kita-ku, Sapporo Hokkaido, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo Hokkaido, Japan
| | - Masayuki Takahashi
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Kita 13, Nishi 8, Kita-ku, Sapporo Hokkaido, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo Hokkaido, Japan
| |
Collapse
|
38
|
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
|
39
|
Kothari P, Johnson C, Sandone C, Iglesias PA, Robinson DN. How the mechanobiome drives cell behavior, viewed through the lens of control theory. J Cell Sci 2019; 132:jcs234476. [PMID: 31477578 PMCID: PMC6771144 DOI: 10.1242/jcs.234476] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cells have evolved sophisticated systems that integrate internal and external inputs to coordinate cell shape changes during processes, such as development, cell identity determination, and cell and tissue homeostasis. Cellular shape-change events are driven by the mechanobiome, the network of macromolecules that allows cells to generate, sense and respond to externally imposed and internally generated forces. Together, these components build the cellular contractility network, which is governed by a control system. Proteins, such as non-muscle myosin II, function as both sensors and actuators, which then link to scaffolding proteins, transcription factors and metabolic proteins to create feedback loops that generate the foundational mechanical properties of the cell and modulate cellular behaviors. In this Review, we highlight proteins that establish and maintain the setpoint, or baseline, for the control system and explore the feedback loops that integrate different cellular processes with cell mechanics. Uncovering the genetic, biophysical and biochemical interactions between these molecular components allows us to apply concepts from control theory to provide a systems-level understanding of cellular processes. Importantly, the actomyosin network has emerged as more than simply a 'downstream' effector of linear signaling pathways. Instead, it is also a significant driver of cellular processes traditionally considered to be 'upstream'.
Collapse
Affiliation(s)
- Priyanka Kothari
- Departments of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cecilia Johnson
- Art as Applied to Medicine, Johns Hopkins University School of Medicine, Baltimore, M 21205, USA
| | - Corinne Sandone
- Art as Applied to Medicine, Johns Hopkins University School of Medicine, Baltimore, M 21205, USA
| | - Pablo A Iglesias
- Departments of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Douglas N Robinson
- Departments of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
40
|
Zhovmer AS, Tabdanov ED, Miao H, Wen H, Chen J, Luo X, Ma X, Provenzano PP, Adelstein RS. The role of nonmuscle myosin 2A and 2B in the regulation of mesenchymal cell contact guidance. Mol Biol Cell 2019; 30:1961-1973. [PMID: 31318315 PMCID: PMC6727766 DOI: 10.1091/mbc.e19-01-0071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Contact guidance refers to the ability of cells to sense the geometrical features of the microenvironment and respond by changing their shape and adopting the appropriate orientation. Inhibition and ablation of nonmuscle myosin 2 (NM2) paralogues have demonstrated their importance for contact guidance. However, the specific roles of the NM2 paralogues have not been systematically studied. In this work we use micropatterned substrates to examine the roles of NM2A and NM2B and to elucidate the relationship of the microenvironment, actomyosin, and microtubules in contact guidance. We show that contact guidance is preserved following loss of NM2B and that expression of NM2A alone is sufficient to establish an appropriate orientation of the cells. Loss of NM2B and overexpression of NM2A result in a prominent cell polarization that is found to be linked to the increased alignment of microtubules with the actomyosin scaffold. Suppression of actomyosin with blebbistatin reduces cell polarity on a flat surface, but not on a surface with contact guidance cues. This indicates that the lost microtubule-actomyosin interactions are compensated for by microtubule-microenvironment interactions, which are sufficient to establish cell polarity through contact guidance.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Erdem D Tabdanov
