1
|
Hart RG, Kota D, Li F, Zhang M, Ramallo D, Price AJ, Otterpohl KL, Smith SJ, Dunn AR, Huising MO, Liu J, Chandrasekar I. Myosin II tension sensors visualize force generation within the actin cytoskeleton in living cells. J Cell Sci 2024; 137:jcs262281. [PMID: 39369303 PMCID: PMC11698044 DOI: 10.1242/jcs.262281] [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: 05/20/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Nonmuscle myosin II (NMII) generates cytoskeletal forces that drive cell division, embryogenesis, muscle contraction and many other cellular functions. However, at present there is no method that can directly measure the forces generated by myosins in living cells. Here, we describe a Förster resonance energy transfer (FRET)-based tension sensor that can detect myosin-associated force along the filamentous actin network. Fluorescence lifetime imaging microscopy (FLIM)-FRET measurements indicate that the forces generated by NMII isoform B (NMIIB) exhibit significant spatial and temporal heterogeneity as a function of donor lifetime and fluorophore energy exchange. These measurements provide a proxy for inferred forces that vary widely along the actin cytoskeleton. This initial report highlights the potential utility of myosin-based tension sensors in elucidating the roles of cytoskeletal contractility in a wide variety of contexts.
Collapse
Affiliation(s)
- Ryan G. Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Fangjia Li
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Mengdi Zhang
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Diego Ramallo
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Andrew J. Price
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Karla L. Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steve J. Smith
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Alexander R. Dunn
- Department of Chemical Biology, Stanford University, Stanford, CA 94305, USA
| | - Mark O. Huising
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jing Liu
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 46907
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
2
|
Wang X, Baster Z, Azizi L, Li L, Rajfur Z, Hytönen VP, Huang C. Talin2 binds to non-muscle myosin IIa and regulates cell attachment and fibronectin secretion. Sci Rep 2024; 14:20175. [PMID: 39215026 PMCID: PMC11364542 DOI: 10.1038/s41598-024-70866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Talin2 is localized to large focal adhesions and is indispensable for traction force generation, invadopodium formation, cell invasion as well as metastasis. Talin2 has a higher affinity toward β-integrin tails than talin1. Moreover, disruption of the talin2-β-integrin interaction inhibits traction force generation, invadopodium formation and cell invasion, indicating that a strong talin2-β-integrin interaction is required for talin2 to fulfill these functions. Nevertheless, the role of talin2 in mediation of these processes remains unknown. Here we show that talin2 binds to the N-terminus of non-muscle myosin IIA (NMIIA) through its F3 subdomain. Moreover, talin2 co-localizes with NMIIA at cell edges as well as at some cytoplasmic spots. Talin2 also co-localizes with cortactin, an invadopodium marker. Furthermore, overexpression of NMIIA promoted the talin2 head binding to the β1-integrin tail, whereas knockdown of NMIIA reduced fibronectin and matrix metalloproteinase secretion as well as inhibited cell attachment on fibronectin-coated substrates. These results suggest that talin2 binds to NMIIA to control the secretion of extracellular matrix proteins and this interaction modulates cell adhesion.
Collapse
Affiliation(s)
- Xiaochuan Wang
- The Second Hospital of Shandong University, Jinan, 250033, Shandong, China.
| | - Zbigniew Baster
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | - Liqing Li
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348, Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| | - Cai Huang
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40506, USA.
- Doer Biologics Inc, 2nd Floor, Building 3, Hexiang Science and Technology Center, Medicine Port Town, Qiantang District, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
3
|
Patwardhan R, Nanda S, Wagner J, Stockter T, Dehmelt L, Nalbant P. Cdc42 activity in the trailing edge is required for persistent directional migration of keratinocytes. Mol Biol Cell 2024; 35:br1. [PMID: 37910204 PMCID: PMC10881163 DOI: 10.1091/mbc.e23-08-0318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Fibroblasts migrate discontinuously by generating transient leading-edge protrusions and irregular, abrupt retractions of a narrow trailing edge. In contrast, keratinocytes migrate persistently and directionally via a single, stable, broad protrusion paired with a stable trailing-edge. The Rho GTPases Rac1, Cdc42 and RhoA are key regulators of cell protrusions and retractions. However, how these molecules mediate cell-type specific migration modes is still poorly understood. In fibroblasts, all three Rho proteins are active at the leading edge, suggesting short-range coordination of protrusive Rac1 and Cdc42 signals with RhoA retraction signals. Here, we show that Cdc42 was surprisingly active in the trailing-edge of migrating keratinocytes. Elevated Cdc42 activity colocalized with the effectors MRCK and N-WASP suggesting that Cdc42 controls both myosin activation and actin polymerization in the back. Indeed, Cdc42 was required to maintain the highly dynamic contractile acto-myosin retrograde flow at the trailing edge of keratinocytes, and its depletion induced ectopic protrusions in the back, leading to decreased migration directionality. These findings suggest that Cdc42 is required to stabilize the dynamic cytoskeletal polarization in keratinocytes, to enable persistent, directional migration.
Collapse
Affiliation(s)
- Rutuja Patwardhan
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Suchet Nanda
- TU Dortmund University, Fakultät für Chemie und Chemische Biologie, 44227 Dortmund, Germany
| | - Jessica Wagner
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Tom Stockter
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Leif Dehmelt
- TU Dortmund University, Fakultät für Chemie und Chemische Biologie, 44227 Dortmund, Germany
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| |
Collapse
|
4
|
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
|
5
|
Baldwin A, Popov IK, Keller R, Wallingford J, Chang C. The RhoGEF protein Plekhg5 regulates medioapical and junctional actomyosin dynamics of apical constriction during Xenopus gastrulation. Mol Biol Cell 2023; 34:ar64. [PMID: 37043306 PMCID: PMC10295481 DOI: 10.1091/mbc.e22-09-0411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
Apical constriction results in apical surface reduction in epithelial cells and is a widely used mechanism for epithelial morphogenesis. Both medioapical and junctional actomyosin remodeling are involved in apical constriction, but the deployment of medial versus junctional actomyosin and their genetic regulation in vertebrate embryonic development have not been fully described. In this study, we investigate actomyosin dynamics and their regulation by the RhoGEF protein Plekhg5 in Xenopus bottle cells. Using live imaging and quantitative image analysis, we show that bottle cells assume different shapes, with rounding bottle cells constricting earlier in small clusters followed by fusiform bottle cells forming between the clusters. Both medioapical and junctional actomyosin signals increase as surface area decreases, though correlation of apical constriction with medioapical actomyosin localization appears to be stronger. F-actin bundles perpendicular to the apical surface form in constricted cells, which may correspond to microvilli previously observed in the apical membrane. Knockdown of plekhg5 disrupts medioapical and junctional actomyosin activity and apical constriction but does not affect initial F-actin dynamics. Taking the results together, our study reveals distinct cell morphologies, uncovers actomyosin behaviors, and demonstrates the crucial role of a RhoGEF protein in controlling actomyosin dynamics during apical constriction of bottle cells in Xenopus gastrulation.
Collapse
Affiliation(s)
- Austin Baldwin
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Ivan K. Popov
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ray Keller
- Biology Department, University of Virginia, Charlottesville, VA 22903
| | - John Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
6
|
Balaban AE, Nguyen LTS, Parajón E, Robinson DN. Nonmuscle myosin IIB is a driver of cellular reprogramming. Mol Biol Cell 2023; 34:ar71. [PMID: 37074945 PMCID: PMC10295488 DOI: 10.1091/mbc.e21-08-0386] [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: 08/09/2021] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/20/2023] Open
Abstract
Nonmuscle myosin IIB (NMIIB) is considered a primary force generator during cell motility. Yet many cell types, including motile cells, do not necessarily express NMIIB. Given the potential of cell engineering for the next wave of technologies, adding back NMIIB could be a strategy for creating supercells with strategically altered cell morphology and motility. However, we wondered what unforeseen consequences could arise from such an approach. Here, we leveraged pancreatic cancer cells, which do not express NMIIB. We generated a series of cells where we added back NMIIB and strategic mutants that increase the ADP-bound time or alter the phosphorylation control of bipolar filament assembly. We characterized the cellular phenotypes and conducted RNA-seq analysis. The addition of NMIIB and the different mutants all have specific consequences for cell morphology, metabolism, cortical tension, mechanoresponsiveness, and gene expression. Major modes of ATP production are shifted, including alterations in spare respiratory capacity and the dependence on glycolysis or oxidative phosphorylation. Several metabolic and growth pathways undergo significant changes in gene expression. This work demonstrates that NMIIB is highly integrated with many cellular systems and simple cell engineering has a profound impact that extends beyond the primary contractile activity presumably being added to the cells.
Collapse
Affiliation(s)
- Amanda E. Balaban
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ly T. S. Nguyen
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Eleana Parajón
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Douglas N. Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Departments of Pharmacology and Molecular Sciences, Medicine, and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
7
|
Actin crosslinking by α-actinin averts viscous dissipation of myosin force transmission in stress fibers. iScience 2023; 26:106090. [PMID: 36852278 PMCID: PMC9958379 DOI: 10.1016/j.isci.2023.106090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Contractile force generated in actomyosin stress fibers (SFs) is transmitted along SFs to the extracellular matrix (ECM), which contributes to cell migration and sensing of ECM rigidity. In this study, we show that efficient force transmission along SFs relies on actin crosslinking by α-actinin. Upon reduction of α-actinin-mediated crosslinks, the myosin II activity induced flows of actin filaments and myosin II along SFs, leading to a decrease in traction force exertion to ECM. The fluidized SFs maintained their cable integrity probably through enhanced actin polymerization throughout SFs. A computational modeling analysis suggested that lowering the density of actin crosslinks caused viscous slippage of actin filaments in SFs and, thereby, dissipated myosin-generated force transmitting along SFs. As a cellular scale outcome, α-actinin depletion attenuated the ECM-rigidity-dependent difference in cell migration speed, which suggested that α-actinin-modulated SF mechanics is involved in the cellular response to ECM rigidity.