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Houxun Miao
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Han Wen
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Jinqiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Paolo P Provenzano
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| |
Collapse
|
41
|
Ultrastructure and dynamics of the actin-myosin II cytoskeleton during mitochondrial fission. Nat Cell Biol 2019; 21:603-613. [PMID: 30988424 PMCID: PMC6499663 DOI: 10.1038/s41556-019-0313-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/14/2019] [Indexed: 02/05/2023]
Abstract
Mitochondrial fission involves the preconstriction of an organelle followed by scission by dynamin-related protein Drp1. Preconstriction is facilitated by actin and non-muscle myosin II through a mechanism that remains unclear, largely due to the unknown cytoskeletal ultrastructure at mitochondrial constrictions. Here, using platinum replica electron microscopy, we show that mitochondria in cells are embedded in an interstitial cytoskeletal network that contains abundant unbranched actin filaments. Both spontaneous and induced mitochondrial constrictions typically associate with a criss-cross array of long actin filaments that comprise part of this interstitial network. Non-muscle myosin II is found adjacent to mitochondria but is not specifically enriched at the constriction sites. During ionomycin-induced mitochondrial fission, F-actin clouds colocalize with mitochondrial constriction sites, whereas dynamic myosin II clouds are present in the vicinity of constrictions. We propose that myosin II promotes mitochondrial constriction by inducing stochastic deformations of the interstitial actin network, which applies pressure on the mitochondrial surface and thus initiates curvature-sensing mechanisms that complete mitochondrial constriction.
Collapse
|
42
|
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
|
43
|
Sao K, Jones TM, Doyle AD, Maity D, Schevzov G, Chen Y, Gunning PW, Petrie RJ. Myosin II governs intracellular pressure and traction by distinct tropomyosin-dependent mechanisms. Mol Biol Cell 2019; 30:1170-1181. [PMID: 30865560 PMCID: PMC6724525 DOI: 10.1091/mbc.e18-06-0355] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Two-dimensional (2D) substrate rigidity promotes myosin II activity to increase traction force in a process negatively regulated by tropomyosin (Tpm) 2.1. We recently discovered that actomyosin contractility can increase intracellular pressure and switch tumor cells from low-pressure lamellipodia to high-pressure lobopodial protrusions during three-dimensional (3D) migration. However, it remains unclear whether these myosin II–generated cellular forces are produced simultaneously, and by the same molecular machinery. Here we identify Tpm 1.6 as a positive regulator of intracellular pressure and confirm that Tpm 2.1 is a negative regulator of traction force. We find that Tpm 1.6 and 2.1 can control intracellular pressure and traction independently, suggesting these myosin II–dependent forces are generated by distinct mechanisms. Further, these tropomyosin-regulated mechanisms can be integrated to control complex cell behaviors on 2D and in 3D environments.
Collapse
Affiliation(s)
- Kimheak Sao
- Department of Biology, Drexel University, Philadelphia, PA 19104
| | - Tia M Jones
- Department of Biology, Drexel University, Philadelphia, PA 19104
| | - Andrew D Doyle
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Debonil Maity
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Galina Schevzov
- School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Peter W Gunning
- School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | - Ryan J Petrie
- Department of Biology, Drexel University, Philadelphia, PA 19104
| |
Collapse
|
44
|
Dasbiswas K, Hu S, Schnorrer F, Safran SA, Bershadsky AD. Ordering of myosin II filaments driven by mechanical forces: experiments and theory. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0114. [PMID: 29632266 DOI: 10.1098/rstb.2017.0114] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/27/2022] Open
Abstract