Collapse
|
8
|
Matsuda M, Chu CW, Sokol SY. Lmo7 recruits myosin II heavy chain to regulate actomyosin contractility and apical domain size in Xenopus ectoderm. Development 2022; 149:275389. [PMID: 35451459 PMCID: PMC9188752 DOI: 10.1242/dev.200236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/30/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Apical constriction, or a reduction in size of the apical domain, underlies many morphogenetic events during development. Actomyosin complexes play an essential role in apical constriction; however, the detailed analysis of molecular mechanisms is still pending. Here, we show that Lim domain only protein 7 (Lmo7), a multidomain adaptor at apical junctions, promotes apical constriction in the Xenopus superficial ectoderm, whereas apical domain size increases in Lmo7-depleted cells. Lmo7 is primarily localized at apical junctions and promotes the formation of the dense circumferential actomyosin belt. Strikingly, Lmo7 binds non-muscle myosin II (NMII) and recruits it to apical junctions and the apical cortex. This NMII recruitment is essential for Lmo7-mediated apical constriction. Lmo7 knockdown decreases NMIIA localization at apical junctions and delays neural tube closure in Xenopus embryos. Our findings suggest that Lmo7 serves as a scaffold that regulates actomyosin contractility and apical domain size.
Collapse
Affiliation(s)
- Miho Matsuda
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
9
|
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
|
10
|
Truong Quang BA, Peters R, Cassani DAD, Chugh P, Clark AG, Agnew M, Charras G, Paluch EK. Extent of myosin penetration within the actin cortex regulates cell surface mechanics. Nat Commun 2021; 12:6511. [PMID: 34764258 PMCID: PMC8586027 DOI: 10.1038/s41467-021-26611-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
In animal cells, shape is mostly determined by the actomyosin cortex, a thin cytoskeletal network underlying the plasma membrane. Myosin motors generate tension in the cortex, and tension gradients result in cellular deformations. As such, many cell morphogenesis studies have focused on the mechanisms controlling myosin activity and recruitment to the cortex. Here, we demonstrate using super-resolution microscopy that myosin does not always overlap with actin at the cortex, but remains restricted towards the cytoplasm in cells with low cortex tension. We propose that this restricted penetration results from steric hindrance, as myosin minifilaments are considerably larger than the cortical actin meshsize. We identify myosin activity and actin network architecture as key regulators of myosin penetration into the cortex, and show that increasing myosin penetration increases cortical tension. Our study reveals that the spatial coordination of myosin and actin at the cortex regulates cell surface mechanics, and unveils an important mechanism whereby myosin size controls its action by limiting minifilament penetration into the cortical actin network. More generally, our findings suggest that protein size could regulate function in dense cytoskeletal structures. Cellular deformations are largely driven by contractile forces generated by myosin motors in the submembraneous actin cortex. Here we show that these forces are controlled not simply by cortical myosin levels, but rather by myosins spatial arrangement, specifically the extent of their overlap with cortical actin.
Collapse
Affiliation(s)
- Binh An Truong Quang
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Ruby Peters
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Davide A D Cassani
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Priyamvada Chugh
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Andrew G Clark
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK.,University of Stuttgart, Institute of Cell Biology and Immunology, Allmandring 31, 70569, Stuttgart, Germany
| | - Meghan Agnew
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.,Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK.
| |
Collapse
|
11
|
Eidell KP, Lovy A, Sylvain NR, Scangarello FA, Muendlein HI, Ophir MJ, Nguyen K, Seminario MC, Bunnell SC. LFA-1 and kindlin-3 enable the collaborative transport of SLP-76 microclusters by myosin and dynein motors. J Cell Sci 2021; 134:270974. [PMID: 34279667 DOI: 10.1242/jcs.258602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/13/2021] [Indexed: 01/10/2023] Open
Abstract
Integrin engagement within the immune synapse enhances T cell activation, but our understanding of this process is incomplete. In response to T cell receptor (TCR) ligation, SLP-76 (LCP2), ADAP (FYB1) and SKAP55 (SKAP1) are recruited into microclusters and activate integrins via the effectors talin-1 and kindlin-3 (FERMT3). We postulated that integrins influence the centripetal transport and signaling of SLP-76 microclusters via these linkages. We show that contractile myosin filaments surround and are co-transported with SLP-76 microclusters, and that TCR ligand density governs the centripetal movement of both structures. Centripetal transport requires formin activity, actomyosin contraction, microtubule integrity and dynein motor function. Although immobilized VLA-4 (α4β1 integrin) and LFA-1 (αLβ2 integrin) ligands arrest the centripetal movement of SLP-76 microclusters and myosin filaments, VLA-4 acts distally, while LFA-1 acts in the lamellum. Integrin β2, kindlin-3 and zyxin are required for complete centripetal transport, while integrin β1 and talin-1 are not. CD69 upregulation is similarly dependent on integrin β2, kindlin-3 and zyxin, but not talin-1. These findings highlight the integration of cytoskeletal systems within the immune synapse and reveal extracellular ligand-independent roles for LFA-1 and kindlin-3. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Keith P Eidell
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Alenka Lovy
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nicholas R Sylvain
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Frank A Scangarello
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Hayley I Muendlein
- Graduate Program in Genetics, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Michael J Ophir
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Ken Nguyen
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | | | - Stephen C Bunnell
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
12
|
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: 10] [Impact Index Per Article: 2.5] [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
|
13
|
A De Novo Mutation in MYH9 in a Child With Severe and Prolonged Macrothrombocytopenia. J Pediatr Hematol Oncol 2021; 43:e7-e10. [PMID: 32520844 DOI: 10.1097/mph.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Congenital macrothrombocytopenia is a diverse group of hereditary disorders caused by mutations in the MYH9 gene, which encodes the nonmuscle myosin heavy chain-A, an important motor protein in hemopoietic cells. Thus, the term MYH9-related disease has been proposed, but the clinicopathologic basis of MYH9 mutations has been poorly investigated. Here, we report a sporadic case of Epstein syndrome, an MYH9 disorder, in a 4-year-old Chinese boy who presented with macrothrombocytopenia. He had no family history of thrombocytopenia, hearing loss, or renal failure. A de novo heterozygous MYH9 mutation, c.287C>T; p. (Ser96Leu), was found in this patient. Genotype-phenotype analysis of all reported mutations suggested a domain-specific relationship between the location of the MYH9 mutation and the penetrance of the nonhematologic characteristics of MYH9-related disorders. Our study highlights the importance of suspecting MYH9-related disease even in cases of chronic macrothrombocytopenia without a family history or extrahematologic symptoms.
Collapse
|
14
|
Kamps D, Koch J, Juma VO, Campillo-Funollet E, Graessl M, Banerjee S, Mazel T, Chen X, Wu YW, Portet S, Madzvamuse A, Nalbant P, Dehmelt L. Optogenetic Tuning Reveals Rho Amplification-Dependent Dynamics of a Cell Contraction Signal Network. Cell Rep 2020; 33:108467. [PMID: 33264629 PMCID: PMC7710677 DOI: 10.1016/j.celrep.2020.108467] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/16/2020] [Accepted: 11/10/2020] [Indexed: 01/21/2023] Open
Abstract
Local cell contraction pulses play important roles in tissue and cell morphogenesis. Here, we improve a chemo-optogenetic approach and apply it to investigate the signal network that generates these pulses. We use these measurements to derive and parameterize a system of ordinary differential equations describing temporal signal network dynamics. Bifurcation analysis and numerical simulations predict a strong dependence of oscillatory system dynamics on the concentration of GEF-H1, an Lbc-type RhoGEF, which mediates the positive feedback amplification of Rho activity. This prediction is confirmed experimentally via optogenetic tuning of the effective GEF-H1 concentration in individual living cells. Numerical simulations show that pulse amplitude is most sensitive to external inputs into the myosin component at low GEF-H1 concentrations and that the spatial pulse width is dependent on GEF-H1 diffusion. Our study offers a theoretical framework to explain the emergence of local cell contraction pulses and their modulation by biochemical and mechanical signals.
Collapse
Affiliation(s)
- Dominic Kamps
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, 44227 Dortmund, Germany
| | - Johannes Koch
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Victor O Juma
- Department of Mathematics, University of Sussex, Pevensey III, Brighton BN1 9QH, UK
| | | | - Melanie Graessl
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Soumya Banerjee
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, 44227 Dortmund, Germany
| | - Tomáš Mazel
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, 44227 Dortmund, Germany
| | - Xi Chen
- Chemical Genomics Centre of the Max-Planck Society, 44227 Dortmund, Germany; The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, P.R. China
| | - Yao-Wen Wu
- Chemical Genomics Centre of the Max-Planck Society, 44227 Dortmund, Germany; Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 901 87 Umeå, Sweden
| | - Stephanie Portet
- Department of Mathematics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Anotida Madzvamuse
- Department of Mathematics, University of Sussex, Pevensey III, Brighton BN1 9QH, UK; Department of Mathematics, University of Johannesburg, South Africa; Universita degli Studi di Bari Aldo Moro, Bari, Italy
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Leif Dehmelt
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund University, 44227 Dortmund, Germany.
| |
Collapse
|
15
|
Horváth ÁI, Gyimesi M, Várkuti BH, Képiró M, Szegvári G, Lőrincz I, Hegyi G, Kovács M, Málnási-Csizmadia A. Effect of allosteric inhibition of non-muscle myosin 2 on its intracellular diffusion. Sci Rep 2020; 10:13341. [PMID: 32769996 PMCID: PMC7415145 DOI: 10.1038/s41598-020-69853-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Subcellular dynamics of non-muscle myosin 2 (NM2) is crucial for a broad-array of cellular functions. To unveil mechanisms of NM2 pharmacological control, we determined how the dynamics of NM2 diffusion is affected by NM2′s allosteric inhibitors, i.e. blebbistatin derivatives, as compared to Y-27632 inhibiting ROCK, NM2′s upstream regulator. We found that NM2 diffusion is markedly faster in central fibers than in peripheral stress fibers. Y-27632 accelerated NM2 diffusion in both peripheral and central fibers, whereas in peripheral fibers blebbistatin derivatives slightly accelerated NM2 diffusion at low, but markedly slowed it at high inhibitor concentrations. In contrast, rapid NM2 diffusion in central fibers was unaffected by direct NM2 inhibition. Using our optopharmacological tool, Molecular Tattoo, sub-effective concentrations of a photo-crosslinkable blebbistatin derivative were increased to effective levels in a small, irradiated area of peripheral fibers. These findings suggest that direct allosteric inhibition affects the diffusion profile of NM2 in a markedly different manner compared to the disruption of the upstream control of NM2. The pharmacological action of myosin inhibitors is channeled through autonomous molecular processes and might be affected by the load acting on the NM2 proteins.
Collapse
Affiliation(s)
- Ádám I Horváth
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Máté Gyimesi
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Boglárka H Várkuti
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Miklós Képiró
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Gábor Szegvári
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - István Lőrincz
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - György Hegyi
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Mihály Kovács
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary.
| | - András Málnási-Csizmadia
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary.
| |
Collapse
|
16
|
Bajaj R, Ambaru B, Gupta CM. Deciphering the role of UBA-like domains in intraflagellar distribution and functions of myosin XXI in Leishmania. PLoS One 2020; 15:e0232116. [PMID: 32343719 PMCID: PMC7188243 DOI: 10.1371/journal.pone.0232116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/07/2020] [Indexed: 11/19/2022] Open
Abstract
Myosin XXI (Myo21) is a novel class of myosin present in all kinetoplastid parasites, such as Trypanosoma and Leishmania. This protein in Leishmania promastigotes is predominantly localized to the proximal region of the flagellum, and is involved in the flagellum assembly, cell motility and intracellular vesicle transport. As Myo21 contains two ubiquitin associated (UBA)-like domains (UBLD) in its amino acid sequence, we considered it of interest to analyze the role of these domains in the intracellular distribution and functions of this protein in Leishmania cells. In this context, we created green fluorescent protein (GFP)-conjugates of Myo21 constructs lacking one of the two UBLDs at a time or both the UBLDs as well as GFP-conjugates of only the two UBLDs and Myo21 tail lacking the two UBLDs and separately expressed them in the Leishmania cells. Our results show that unlike Myo21-GFP, Myo21-GFP constructs lacking either one or both the UBLDs failed to concentrate and co-distribute with actin in the proximal region of the flagellum. Nevertheless, the GFP conjugate of only the two UBLDs was found to predominantly localize to the flagellum base. Additionally, the cells that expressed only one or both the UBLDs-deleted Myo21-GFP constructs possessed shorter flagellum and displayed slower motility, compared to Myo21-GFP expressing cells. Further, the intracellular vesicle transport and cell growth were severely impaired in the cells that expressed both the UBLDs deleted Myo21-GFP construct, but in contrast, virtually no effect was observed on the intracellular vesicle transport and growth in the cells that expressed single UBLD deleted mutant proteins. Moreover, the observed slower growth of both the UBLDs-deleted Myo21-GFP expressing cells was primarily due to delayed G2/M phase caused by aberrant nuclear and daughter cell segregation during their cell division process. These results taken together clearly reveal that the presence of UBLDs in Myo21 are essentially required for its predominant localization to the flagellum base, and perhaps also in its involvement in the flagellum assembly and cell division. Possible role of UBLDs in involvement of Myo21 during Leishmania flagellum assembly and cell cycle is discussed.
Collapse
Affiliation(s)
- Rani Bajaj
- Institute of Bioinformatics & Applied Biotechnology, Bengaluru, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Bindu Ambaru
- Institute of Bioinformatics & Applied Biotechnology, Bengaluru, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Chhitar M. Gupta
- Institute of Bioinformatics & Applied Biotechnology, Bengaluru, Karnataka, India
| |
Collapse
|
17
|
Zhang Y, Thery F, Wu NC, Luhmann EK, Dussurget O, Foecke M, Bredow C, Jiménez-Fernández D, Leandro K, Beling A, Knobeloch KP, Impens F, Cossart P, Radoshevich L. The in vivo ISGylome links ISG15 to metabolic pathways and autophagy upon Listeria monocytogenes infection. Nat Commun 2019; 10:5383. [PMID: 31772204 PMCID: PMC6879477 DOI: 10.1038/s41467-019-13393-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
ISG15 is an interferon-stimulated, ubiquitin-like protein, with anti-viral and anti-bacterial activity. Here, we map the endogenous in vivo ISGylome in the liver following Listeria monocytogenes infection by combining murine models of reduced or enhanced ISGylation with quantitative proteomics. Our method identifies 930 ISG15 sites in 434 proteins and also detects changes in the host ubiquitylome. The ISGylated targets are enriched in proteins which alter cellular metabolic processes, including upstream modulators of the catabolic and antibacterial pathway of autophagy. Computational analysis of substrate structures reveals that a number of ISG15 modifications occur at catalytic sites or dimerization interfaces of enzymes. Finally, we demonstrate that animals and cells with enhanced ISGylation have increased basal and infection-induced autophagy through the modification of mTOR, WIPI2, AMBRA1, and RAB7. Taken together, these findings ascribe a role of ISGylation to temporally reprogram organismal metabolism following infection through direct modification of a subset of enzymes in the liver.
Collapse
Affiliation(s)
- Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Fabien Thery
- Center for Medical Biotechnology, VIB, 9000, Gent, Belgium
- Department for Biomolecular Medicine, Gent University, 9000, Gent, Belgium
| | - Nicholas C Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Emma K Luhmann
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Olivier Dussurget
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, 75015, Paris, France
- Inserm, U604, 75015, Paris, France
- National Institute for Agronomic Research (INRA), Unité sous-contrat 2020, 75015, Paris, France
| | - Mariko Foecke
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, 75015, Paris, France
- Inserm, U604, 75015, Paris, France
- National Institute for Agronomic Research (INRA), Unité sous-contrat 2020, 75015, Paris, France
| | - Clara Bredow
- Charité-Universitäts medizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
| | | | - Kevin Leandro
- Center for Medical Biotechnology, VIB, 9000, Gent, Belgium
- Department for Biomolecular Medicine, Gent University, 9000, Gent, Belgium
| | - Antje Beling
- Charité-Universitäts medizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Biochemistry, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Partner Site Berlin, Berlin, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Francis Impens
- Center for Medical Biotechnology, VIB, 9000, Gent, Belgium.
- Department for Biomolecular Medicine, Gent University, 9000, Gent, Belgium.
- VIB Proteomics Core, VIB, 9000, Gent, Belgium.
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, 75015, Paris, France.
- Inserm, U604, 75015, Paris, France.
- National Institute for Agronomic Research (INRA), Unité sous-contrat 2020, 75015, Paris, France.
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
18
|
Brito C, Mesquita FS, Bleck CKE, Sellers JR, Cabanes D, Sousa S. Perfringolysin O-Induced Plasma Membrane Pores Trigger Actomyosin Remodeling and Endoplasmic Reticulum Redistribution. Toxins (Basel) 2019; 11:toxins11070419. [PMID: 31319618 PMCID: PMC6669444 DOI: 10.3390/toxins11070419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
Clostridium perfringens produces an arsenal of toxins that act together to cause severe infections in humans and livestock animals. Perfringolysin O (PFO) is a cholesterol-dependent pore-forming toxin encoded in the chromosome of virtually all C. perfringens strains and acts in synergy with other toxins to determine the outcome of the infection. However, its individual contribution to the disease is poorly understood. Here, we intoxicated human epithelial and endothelial cells with purified PFO to evaluate the host cytoskeletal responses to PFO-induced damage. We found that, at sub-lytic concentrations, PFO induces a profound reorganization of the actomyosin cytoskeleton culminating into the assembly of well-defined cortical actomyosin structures at sites of plasma membrane (PM) remodeling. The assembly of such structures occurs concomitantly with the loss of the PM integrity and requires pore-formation, calcium influx, and myosin II activity. The recovery from the PM damage occurs simultaneously with the disassembly of cortical structures. PFO also targets the endoplasmic reticulum (ER) by inducing its disruption and vacuolation. ER-enriched vacuoles were detected at the cell cortex within the PFO-induced actomyosin structures. These cellular events suggest the targeting of the endothelium integrity at early stages of C. perfringens infection, in which secreted PFO is at sub-lytic concentrations.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel, Salazar, Universidade do Porto, 4099-002 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Christopher K E Bleck
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal.
| |
Collapse
|
19
|
Angiotensin II-mediated MYH9 downregulation causes structural and functional podocyte injury in diabetic kidney disease. Sci Rep 2019; 9:7679. [PMID: 31118506 PMCID: PMC6531474 DOI: 10.1038/s41598-019-44194-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/10/2019] [Indexed: 12/26/2022] Open
Abstract
MYH9, a widely expressed gene encoding nonmuscle myosin heavy chain, is also expressed in podocytes and is associated with glomerular pathophysiology. However, the mechanisms underlying MYH9-related glomerular diseases associated with proteinuria are poorly understood. Therefore, we investigated the role and mechanism of MYH9 in diabetic kidney injury. MYH9 expression was decreased in glomeruli from diabetic patients and animals and in podocytes treated with Ang II in vitro. Ang II treatment and siRNA-mediated MYH9 knockdown in podocytes resulted in actin cytoskeleton reorganization, reduced cell adhesion, actin-associated protein downregulation, and increased albumin permeability. Ang II treatment increased NOX4 expression and ROS generation. The Ang II receptor blocker losartan and the ROS scavenger NAC restored MYH9 expression in Ang II-treated podocytes, attenuated disrupted actin cytoskeleton and decreased albumin permeability. Furthermore, MYH9 overexpression in podocytes restored the effects of Ang II on the actin cytoskeleton and actin-associated proteins. Ang II-mediated TRPC6 activation reduced MYH9 expression. These results suggest that Ang II-mediated MYH9 depletion in diabetic nephropathy may increase filtration barrier permeability by inducing structural and functional podocyte injury through TRPC6-mediated Ca2+ influx by NOX4-mediated ROS generation. These findings reveal a novel MYH9 function in maintaining urinary filtration barrier integrity. MYH9 may be a potential target for treating diabetic nephropathy.