Myosin II filaments form ordered superstructures in both cross-striated muscle and non-muscle cells. In cross-striated muscle, myosin II (thick) filaments, actin (thin) filaments and elastic titin filaments comprise the stereotypical contractile units of muscles called sarcomeres. Linear chains of sarcomeres, called myofibrils, are aligned laterally in registry to form cross-striated muscle cells. The experimentally observed dependence of the registered organization of myofibrils on extracellular matrix elasticity has been proposed to arise from the interactions of sarcomeric contractile elements (considered as force dipoles) through the matrix. Non-muscle cells form small bipolar filaments built of less than 30 myosin II molecules. These filaments are associated in registry forming superstructures ('stacks') orthogonal to actin filament bundles. Formation of myosin II filament stacks requires the myosin II ATPase activity and function of the actin filament crosslinking, polymerizing and depolymerizing proteins. We propose that the myosin II filaments embedded into elastic, intervening actin network (IVN) function as force dipoles that interact attractively through the IVN. This is in analogy with the theoretical picture developed for myofibrils where the elastic medium is now the actin cytoskeleton itself. Myosin stack formation in non-muscle cells provides a novel mechanism for the self-organization of the actin cytoskeleton at the level of the entire cell.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Kinjal Dasbiswas
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shiqiong Hu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Samuel A Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore .,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
45
|
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
|
46
|
Jiu Y, Kumari R, Fenix AM, Schaible N, Liu X, Varjosalo M, Krishnan R, Burnette DT, Lappalainen P. Myosin-18B Promotes the Assembly of Myosin II Stacks for Maturation of Contractile Actomyosin Bundles. Curr Biol 2018; 29:81-92.e5. [PMID: 30581023 DOI: 10.1016/j.cub.2018.11.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/12/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Abstract
Cell adhesion, morphogenesis, mechanosensing, and muscle contraction rely on contractile actomyosin bundles, where the force is produced through sliding of bipolar myosin II filaments along actin filaments. The assembly of contractile actomyosin bundles involves registered alignment of myosin II filaments and their subsequent fusion into large stacks. However, mechanisms underlying the assembly of myosin II stacks and their physiological functions have remained elusive. Here, we identified myosin-18B, an unconventional myosin, as a stable component of contractile stress fibers. Myosin-18B co-localized with myosin II motor domains in stress fibers and was enriched at the ends of myosin II stacks. Importantly, myosin-18B deletion resulted in drastic defects in the concatenation and persistent association of myosin II filaments with each other and thus led to severely impaired assembly of myosin II stacks. Consequently, lack of myosin-18B resulted in defective maturation of actomyosin bundles from their precursors in osteosarcoma cells. Moreover, myosin-18B knockout cells displayed abnormal morphogenesis, migration, and ability to exert forces to the environment. These results reveal a critical role for myosin-18B in myosin II stack assembly and provide evidence that myosin II stacks are important for a variety of vital processes in cells.
Collapse
Affiliation(s)
- Yaming Jiu
- Institute of Biotechnology, P.O. Box 56, University of Helsinki, 00014 Helsinki, Finland; CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Reena Kumari
- Institute of Biotechnology, P.O. Box 56, University of Helsinki, 00014 Helsinki, Finland
| | - Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Niccole Schaible
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaonan Liu
- Institute of Biotechnology, P.O. Box 56, University of Helsinki, 00014 Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, P.O. Box 56, University of Helsinki, 00014 Helsinki, Finland