Collapse
|
20
|
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: 90] [Impact Index Per Article: 15.0] [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
|
21
|
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
|
22
|
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
|
23
|
Dash B, Dib-Hajj SD, Waxman SG. Multiple myosin motors interact with sodium/potassium-ATPase alpha 1 subunits. Mol Brain 2018; 11:45. [PMID: 30086768 PMCID: PMC6081954 DOI: 10.1186/s13041-018-0388-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/20/2018] [Indexed: 11/10/2022] Open
Abstract
The alpha1 (α1) subunit of the sodium/potassium ATPase (i.e., Na+/K+-ATPase α1), the prototypical sodium pump, is expressed in each eukaryotic cell. They pump out three sodium ions in exchange for two extracellular potassium ions to establish a cellular electrochemical gradient important for firing of neuronal and cardiac action potentials. We hypothesized that myosin (myo or myh) motor proteins might interact with Na+/K+-ATPase α1 subunits in order for them to play an important role in the transport and trafficking of sodium pump. To this end immunoassays were performed to determine whether class II non-muscle myosins (i.e., NMHC-IIA/myh9, NMHC-IIB/myh10 or NMHC-IIC/myh14), myosin Va (myoVa) and myosin VI (myoVI) would interact with Na+/K+-ATPase α1 subunits. Immunoprecipitation of myh9, myh10, myh14, myoVa and myoVI from rat brain tissues led to the co-immunoprecipitation of Na+/K+-ATPase α1 subunits expressed there. Heterologous expression studies using HEK293 cells indicated that recombinant myh9, myh10, myh14 and myoVI interact with Na+/K+-ATPase α1 subunits expressed in HEK293 cells. Additional results indicated that loss of tail regions in recombinant myh9, myh10, myh14 and myoVI did not affect their interaction with Na+/K+-ATPase α1 subunits. However, recombinant myh9, myh10 and myh14 mutants having reduced or no actin binding ability, as a result of loss of their actin binding sites, displayed greatly reduced or null interaction with Na+/K+-ATPase α1 subunits. These results suggested the involvement of the actin binding site, but not tail regions, of NMHC-IIs in their interaction with Na+/K+-ATPase α1 subunits. Overall these results suggest a role for these diverse myosins in the trafficking and transport of sodium pump in neuronal and non-neuronal tissues.
Collapse
Affiliation(s)
- Bhagirathi Dash
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA.,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA.,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University Schoolof Medicine, New Haven, CT, 06510, USA. .,Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT, 06510, USA. .,Rehabilitation Research center, VA Connecticut Healthcare System, 950 Campbell Avenue, Bldg. 34, West Haven, CT, 06516, USA.
| |
Collapse
|
24
|
Dash B, Han C, Waxman SG, Dib-Hajj SD. Nonmuscle myosin II isoforms interact with sodium channel alpha subunits. Mol Pain 2018; 14:1744806918788638. [PMID: 29956586 PMCID: PMC6052497 DOI: 10.1177/1744806918788638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Sodium channels play pivotal roles in health and diseases due to their ability to control cellular excitability. The pore-forming α-subunits (sodium channel alpha subunits) of the voltage-sensitive channels (i.e., Nav1.1–1.9) and the nonvoltage-dependent channel (i.e., Nax) share a common structural motif and selectivity for sodium ions. We hypothesized that the actin-based nonmuscle myosin II motor proteins, nonmuscle myosin heavy chain-IIA/myh9, and nonmuscle myosin heavy chain-IIB/myh10 might interact with sodium channel alpha subunits to play an important role in their transport, trafficking, and/or function. Immunochemical and electrophysiological assays were conducted using rodent nervous (brain and dorsal root ganglia) tissues and ND7/23 cells coexpressing Nav subunits and recombinant myosins. Immunoprecipitation of myh9 and myh10 from rodent brain tissues led to the coimmunoprecipitation of Nax, Nav1.2, and Nav1.3 subunits, but not Nav1.1 and Nav1.6 subunits, expressed there. Similarly, immunoprecipitation of myh9 and myh10 from rodent dorsal root ganglia tissues led to the coimmunoprecipitation of Nav1.7 and Nav1.8 subunits, but not Nav1.9 subunits, expressed there. The functional implication of one of these interactions was assessed by coexpressing myh10 along with Nav1.8 subunits in ND7/23 cells. Myh10 overexpression led to three-fold increase (P < 0.01) in the current density of Nav1.8 channels expressed in ND7/23 cells. Myh10 coexpression also hyperpolarized voltage-dependent activation and steady-state fast inactivation of Nav1.8 channels. In addition, coexpression of myh10 reduced (P < 0.01) the offset of fast inactivation and the amplitude of the ramp currents of Nav1.8 channels. These results indicate that nonmuscle myosin heavy chain-IIs interact with sodium channel alpha subunits subunits in an isoform-dependent manner and influence their functional properties.
Collapse
Affiliation(s)
- Bhagirathi Dash
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Chongyang Han
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Sulayman D Dib-Hajj
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, USA.,3 Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
25
|
Ozawa M. Nonmuscle myosin IIA is involved in recruitment of apical junction components through activation of α-catenin. Biol Open 2018; 7:bio.031369. [PMID: 29654115 PMCID: PMC5992523 DOI: 10.1242/bio.031369] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
MDCK dog kidney epithelial cells express two isoforms of nonmuscle myosin heavy chain II, IIA and IIB. Using the CRISPR/Cas9 system, we established cells in which the IIA gene was ablated. These cells were then transfected with a vector that expresses GFP-IIA chimeric molecule under the control of a tetracycline-responsible element. In the absence of Dox (doxycyclin), when GFP-IIA is expressed (GFP-IIA+), the cells exhibit epithelial cell morphology, but in the presence of Dox, when expression of GFP-IIA is repressed (GFP-IIA-), the cells lose epithelial morphology and strong cell-cell adhesion. Consistent with these observations, GFP-IIA- cells failed to assemble junction components such as E-cadherin, desmoplakin, and occludin at cell-cell contact sites. Therefore, IIA is required for assembly of junction complexes. MDCK cells with an ablation of the α-catenin gene also exhibited the same phenotype. However, when in GFP-IIA- cells expressed α-catenin lacking the inhibitory region or E-cadherin/α-catenin chimeras, the cells acquired the ability to establish the junction complex. These experiments reveal that IIA acts as an activator of α-catenin in junction assembly.
Collapse
Affiliation(s)
- Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
26
|
Kuragano M, Uyeda TQP, Kamijo K, Murakami Y, Takahashi M. Different contributions of nonmuscle myosin IIA and IIB to the organization of stress fiber subtypes in fibroblasts. Mol Biol Cell 2018; 29:911-922. [PMID: 29467250 PMCID: PMC5896930 DOI: 10.1091/mbc.e17-04-0215] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 12/25/2022] Open
Abstract
We demonstrated that myosin IIA and IIB are essential for the formation of transverse arcs and ventral stress fibers, respectively. Furthermore, we illustrated the roles of both isoforms in lamellar flattening and also raised the possibility that actin filaments in ventral stress fibers are in a stretched conformation.
Collapse
Affiliation(s)
- Masahiro Kuragano
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
| | - Taro Q. P. Uyeda
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Keiju Kamijo
- Department of Anatomy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Yota Murakami
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Masayuki Takahashi
- Graduate School of Chemical Science and Engineering, Hokkaido University, Sapporo, Hokkaido 060-8628, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| |
Collapse
|
27
|
Jiao M, Wu D, Wei Q. Myosin II-interacting guanine nucleotide exchange factor promotes bleb retraction via stimulating cortex reassembly at the bleb membrane. Mol Biol Cell 2018; 29:643-656. [PMID: 29321250 PMCID: PMC6004584 DOI: 10.1091/mbc.e17-10-0579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 01/03/2018] [Indexed: 11/11/2022] Open
Abstract
Blebs are involved in various biological processes such as cell migration, cytokinesis, and apoptosis. While the expansion of blebs is largely an intracellular pressure-driven process, the retraction of blebs is believed to be driven by RhoA activation that leads to the reassembly of the actomyosin cortex at the bleb membrane. However, it is still poorly understood how RhoA is activated at the bleb membrane. Here, we provide evidence demonstrating that myosin II-interacting guanine nucleotide exchange factor (MYOGEF) is implicated in bleb retraction via stimulating RhoA activation and the reassembly of an actomyosin network at the bleb membrane during bleb retraction. Interaction of MYOGEF with ezrin, a well-known regulator of bleb retraction, is required for MYOGEF localization to retracting blebs. Notably, knockout of MYOGEF or ezrin not only disrupts RhoA activation at the bleb membrane, but also interferes with nonmuscle myosin II localization and activation, as well as actin polymerization in retracting blebs. Importantly, MYOGEF knockout slows down bleb retraction. We propose that ezrin interacts with MYOGEF and recruits it to retracting blebs, where MYOGEF activates RhoA and promotes the reassembly of the cortical actomyosin network at the bleb membrane, thus contributing to the regulation of bleb retraction.
Collapse
Affiliation(s)
- Meng Jiao
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| | - Di Wu
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| | - Qize Wei
- Department of Biological Sciences, Fordham University, Bronx, NY 10458
| |
Collapse
|
28
|
Li YR, Yang WX. Myosins as fundamental components during tumorigenesis: diverse and indispensable. Oncotarget 2018; 7:46785-46812. [PMID: 27121062 PMCID: PMC5216836 DOI: 10.18632/oncotarget.8800] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/10/2016] [Indexed: 12/11/2022] Open
Abstract
Myosin is a kind of actin-based motor protein. As the crucial functions of myosin during tumorigenesis have become increasingly apparent, the profile of myosin in the field of cancer research has also been growing. Eighteen distinct classes of myosins have been discovered in the past twenty years and constitute a diverse superfamily. Various myosins share similar structures. They all convert energy from ATP hydrolysis to exert mechanical stress upon interactions with microfilaments. Ongoing research is increasingly suggesting that at least seven kinds of myosins participate in the formation and development of cancer. Myosins play essential roles in cytokinesis failure, chromosomal and centrosomal amplification, multipolar spindle formation and DNA microsatellite instability. These are all prerequisites of tumor formation. Subsequently, myosins activate various processes of tumor invasion and metastasis development including cell migration, adhesion, protrusion formation, loss of cell polarity and suppression of apoptosis. In this review, we summarize the current understanding of the roles of myosins during tumorigenesis and discuss the factors and mechanisms which may regulate myosins in tumor progression. Furthermore, we put forward a completely new concept of “chromomyosin” to demonstrate the pivotal functions of myosins during karyokinesis and how this acts to optimize the functions of the members of the myosin superfamily.