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, P.O. Box 56, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
47
|
Wang Y, Yang Q, Cheng Y, Gao M, Kuang L, Wang C. Myosin Heavy Chain 10 (MYH10) Gene Silencing Reduces Cell Migration and Invasion in the Glioma Cell Lines U251, T98G, and SHG44 by Inhibiting the Wnt/β-Catenin Pathway. Med Sci Monit 2018; 24:9110-9119. [PMID: 30552850 PMCID: PMC6319164 DOI: 10.12659/msm.911523] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The myosin heavy chain 10 or MYH10 gene encodes non-muscle myosin II B (NM IIB), and is involved in tumor cell migration, invasion, extracellular matrix (ECM) production, and epithelial-mesenchymal transition (EMT). This study aimed to investigate the effects of the MYH10 gene on normal human glial cells and glioma cell lines in vitro, by gene silencing, and to determine the signaling pathways involved. Material/Methods The normal human glial cell line HEB, and the glioma cell lines, U251, T98G, and SHG44 were studied. Plasmid transfection silenced the MYH10 gene. The cell counting kit-8 (CCK-8) assay evaluated cell viability. Cell migration and invasion were evaluated using scratch and transwell assays. Western blot measured the protein expression levels, and quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the mRNA expression levels, for MYH10, metastasis-associated protein 1 (MTA-1), matrix metalloproteinase (MMP)-1, MMP-9, tissue inhibitor of metalloproteinases 2 (TIMP2), collagen 1, E-cadherin, vimentin, Wnt3a, β-catenin, and cyclin D1. Results The MYH10 gene was overexpressed in U251, T98G, and SHG44 cells. MYH10 expression was down-regulated following siMYH10 plasmid interference, which also inhibited glioma cell migration and invasion. MYH10 gene silencing resulted in reduced expression of MTA-1, MPP-2, MMP-9 and vimentin, and increased expression of TIMP-2, E-cadherin and collagen 1 at the protein and mRNA level, and inhibited the Wnt/β-catenin pathway. Conclusions In human glioma cell lines, silencing the MYH10 gene reduced cell migration and invasion, by inhibiting the Wnt/β-catenin pathway, which may regulate the ECM and inhibit EMT in human glioma.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurosurgery, 2nd Ward, Taihe Hospital, Shiyan, Hubei, China (mainland)
| | - Qi Yang
- Department of Orthopedic Surgery, 3rd Ward, Taihe Hospital, Shiyan, Hubei, China (mainland)
| | - Yanli Cheng
- Skin Department, Taihe Hospital, Shiyan, Hubei, China (mainland)
| | - Meng Gao
- Department of Ophthalmology and Otolaryngology, Weifang Maternal and Child Health Care Hospital, Weifang, Shandong, China (mainland)
| | - Lei Kuang
- Department of Neurosurgery, 3rd Ward, Taihe Hospital, Shiyan, Hubei, China (mainland)
| | - Chun Wang
- Department of Neurosurgery, Suizhou Central Hospital, Suizhou, Hubei, China (mainland)
| |
Collapse
|
48
|
Fenix AM, Neininger AC, Taneja N, Hyde K, Visetsouk MR, Garde RJ, Liu B, Nixon BR, Manalo AE, Becker JR, Crawley SW, Bader DM, Tyska MJ, Liu Q, Gutzman JH, Burnette DT. Muscle-specific stress fibers give rise to sarcomeres in cardiomyocytes. eLife 2018; 7:42144. [PMID: 30540249 PMCID: PMC6307863 DOI: 10.7554/elife.42144] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/11/2018] [Indexed: 11/13/2022] Open
Abstract
The sarcomere is the contractile unit within cardiomyocytes driving heart muscle contraction. We sought to test the mechanisms regulating actin and myosin filament assembly during sarcomere formation. Therefore, we developed an assay using human cardiomyocytes to monitor sarcomere assembly. We report a population of muscle stress fibers, similar to actin arcs in non-muscle cells, which are essential sarcomere precursors. We show sarcomeric actin filaments arise directly from muscle stress fibers. This requires formins (e.g., FHOD3), non-muscle myosin IIA and non-muscle myosin IIB. Furthermore, we show short cardiac myosin II filaments grow to form ~1.5 μm long filaments that then 'stitch' together to form the stack of filaments at the core of the sarcomere (i.e., the A-band). A-band assembly is dependent on the proper organization of actin filaments and, as such, is also dependent on FHOD3 and myosin IIB. We use this experimental paradigm to present evidence for a unifying model of sarcomere assembly.