Collapse
Affiliation(s)
- Yan-Ruide Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Graessl M, Koch J, Calderon A, Kamps D, Banerjee S, Mazel T, Schulze N, Jungkurth JK, Patwardhan R, Solouk D, Hampe N, Hoffmann B, Dehmelt L, Nalbant P. An excitable Rho GTPase signaling network generates dynamic subcellular contraction patterns. J Cell Biol 2017; 216:4271-4285. [PMID: 29055010 PMCID: PMC5716289 DOI: 10.1083/jcb.201706052] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/25/2017] [Accepted: 09/08/2017] [Indexed: 12/22/2022] Open
Abstract
Rho GTPase-based signaling networks control cellular dynamics by coordinating protrusions and retractions in space and time. Here, we reveal a signaling network that generates pulses and propagating waves of cell contractions. These dynamic patterns emerge via self-organization from an activator-inhibitor network, in which the small GTPase Rho amplifies its activity by recruiting its activator, the guanine nucleotide exchange factor GEF-H1. Rho also inhibits itself by local recruitment of actomyosin and the associated RhoGAP Myo9b. This network structure enables spontaneous, self-limiting patterns of subcellular contractility that can explore mechanical cues in the extracellular environment. Indeed, actomyosin pulse frequency in cells is altered by matrix elasticity, showing that coupling of contractility pulses to environmental deformations modulates network dynamics. Thus, our study reveals a mechanism that integrates intracellular biochemical and extracellular mechanical signals into subcellular activity patterns to control cellular contractility dynamics.
Collapse
Affiliation(s)
- Melanie Graessl
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Johannes Koch
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Abram Calderon
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Dominic Kamps
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Soumya Banerjee
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Tomáš Mazel
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Nina Schulze
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Jana Kathrin Jungkurth
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Rutuja Patwardhan
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Djamschid Solouk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Nico Hampe
- Institute of Complex Systems, Forschungszentrum Jülich, Jülich, Germany
| | - Bernd Hoffmann
- Institute of Complex Systems, Forschungszentrum Jülich, Jülich, Germany
| | - Leif Dehmelt
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology and Fakultät für Chemie und Chemische Biologie, TU Dortmund University, Dortmund, Germany
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
30
|
Lehtimäki JI, Fenix AM, Kotila TM, Balistreri G, Paavolainen L, Varjosalo M, Burnette DT, Lappalainen P. UNC-45a promotes myosin folding and stress fiber assembly. J Cell Biol 2017; 216:4053-4072. [PMID: 29055011 PMCID: PMC5716280 DOI: 10.1083/jcb.201703107] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/28/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022] Open
Abstract
Contractile actomyosin bundles, stress fibers, are crucial for adhesion, morphogenesis, and mechanosensing in nonmuscle cells. However, the mechanisms by which nonmuscle myosin II (NM-II) is recruited to those structures and assembled into functional bipolar filaments have remained elusive. We report that UNC-45a is a dynamic component of actin stress fibers and functions as a myosin chaperone in vivo. UNC-45a knockout cells display severe defects in stress fiber assembly and consequent abnormalities in cell morphogenesis, polarity, and migration. Experiments combining structured-illumination microscopy, gradient centrifugation, and proteasome inhibition approaches revealed that a large fraction of NM-II and myosin-1c molecules fail to fold in the absence of UNC-45a. The remaining properly folded NM-II molecules display defects in forming functional bipolar filaments. The C-terminal UNC-45/Cro1/She4p domain of UNC-45a is critical for NM-II folding, whereas the N-terminal tetratricopeptide repeat domain contributes to the assembly of functional stress fibers. Thus, UNC-45a promotes generation of contractile actomyosin bundles through synchronized NM-II folding and filament-assembly activities.
Collapse
Affiliation(s)
| | - Aidan M Fenix
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Tommi M Kotila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Giuseppe Balistreri
- Department of Biosciences, Division of General Microbiology, University of Helsinki, Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Coaxum SD, Tiedeken J, Garrett-Mayer E, Myers J, Rosenzweig SA, Neskey DM. The tumor suppressor capability of p53 is dependent on non-muscle myosin IIA function in head and neck cancer. Oncotarget 2017; 8:22991-23007. [PMID: 28160562 PMCID: PMC5410280 DOI: 10.18632/oncotarget.14967] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/10/2017] [Indexed: 11/26/2022] Open
Abstract
Over 300,000 patients develop squamous cell carcinoma of the head and neck (HNSCC) worldwide with 25-30% of patients ultimately dying from their disease. Currently, molecular biomarkers are not used in HNSCC but several genes have been identified including mutant TP53 (mutp53) Our recent work has identified an approach to stratify patients with tumors harboring high or low risk TP53 mutations. Non-muscle Myosin IIA (NMIIA) was recently identified as a tumor suppressor in HNSCC. We now demonstrate that low MYH9 expression is associated with decreased survival in patients with head and neck cancer harboring low-risk mutp53 but not high-risk mutp53. Furthermore, inhibition of NMIIA leads to increased invasion in cells harboring wildtype p53 (wtp53), which was not observed in high-risk mutp53 cells. This increased invasiveness of wtp53 following NMIIA inhibition was associated with reduced p53 target gene expression and was absent in cells expressing mutp53. This reduced expression may be due, in part, to a decrease in nuclear localization of wtp53. These findings suggest that the tumor suppressor capability of wtp53 is dependent upon functional NMIIA and that the invasive phenotype of high-risk mutp53 is independent of NMIIA.
Collapse
Affiliation(s)
- Sonya D Coaxum
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica Tiedeken
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Jeffrey Myers
- Department of Head & Neck Surgery, M.D. Anderson Medical Center, Houston, TX, USA
| | - Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - David M Neskey
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA.,Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
32
|
Hiruma S, Kamasaki T, Otomo K, Nemoto T, Uehara R. Dynamics and function of ERM proteins during cytokinesis in human cells. FEBS Lett 2017; 591:3296-3309. [DOI: 10.1002/1873-3468.12844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Shota Hiruma
- Graduate School of Life Science; Hokkaido University; Japan
| | | | - Kohei Otomo
- Research Institute for Electronic Science; Hokkaido University; Japan
| | - Tomomi Nemoto
- Research Institute for Electronic Science; Hokkaido University; Japan
| | - Ryota Uehara
- Graduate School of Life Science; Hokkaido University; Japan
- Creative Research Institution; Hokkaido University; Japan
| |
Collapse
|
33
|
Roberts B, Haupt A, Tucker A, Grancharova T, Arakaki J, Fuqua MA, Nelson A, Hookway C, Ludmann SA, Mueller IA, Yang R, Horwitz R, Rafelski SM, Gunawardane RN. Systematic gene tagging using CRISPR/Cas9 in human stem cells to illuminate cell organization. Mol Biol Cell 2017; 28:2854-2874. [PMID: 28814507 PMCID: PMC5638588 DOI: 10.1091/mbc.e17-03-0209] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
The generation of a collection of human induced pluripotent stem cell (hiPSC) lines expressing endogenously GFP-tagged proteins using CRISPR/Cas9 methods is described. The methods used and the genomic and cell biological data validating the GFP-tagged hiPSC lines are also presented. We present a CRISPR/Cas9 genome-editing strategy to systematically tag endogenous proteins with fluorescent tags in human induced pluripotent stem cells (hiPSC). To date, we have generated multiple hiPSC lines with monoallelic green fluorescent protein tags labeling 10 proteins representing major cellular structures. The tagged proteins include alpha tubulin, beta actin, desmoplakin, fibrillarin, nuclear lamin B1, nonmuscle myosin heavy chain IIB, paxillin, Sec61 beta, tight junction protein ZO1, and Tom20. Our genome-editing methodology using Cas9/crRNA ribonuclear protein and donor plasmid coelectroporation, followed by fluorescence-based enrichment of edited cells, typically resulted in <0.1–4% homology-directed repair (HDR). Twenty-five percent of clones generated from each edited population were precisely edited. Furthermore, 92% (36/39) of expanded clonal lines displayed robust morphology, genomic stability, expression and localization of the tagged protein to the appropriate subcellular structure, pluripotency-marker expression, and multilineage differentiation. It is our conclusion that, if cell lines are confirmed to harbor an appropriate gene edit, pluripotency, differentiation potential, and genomic stability are typically maintained during the clonal line–generation process. The data described here reveal general trends that emerged from this systematic gene-tagging approach. Final clonal lines corresponding to each of the 10 cellular structures are now available to the research community.
Collapse
Affiliation(s)
| | - Amanda Haupt
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | | - Joy Arakaki
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | | | | | | | | - Ruian Yang
- Allen Institute for Cell Science, Seattle, WA 98109
| | - Rick Horwitz
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | |
Collapse
|
34
|
Shutova MS, Asokan SB, Talwar S, Assoian RK, Bear JE, Svitkina TM. Self-sorting of nonmuscle myosins IIA and IIB polarizes the cytoskeleton and modulates cell motility. J Cell Biol 2017; 216:2877-2889. [PMID: 28701425 PMCID: PMC5584186 DOI: 10.1083/jcb.201705167] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/21/2023] Open
Abstract
Nonmuscle myosin II (NMII) is uniquely responsible for cell contractility and thus defines multiple aspects of cell behavior. To generate contraction, NMII molecules polymerize into bipolar minifilaments. Different NMII paralogs are often coexpressed in cells and can copolymerize, suggesting that they may cooperate to facilitate cell motility. However, whether such cooperation exists and how it may work remain unknown. We show that copolymerization of NMIIA and NMIIB followed by their differential turnover leads to self-sorting of NMIIA and NMIIB along the front-rear axis, thus producing a polarized actin-NMII cytoskeleton. Stress fibers newly formed near the leading edge are enriched in NMIIA, but over time, they become progressively enriched with NMIIB because of faster NMIIA turnover. In combination with retrograde flow, this process results in posterior accumulation of more stable NMIIB-rich stress fibers, thus strengthening cell polarity. By copolymerizing with NMIIB, NMIIA accelerates the intrinsically slow NMIIB dynamics, thus increasing cell motility and traction and enabling chemotaxis.
Collapse
Affiliation(s)
- Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Sreeja B Asokan
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shefali Talwar
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Richard K Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - James E Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | |
Collapse
|
35
|
Jossin Y, Lee M, Klezovitch O, Kon E, Cossard A, Lien WH, Fernandez TE, Cooper JA, Vasioukhin V. Llgl1 Connects Cell Polarity with Cell-Cell Adhesion in Embryonic Neural Stem Cells. Dev Cell 2017; 41:481-495.e5. [PMID: 28552558 DOI: 10.1016/j.devcel.2017.05.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 04/11/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.