Collapse
Affiliation(s)
- Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Abigail C Neininger
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Karren Hyde
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Mike R Visetsouk
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Ryan J Garde
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Baohong Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, United States
| | - Benjamin R Nixon
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Annabelle E Manalo
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Jason R Becker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Scott W Crawley
- Department of Biological Sciences, The University of Toledo, Toledo, United States
| | - David M Bader
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Qi Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, United States
| | - Jennifer H Gutzman
- Department of Biological Sciences, Cell and Molecular Biology, University of Wisconsin Milwaukee, Milwaukee, United States
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| |
Collapse
|
49
|
Fenix AM, Burnette DT. Assembly of myosin II filament arrays: Network Contraction versus Expansion. Cytoskeleton (Hoboken) 2018; 75:545-549. [PMID: 30126071 PMCID: PMC6368487 DOI: 10.1002/cm.21487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/31/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022]
Abstract
How cellular contractile systems assemble has fascinated scientists for generations. The major molecule responsible for cellular force generation is the molecular motor, non-muscle myosin II (NMII). NMII molecules are organized into single myosin filaments and larger arrays of filaments called NMII stacks, which are capable of generating increasing amounts of force. The textbook model of NMII stack assembly is the Network Contraction Model, where ensembles of distinct NMII filaments condense into a NMII stack by pulling on actin filaments. While this model has been widely accepted for ~20 years, it has been difficult to test inside cells due to the small size of NMII filaments. Recently, interest in how NMII stacks form has been reinvigorated by the advent of super-resolution microscopy techniques which have afforded unprecedented resolution of NMII filaments inside cells. A number of recent publications using these techniques have called into question key aspects of the Network Contraction Model, and our understanding of how NMII stacks assemble.
Collapse
Affiliation(s)
- Aidan M. Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine
| |
Collapse
|
50
|
Ecsédi P, Billington N, Pálfy G, Gógl G, Kiss B, Bulyáki É, Bodor A, Sellers JR, Nyitray L. Multiple S100 protein isoforms and C-terminal phosphorylation contribute to the paralog-selective regulation of nonmuscle myosin 2 filaments. J Biol Chem 2018; 293:14850-14867. [PMID: 30087119 PMCID: PMC6153290 DOI: 10.1074/jbc.ra118.004277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/06/2018] [Indexed: 12/27/2022] Open
Abstract
Nonmuscle myosin 2 (NM2) has three paralogs in mammals, NM2A, NM2B, and NM2C, which have both unique and overlapping functions in cell migration, formation of cell-cell adhesions, and cell polarity. Their assembly into homo- and heterotypic bipolar filaments in living cells is primarily regulated by phosphorylation of the N-terminally bound regulatory light chain. Here, we present evidence that the equilibrium between these filaments and single NM2A and NM2B molecules can be controlled via S100 calcium-binding protein interactions and phosphorylation at the C-terminal end of the heavy chains. Furthermore, we show that in addition to S100A4, other members of the S100 family can also mediate disassembly of homotypic NM2A filaments. Importantly, these proteins can selectively remove NM2A molecules from heterotypic filaments. We also found that tail phosphorylation (at Ser-1956 and Ser-1975) of NM2B by casein kinase 2, as well as phosphomimetic substitutions at sites targeted by protein kinase C (PKC) and transient receptor potential cation channel subfamily M member 7 (TRPM7), down-regulates filament assembly in an additive fashion. Tail phosphorylation of NM2A had a comparatively minor effect on filament stability. S100 binding and tail phosphorylation therefore preferentially disassemble NM2A and NM2B, respectively. These two distinct mechanisms are likely to contribute to the temporal and spatial sorting of the two NM2 paralogs within heterotypic filaments. The existence of multiple NM2A-depolymerizing S100 paralogs offers the potential for diverse regulatory inputs modulating NM2A filament disassembly in cells and provides functional redundancy under both physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Neil Billington
- the Laboratory of Physiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Gyula Pálfy
- the Laboratory of Structural Chemistry and Biology, Institute of Chemistry, and
| | | | | | - Éva Bulyáki
- From the Department of Biochemistry
- the ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/C, 1117 Budapest, Hungary and
| | - Andrea Bodor
- the Laboratory of Structural Chemistry and Biology, Institute of Chemistry, and
| | - James R Sellers
- the Laboratory of Physiology, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|