Collapse
Affiliation(s)
- Yves Jossin
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Minhui Lee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Olga Klezovitch
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elif Kon
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alexia Cossard
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Tania E Fernandez
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Valera Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA; Department of Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
36
|
Pfisterer SG, Gateva G, Horvath P, Pirhonen J, Salo VT, Karhinen L, Varjosalo M, Ryhänen SJ, Lappalainen P, Ikonen E. Role for formin-like 1-dependent acto-myosin assembly in lipid droplet dynamics and lipid storage. Nat Commun 2017; 8:14858. [PMID: 28361956 PMCID: PMC5380971 DOI: 10.1038/ncomms14858] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 02/01/2017] [Indexed: 11/10/2022] Open
Abstract
Lipid droplets (LDs) are cellular organelles specialized in triacylglycerol (TG) storage undergoing homotypic clustering and fusion. In non-adipocytic cells with numerous LDs this is balanced by poorly understood droplet dissociation mechanisms. We identify non-muscle myosin IIa (NMIIa/MYH-9) and formin-like 1 (FMNL1) in the LD proteome. NMIIa and actin filaments concentrate around LDs, and form transient foci between dissociating LDs. NMIIa depletion results in decreased LD dissociations, enlarged LDs, decreased hydrolysis and increased storage of TGs. FMNL1 is required for actin assembly on LDs in vitro and for NMIIa recruitment to LDs in cells. We propose a novel acto-myosin structure regulating lipid storage: FMNL1-dependent assembly of myosin II-functionalized actin filaments on LDs facilitates their dissociation, thereby affecting LD surface-to-volume ratio and enzyme accessibility to TGs. In neutrophilic leucocytes from MYH9-related disease patients NMIIa inclusions are accompanied by increased lipid storage in droplets, suggesting that NMIIa dysfunction may contribute to lipid imbalance in man.
Collapse
Affiliation(s)
- Simon G. Pfisterer
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Gergana Gateva
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Peter Horvath
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00290, Finland
- Synthetic and Systems Biology Unit, Hungarian Academy of Sciences, BRC, Szeged H-6726, Hungary
| | - Juho Pirhonen
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Veijo T. Salo
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| | - Leena Karhinen
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Samppa J. Ryhänen
- Division of Hematology-Oncology and Stem Cell Transplantation, Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki 00290, Finland
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, Helsinki 00790, Finland
| | - Elina Ikonen
- Department of Anatomy and Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki 00290, Finland
| |
Collapse
|
37
|
Kanzaki H, Movila A, Kayal R, Napimoga MH, Egashira K, Dewhirst F, Sasaki H, Howait M, Al-Dharrab A, Mira A, Han X, Taubman MA, Nichols FC, Kawai T. Phosphoglycerol dihydroceramide, a distinctive ceramide produced by Porphyromonas gingivalis, promotes RANKL-induced osteoclastogenesis by acting on non-muscle myosin II-A (Myh9), an osteoclast cell fusion regulatory factor. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:452-462. [PMID: 28153611 DOI: 10.1016/j.bbalip.2017.01.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 12/29/2016] [Accepted: 01/30/2017] [Indexed: 12/31/2022]
Abstract
Among several virulence factors produced by the periodontal pathogen Porphyromonas gingivalis (Pg), a recently identified novel class of dihydroceramide lipids that contains a long acyl-chain has the potential to play a pathogenic role in periodontitis because of its higher level of tissue penetration compared to other lipid classes produced by Pg. However, the possible impact of Pg ceramides on osteoclastogenesis is largely unknown. In the present study, we report that the phosphoglycerol dihydroceramide (PGDHC) isolated from Pg enhanced osteoclastogenesis in vitro and in vivo. Using RAW264.7 cells, in vitro assays indicated that PGDHC can promote RANKL-induced osteoclastogenesis by generating remarkably larger TRAP+ multinuclear osteoclasts compared to Pg LPS in a TLR2/4-independent manner. According to fluorescent confocal microscopy, co-localization of non-muscle myosin II-A (Myh9) and PGDHC was observed in the cytoplasm of osteoclasts, indicating the membrane-permeability of PGDHC. Loss- and gain-of-function assays using RNAi-based Myh9 gene silencing, as well as overexpression of the Myh9 gene, in RAW264.7 cells showed that interaction of PGDHC with Myh9 enhances RANKL-induced osteoclastogenesis. It was also demonstrated that PGDHC can upregulate the expression of dendritic cell-specific transmembrane protein (DC-STAMP), an important osteoclast fusogen, through signaling that involves Rac1, suggesting that interaction of PGDHC with Myh9 can elicit the cell signal that promotes osteoclast cell fusion. Taken together, our data indicated that PGDHC is a Pg-derived, cell-permeable ceramide that possesses a unique property of promoting osteoclastogenesis via interaction with Myh9 which, in turn, activates a Rac1/DC-STAMP pathway for upregulation of osteoclast cell fusion.
Collapse
Affiliation(s)
- Hiroyuki Kanzaki
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Tsurumi University, School of Dental Medicine, Department of Orthodontics, Kanagawa, Japan.
| | - Alexandru Movila
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA.
| | - Rayyan Kayal
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Marcelo H Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, São Paulo, Brazil.
| | - Kenji Egashira
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA; LION Corporation, Research and Development Headquarters, Odawara, Kanagawa, Japan
| | - Floyd Dewhirst
- The Forsyth Institute, Department of Microbiology, Cambridge, MA, USA; Harvard School of Dental Medicine, Department of Oral Medicine, Infection and Immunity, Boston, MA, USA.
| | - Hajime Sasaki
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Harvard School of Dental Medicine, Department of Oral Medicine, Infection and Immunity, Boston, MA, USA.
| | - Mohammed Howait
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ayman Al-Dharrab
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Abdulghani Mira
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Xiaozhe Han
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Harvard School of Dental Medicine, Department of Oral Medicine, Infection and Immunity, Boston, MA, USA.
| | - Martin A Taubman
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Harvard School of Dental Medicine, Department of Oral Medicine, Infection and Immunity, Boston, MA, USA.
| | - Frank C Nichols
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA.
| | - Toshihisa Kawai
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, USA; Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA; NOVA Southeastern University College of Dental Medicine, Department of Periodontology, Fort Lauderdale, FL, USA.
| |
Collapse
|
38
|
Mesquita FS, Brito C, Mazon Moya MJ, Pinheiro JC, Mostowy S, Cabanes D, Sousa S. Endoplasmic reticulum chaperone Gp96 controls actomyosin dynamics and protects against pore-forming toxins. EMBO Rep 2016; 18:303-318. [PMID: 28039206 DOI: 10.15252/embr.201642833] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 11/09/2022] Open
Abstract
During infection, plasma membrane (PM) blebs protect host cells against bacterial pore-forming toxins (PFTs), but were also proposed to promote pathogen dissemination. However, the details and impact of blebbing regulation during infection remained unclear. Here, we identify the endoplasmic reticulum chaperone Gp96 as a novel regulator of PFT-induced blebbing. Gp96 interacts with non-muscle myosin heavy chain IIA (NMHCIIA) and controls its activity and remodelling, which is required for appropriate coordination of bleb formation and retraction. This mechanism involves NMHCIIA-Gp96 interaction and their recruitment to PM blebs and strongly resembles retraction of uropod-like structures from polarized migrating cells, a process that also promotes NMHCIIA-Gp96 association. Consistently, Gp96 and NMHCIIA not only protect the PM integrity from listeriolysin O (LLO) during infection by Listeria monocytogenes but also affect cytoskeletal organization and cell migration. Finally, we validate the association between Gp96 and NMHCIIA in vivo and show that Gp96 is required to protect hosts from LLO-dependent killing.
Collapse
Affiliation(s)
- Francisco Sarmento Mesquita
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC, Universidade do Porto, Porto, Portugal
| | - Cláudia Brito
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Maria J Mazon Moya
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection (CMBI), Imperial College London, London, UK
| | - Jorge Campos Pinheiro
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Serge Mostowy
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection (CMBI), Imperial College London, London, UK
| | - Didier Cabanes
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal .,Group of Molecular Microbiology, IBMC, Universidade do Porto, Porto, Portugal
| | - Sandra Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal .,Group of Molecular Microbiology, IBMC, Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Hartzell CA, Jankowska KI, Burkhardt JK, Lewis RS. Calcium influx through CRAC channels controls actin organization and dynamics at the immune synapse. eLife 2016; 5. [PMID: 27440222 PMCID: PMC4956410 DOI: 10.7554/elife.14850] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/13/2016] [Indexed: 11/21/2022] Open
Abstract
T cell receptor (TCR) engagement opens Ca2+ release-activated Ca2+ (CRAC) channels and triggers formation of an immune synapse between T cells and antigen-presenting cells. At the synapse, actin reorganizes into a concentric lamellipod and lamella with retrograde actin flow that helps regulate the intensity and duration of TCR signaling. We find that Ca2+ influx is required to drive actin organization and dynamics at the synapse. Calcium acts by promoting actin depolymerization and localizing actin polymerization and the actin nucleation promotion factor WAVE2 to the periphery of the lamellipod while suppressing polymerization elsewhere. Ca2+-dependent retrograde actin flow corrals ER tubule extensions and STIM1/Orai1 complexes to the synapse center, creating a self-organizing process for CRAC channel localization. Our results demonstrate a new role for Ca2+ as a critical regulator of actin organization and dynamics at the synapse, and reveal potential feedback loops through which Ca2+ influx may modulate TCR signaling. DOI:http://dx.doi.org/10.7554/eLife.14850.001 An effective immune response requires the immune system to rapidly recognize and respond to foreign invaders. Immune cells known as T cells recognize infection through a protein on their surface known as the T cell receptor. The T cell receptor binds to foreign proteins displayed on the surface of other cells. This interaction initiates a chain of events, including the opening of calcium channels embedded in the T cell membrane known as CRAC channels, which allows calcium ions to flow into the cell. These events ultimately lead to the activation of the T cell, enabling it to mount an immune response against the foreign invader. As part of the activation process, the T cell spreads over the surface of the cell that is displaying foreign proteins to form an extensive interface known as an immune synapse. The movement of the T cell's internal skeleton (the cytoskeleton) is crucial for the formation and function of the synapse. Actin filaments, a key component of the cytoskeleton, flow from the edge of the synapse toward the center; these rearrangements of the actin cytoskeleton help to transport clusters of T cell receptors to the center of the synapse and enable the T cell receptors to transmit signals that lead to the T cell being activated. It is not entirely clear how the binding of T cell receptors to foreign proteins drives the actin rearrangements, but there is indirect evidence suggesting that calcium ions may be involved. Hartzell et al. have now investigated the interactions between calcium and the actin cytoskeleton at the immune synapse in human T cells. T cells were placed on glass so that they formed immune synapse-like connections with the surface, and actin movements at the synapse were visualized using a specialized type of fluorescence microscopy. When calcium ions were prevented from entering the T cell, the movement of actin stopped almost entirely. Thus, the flow of calcium ions into the T cell through CRAC channels is essential for driving the actin movements that underlie immune synapse development and T cell activation. In further experiments, Hartzell et al. tracked the movements of CRAC channels and actin at the synapse and found that actin filaments create a constricting “corral” that concentrates CRAC channels in the center of the synapse. Thus, by driving cytoskeleton movement, calcium ions also help to organize calcium channels at the immune synapse. Future work will focus on identifying the actin remodeling proteins that enable calcium ions to control this process. DOI:http://dx.doi.org/10.7554/eLife.14850.002
Collapse
Affiliation(s)
- Catherine A Hartzell
- Immunology Program, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Katarzyna I Jankowska
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Richard S Lewis
- Immunology Program, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| |
Collapse
|
40
|
aPKC regulates apical localization of Lgl to restrict elongation of microridges in developing zebrafish epidermis. Nat Commun 2016; 7:11643. [PMID: 27249668 PMCID: PMC4895443 DOI: 10.1038/ncomms11643] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/15/2016] [Indexed: 12/05/2022] Open
Abstract
Epithelial cells exhibit apical membrane protrusions, which confer specific functions to epithelial tissues. Microridges are short actin protrusions that are laterally long and form a maze-like pattern in the apical domain. They are widely found on vertebrate squamous epithelia including epidermis and have functions in mucous retention, membrane storage and abrasion resistance. It is largely unknown how the formation of these laterally long actin projections is regulated. Here, we show that antagonistic interactions between aPKC and Lgl–regulators of apical and basolateral domain identity, respectively,–control the length of microridges in the zebrafish periderm, the outermost layer of the epidermis. aPKC regulates the levels of Lgl and the active form of non-muscle myosinII at the apical cortex to prevent actin polymerization-dependent precocious fusion and elongation of microridges. Our data unravels the functional significance of exclusion of Lgl from the apical domain in epithelial cells. Squamous epithelia present actin-rich microridges on the apical surface, but the mechanism of their formation is not known. Here the authors show that, in zebrafish epidermis, the exclusion of the basolateral regulator Lgl from the apical domain by atypical protein kinase C prevents precocious elongation and fusion of microridges.
Collapse
|
41
|
Uehara R, Kamasaki T, Hiruma S, Poser I, Yoda K, Yajima J, Gerlich DW, Goshima G. Augmin shapes the anaphase spindle for efficient cytokinetic furrow ingression and abscission. Mol Biol Cell 2016; 27:812-27. [PMID: 26764096 PMCID: PMC4803307 DOI: 10.1091/mbc.e15-02-0101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/11/2022] Open
Abstract
During anaphase, distinct populations of microtubules (MTs) form by either centrosome-dependent or augmin-dependent nucleation. It remains largely unknown whether these different MT populations contribute distinct functions to cytokinesis. Here we show that augmin-dependent MTs are required for the progression of both furrow ingression and abscission. Augmin depletion reduced the accumulation of anillin, a contractile ring regulator at the cell equator, yet centrosomal MTs were sufficient to mediate RhoA activation at the furrow. This defect in contractile ring organization, combined with incomplete spindle pole separation during anaphase, led to impaired furrow ingression. During the late stages of cytokinesis, astral MTs formed bundles in the intercellular bridge, but these failed to assemble a focused midbody structure and did not establish tight linkage to the plasma membrane, resulting in furrow regression. Thus augmin-dependent acentrosomal MTs and centrosomal MTs contribute to nonredundant targeting mechanisms of different cytokinesis factors, which are required for the formation of a functional contractile ring and midbody.
Collapse
Affiliation(s)
- Ryota Uehara
- Creative Research Institution, Hokkaido University, Sapporo 001-0021, Japan Department of Life Sciences, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Tomoko Kamasaki
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Shota Hiruma
- Creative Research Institution, Hokkaido University, Sapporo 001-0021, Japan
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Kinya Yoda
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Junichiro Yajima
- Department of Life Sciences, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter Campus, 1030 Vienna, Austria
| | - Gohta Goshima
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| |
Collapse
|
42
|
Newell-Litwa KA, Badoual M, Asmussen H, Patel H, Whitmore L, Horwitz AR. ROCK1 and 2 differentially regulate actomyosin organization to drive cell and synaptic polarity. J Cell Biol 2015; 210:225-42. [PMID: 26169356 PMCID: PMC4508895 DOI: 10.1083/jcb.201504046] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/04/2015] [Indexed: 01/19/2023] Open
Abstract
RhoGTPases organize the actin cytoskeleton to generate diverse polarities, from front-back polarity in migrating cells to dendritic spine morphology in neurons. For example, RhoA through its effector kinase, RhoA kinase (ROCK), activates myosin II to form actomyosin filament bundles and large adhesions that locally inhibit and thereby polarize Rac1-driven actin polymerization to the protrusions of migratory fibroblasts and the head of dendritic spines. We have found that the two ROCK isoforms, ROCK1 and ROCK2, differentially regulate distinct molecular pathways downstream of RhoA, and their coordinated activities drive polarity in both cell migration and synapse formation. In particular, ROCK1 forms the stable actomyosin filament bundles that initiate front-back and dendritic spine polarity. In contrast, ROCK2 regulates contractile force and Rac1 activity at the leading edge of migratory cells and the spine head of neurons; it also specifically regulates cofilin-mediated actin remodeling that underlies the maturation of adhesions and the postsynaptic density of dendritic spines.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Mathilde Badoual
- Laboratoire Imagerie et Modélisation en Neurobiologie et Cancérologie (IMNC), UMR 8165, Centre National de la Recherche Scientifique, University Paris-Sud and University Paris Diderot, 91405 Orsay, France
| | - Hannelore Asmussen
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Heather Patel
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Leanna Whitmore
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
43
|
Almeida MT, Mesquita FS, Cruz R, Osório H, Custódio R, Brito C, Vingadassalom D, Martins M, Leong JM, Holden DW, Cabanes D, Sousa S. Src-dependent tyrosine phosphorylation of non-muscle myosin heavy chain-IIA restricts Listeria monocytogenes cellular infection. J Biol Chem 2015; 290:8383-95. [PMID: 25635050 DOI: 10.1074/jbc.m114.591313] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Bacterial pathogens often interfere with host tyrosine phosphorylation cascades to control host responses and cause infection. Given the role of tyrosine phosphorylation events in different human infections and our previous results showing the activation of the tyrosine kinase Src upon incubation of cells with Listeria monocytogenes, we searched for novel host proteins undergoing tyrosine phosphorylation upon L. monocytogenes infection. We identify the heavy chain of the non-muscle myosin IIA (NMHC-IIA) as being phosphorylated in a specific tyrosine residue in response to L. monocytogenes infection. We characterize this novel post-translational modification event and show that, upon L. monocytogenes infection, Src phosphorylates NMHC-IIA in a previously uncharacterized tyrosine residue (Tyr-158) located in its motor domain near the ATP-binding site. In addition, we found that other intracellular and extracellular bacterial pathogens trigger NMHC-IIA tyrosine phosphorylation. We demonstrate that NMHC-IIA limits intracellular levels of L. monocytogenes, and this is dependent on the phosphorylation of Tyr-158. Our data suggest a novel mechanism of regulation of NMHC-IIA activity relying on the phosphorylation of Tyr-158 by Src.
Collapse
Affiliation(s)
- Maria Teresa Almeida
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal, the Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Francisco S Mesquita
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal, the Medical Research Council, Centre for Molecular Bacteriology and Infection, Imperial College, London, London SW7 2AZ, United Kingdom
| | - Rui Cruz
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal, the Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Hugo Osório
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Institute of Molecular Pathology and Immunology, University of Porto, 4200-465 Porto, Portugal, and
| | - Rafael Custódio
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - Cláudia Brito
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal, the Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Didier Vingadassalom
- the Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Mariana Martins
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal
| | - John M Leong
- the Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - David W Holden
- the Medical Research Council, Centre for Molecular Bacteriology and Infection, Imperial College, London, London SW7 2AZ, United Kingdom
| | - Didier Cabanes
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal,
| | - Sandra Sousa
- From the Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200 Porto, Portugal, the Group of Molecular Microbiology, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150-180 Porto, Portugal,
| |
Collapse
|
44
|
Cdk1-dependent mitotic enrichment of cortical myosin II promotes cell rounding against confinement. Nat Cell Biol 2015; 17:148-59. [DOI: 10.1038/ncb3098] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/17/2014] [Indexed: 12/16/2022]
|
45
|
Galic M, Tsai FC, Collins SR, Matis M, Bandara S, Meyer T. Dynamic recruitment of the curvature-sensitive protein ArhGAP44 to nanoscale membrane deformations limits exploratory filopodia initiation in neurons. eLife 2014; 3:e03116. [PMID: 25498153 PMCID: PMC4381785 DOI: 10.7554/elife.03116] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 11/13/2014] [Indexed: 12/14/2022] Open
Abstract
In the vertebrate central nervous system, exploratory filopodia transiently form on
dendritic branches to sample the neuronal environment and initiate new trans-neuronal
contacts. While much is known about the molecules that control filopodia extension
and subsequent maturation into functional synapses, the mechanisms that regulate
initiation of these dynamic, actin-rich structures have remained elusive. Here, we
find that filopodia initiation is suppressed by recruitment of ArhGAP44 to
actin-patches that seed filopodia. Recruitment is mediated by binding of a membrane
curvature-sensing ArhGAP44 N-BAR domain to plasma membrane sections that were
deformed inward by acto-myosin mediated contractile forces. A GAP domain in ArhGAP44
triggers local Rac-GTP hydrolysis, thus reducing actin polymerization required for
filopodia formation. Additionally, ArhGAP44 expression increases during neuronal
development, concurrent with a decrease in the rate of filopodia formation. Together,
our data reveals a local auto-regulatory mechanism that limits initiation of
filopodia via protein recruitment to nanoscale membrane deformations. DOI:http://dx.doi.org/10.7554/eLife.03116.001 Our brains contain a vast network of many billions of cells that communicate with,
and are connected to, each other. Each brain cell, or neuron, can form connections
with as many as 10,000 other neurons—and signals pass from one neuron to the
next at sites known as synapses. A neuron's surface has numerous finger-like protrusions known as filopodia that are
important for sensing the environment around the cells. Filopodia are highly
changeable and constantly extend and retract as the filaments that support
them—which are made up of a protein called actin—grow and shrink back.
Neurons use their filopodia to explore and seek out other neurons in the brain, and
when they make contact with the right neuron, it leads to the formation of a synapse.
However, how filopodial extensions begin to grow—and what stops a neuron from
forming too many filopodia—is not fully understood. Galic et al. now show that a protein called ArhGAP44 limits the formation of new
filopodia in neurons. The ArhGAP44 protein is recruited to patches of the surface
membrane that have a lot of actin and that curve inwards. ArhGAP44 then locally
inhibits other proteins that are normally required to extend the actin filaments and
drive the growth of filopodia out from the surface of the cell. Galic et al. also show that more ArhGAP44 is produced with age—levels are low
in embryos and high in adults—and this increase in the amount of protein
correlates with a decrease in the number of filopodia formed. When Galic et al.
engineered rat neurons to produce more of the ArhGAP44 protein, fewer filopodia
formed on the surface of the neurons. Decreasing the amount of this protein had the
opposite effect. Moreover, ArhGAP44 was shown to mainly stop new filopodia from
forming and had little effect on existing filopodia. Together, these findings suggest
that ArhGAP44 may help neurons transition from a dynamic exploratory mode to a
mature, more static, state; this is a characteristic of the development of the
nervous system. DOI:http://dx.doi.org/10.7554/eLife.03116.002
Collapse
Affiliation(s)
- Milos Galic
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| | - Feng-Chiao Tsai
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| | - Sean R Collins
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| | - Maja Matis
- Department of Pathology, Stanford University, Stanford, United States
| | - Samuel Bandara
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, United States
| |
Collapse
|
46
|
Rao Y, Hao R, Wang B, Yao TP. A Mec17-Myosin II Effector Axis Coordinates Microtubule Acetylation and Actin Dynamics to Control Primary Cilium Biogenesis. PLoS One 2014; 9:e114087. [PMID: 25494100 PMCID: PMC4262394 DOI: 10.1371/journal.pone.0114087] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/04/2014] [Indexed: 01/31/2023] Open
Abstract
Primary cilia are specialized, acetylated microtubule-based signaling processes. Cilium assembly is activated by cellular quiescence and requires reconfiguration of microtubules, the actin cytoskeleton, and vesicular trafficking machinery. How these components are coordinated to activate ciliogenesis remains unknown. Here we identify the microtubule acetyltransferase Mec-17 and myosin II motors as the key effectors in primary cilium biogenesis. We found that myosin IIB (Myh10) is required for cilium formation; however, myosin IIA (Myh9) suppresses it. Myh10 binds and antagonizes Myh9 to increase actin dynamics, which facilitates the assembly of the pericentrosomal preciliary complex (PPC) that supplies materials for cilium growth. Importantly, Myh10 expression is upregulated by serum-starvation and this induction requires Mec-17, which is itself accumulated upon cellular quiescence. Pharmacological stimulation of microtubule acetylation also induces Myh10 expression and cilium formation. Thus cellular quiescence induces Mec17 to couple the production of acetylated microtubules and Myh10, whose accumulation overcomes the inhibitory role of Myh9 and initiates ciliogenesis.
Collapse
Affiliation(s)
- Yanhua Rao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Rui Hao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Bin Wang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
| | - Tso-Pang Yao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
47
|
Borg M, Bakke O, Progida C. A novel interaction between Rab7b and actomyosin reveals a dual role in intracellular transport and cell migration. J Cell Sci 2014; 127:4927-39. [PMID: 25217632 DOI: 10.1242/jcs.155861] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Rab proteins are small GTPases that regulate transport between the different compartments of the endomembrane system in eukaryotic cells. Here, we show that Rab7b, a Rab that controls the transport between late endosomes and the trans Golgi network, interacts directly with myosin II. We illustrate the functional relevance of this interaction, demonstrating that myosin II mediates the transport of Rab7b endosomes, as Rab7b dynamics are strongly affected after myosin II depletion or inhibition. We also demonstrate that a member of the Rab family regulates actin remodeling and, consequently, influences cell adhesion, polarization and migration. We find the molecular mechanism by which Rab7b influences stress fiber formation - through controlling the activation status of the small GTPase RhoA and therefore influencing myosin light chain phosphorylation. Our findings reveal a newly identified role for Rab proteins outside of their canonical role in intracellular trafficking, identifying Rab7b as a coordinator of cytoskeletal organization.
Collapse
Affiliation(s)
- Marita Borg
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Oddmund Bakke
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| |
Collapse
|
48
|
Lum M, Morona R. Myosin IIA is essential for Shigella flexneri cell-to-cell spread. Pathog Dis 2014; 72:174-87. [PMID: 24989342 DOI: 10.1111/2049-632x.12202] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/29/2014] [Accepted: 06/24/2014] [Indexed: 11/26/2022] Open
Abstract
A key feature of Shigella pathogenesis is the ability to spread from cell-to-cell post-invasion. This is dependent on the bacteria's ability to initiate de novo F-actin tail polymerisation, followed by protrusion formation, uptake of bacteria-containing protrusion and finally, lysis of the double membrane vacuole in the adjacent cell. In epithelial cells, cytoskeletal tension is maintained by the actin-myosin II networks. In this study, the role of myosin II and its specific kinase, myosin light chain kinase (MLCK), during Shigella intercellular spreading was investigated in HeLa cells. Inhibition of MLCK and myosin II, as well as myosin IIA knockdown, significantly reduced Shigella plaque and infectious focus formation. Protrusion formation and intracellular bacterial growth was not affected. Low levels of myosin II were localised to the Shigella F-actin tail. HeLa cells were also infected with Shigella strains defective in cell-to-cell spreading. Unexpectedly loss of myosin IIA labelling was observed in HeLa cells infected with these mutant strains. This phenomenon was not observed with WT Shigella or with the less abundant myosin IIB isoform, suggesting a critical role for myosin IIA.
Collapse
Affiliation(s)
- Mabel Lum
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | | |
Collapse
|
49
|
Ma X, Adelstein RS. A point mutation in Myh10 causes major defects in heart development and body wall closure. ACTA ACUST UNITED AC 2014; 7:257-65. [PMID: 24825879 DOI: 10.1161/circgenetics.113.000455] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The 3 isoforms of nonmuscle myosin (NM) II (NMII-A, NMII-B, and NMII-C) play various roles during mouse embryonic development. Previous work, using knockout and hypomorphic mice, showed that Myh10 encoding myosin heavy chain II-B is critical for cardiac and brain development. Ablating or decreasing NMII-B by 80% results in cardiac (ventricular septal defect, double outlet of the right ventricle) and brain defects but not midline fusion defects. Neither NMII-A nor II-C seems to play roles in early myocardial development. METHODS AND RESULTS We had previously generated point mutant knock-in mice and now report novel findings as a result of expressing motor-deficient NMII-B at wild-type levels. Homozygous mice die at embryonic day 14.5 in cardiac failure, exhibiting abnormalities not seen in NMII-B null and hypomorphic mice: a failure in midline fusion resulting in a cleft palate, ectopia cordis, and a large omphalocele. Fusion of the sternum and endocardial cushions is impaired in the mutant mice associated with a failure in apoptosis of the mesenchymal cells. Failure to disassemble myocyte cell-cell adhesions during cardiac outflow tract development contributes to impaired outflow tract myocardialization and displacement of the aorta to the right ventricle. CONCLUSIONS Expression of motor-impaired NMII-B disrupts normal ventral body wall closure because of a dominant-negative effect. This is not because of the loss of NMII-B function but rather a gain-of-function resulting from prolonged cross-linking of NMII-B to actin filaments, thereby interfering with the dynamics of actomyosin cytoskeletal structure. Furthermore, impaired NMII-B motor activity inhibits outflow tract myocardialization, leading to mislocalization of the aorta.
Collapse
Affiliation(s)
- Xuefei Ma
- From the Laboratory of Molecular Cardiology, Genetics & Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Robert S Adelstein
- From the Laboratory of Molecular Cardiology, Genetics & Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
50
|
Schulze N, Graessl M, Blancke Soares A, Geyer M, Dehmelt L, Nalbant P. FHOD1 regulates stress fiber organization by controlling the dynamics of transverse arcs and dorsal fibers. J Cell Sci 2014; 127:1379-93. [PMID: 24481812 DOI: 10.1242/jcs.134627] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The formin FHOD1 (formin homology 2 domain containing protein 1) can act as a capping and bundling protein in vitro. In cells, active FHOD1 stimulates the formation of ventral stress fibers. However, the cellular mechanisms by which this phenotype is produced and the physiological relevance of FHOD1 function are not currently understood. Here, we first show that FHOD1 controls the formation of two distinct stress fiber precursors differentially. On the one hand, it inhibits dorsal fiber growth, which requires the polymerization of parallel bundles of long actin filaments. On the other hand, it stimulates transverse arcs that are formed by the fusion of short antiparallel actin filaments. This combined action is crucial for the maturation of stress fibers and their spatio-temporal organization, and a lack of FHOD1 function perturbs dynamic cell behavior during cell migration. Furthermore, we show that the GTPase-binding and formin homology 3 domains (GBD and FH3) are responsible for stress fiber association and colocalization with myosin. Surprisingly, a version of FHOD1 that lacks these domains nevertheless retains its full capacity to stimulate arc and ventral stress fiber formation. Based on our findings, we propose a mechanism in which FHOD1 promotes the formation of short actin filaments and transiently associates with transverse arcs, thus providing tight temporal and spatial control of the formation and turnover of transverse arcs into mature ventral stress fibers during dynamic cell behavior.
Collapse
Affiliation(s)
- Nina Schulze
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | | | | | | | | | | |
Collapse
|