51
|
Sadahiro R, Knight B, James F, Hannon E, Charity J, Daniels IR, Burrage J, Knox O, Crawford B, Smart NJ, Mill J. Major surgery induces acute changes in measured DNA methylation associated with immune response pathways. Sci Rep 2020; 10:5743. [PMID: 32238836 PMCID: PMC7113299 DOI: 10.1038/s41598-020-62262-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Surgery is an invasive procedure evoking acute inflammatory and immune responses that can influence risk for postoperative complications including cognitive dysfunction and delirium. Although the specific mechanisms driving these responses have not been well-characterized, they are hypothesized to involve the epigenetic regulation of gene expression. We quantified genome-wide levels of DNA methylation in peripheral blood mononuclear cells (PBMCs) longitudinally collected from a cohort of elderly patients undergoing major surgery, comparing samples collected at baseline to those collected immediately post-operatively and at discharge from hospital. We identified acute changes in measured DNA methylation at sites annotated to immune system genes, paralleling changes in serum-levels of markers including C-reactive protein (CRP) and Interleukin 6 (IL-6) measured in the same individuals. Many of the observed changes in measured DNA methylation were consistent across different types of major surgery, although there was notable heterogeneity between surgery types at certain loci. The acute changes in measured DNA methylation induced by surgery are relatively stable in the post-operative period, generally persisting until discharge from hospital. Our results highlight the dramatic alterations in gene regulation induced by invasive surgery, primarily reflecting upregulation of the immune system in response to trauma, wound healing and anaesthesia.
Collapse
Affiliation(s)
- Ryoichi Sadahiro
- Department of Immune Medicine, National Cancer Center Research Institute, National Cancer Center Japan, Tokyo, Japan. .,University of Exeter Medical School, University of Exeter, Exeter, United Kingdom.
| | - Bridget Knight
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom.,Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Ffion James
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Eilis Hannon
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - John Charity
- Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Ian R Daniels
- Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Joe Burrage
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Olivia Knox
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Bethany Crawford
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Neil J Smart
- Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom.
| |
Collapse
|
52
|
He S, Green Y, Saeidi N, Li X, Fredberg JJ, Ji B, Pismen LM. A theoretical model of collective cell polarization and alignment. JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS 2020; 137:103860. [PMID: 33518805 PMCID: PMC7842695 DOI: 10.1016/j.jmps.2019.103860] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Collective cell polarization and alignment play important roles in tissue morphogenesis, wound healing and cancer metastasis. How cells sense the direction and position in these processes, however, has not been fully understood. Here we construct a theoretical model based on describing cell layer as a nemato-elastic medium, by which the cell polarization, cell alignment and cell active contraction are explicitly expressed as functions of components of the nematic order parameter. To determine the order parameter we derive two sets of governing equations, one for the force equilibrium of the system, and the other for the minimization of the system's free energy including the energy of cell polarization and alignment. By solving these coupled governing equations, we can predict the effects of substrate stiffness, geometries of cell layers, external forces and myosin activity on the direction- and position-dependent cell aspect ratio and cell orientation. Moreover, the axisymmetric problem with cells on a ring-like pattern is solved analytically, and the analytical solution for cell aspect ratio are governed by parameter groups which include the stiffness of the cell and the substrate, the strength of myosin activity and the external forces. Our predictions of the cell aspect ratio and orientation are generally comparable to experimental observations. These results show that the pattern of cell polarization is determined by the anisotropic degree of active contractile stress, and suggest a stress-driven polarization mechanism that enables cells to sense their spatial positions to develop direction- and position-dependent behavior. This, in turn, sheds light on the ways to control pattern formation in tissue engineering for potential biomedical applications.
Collapse
Affiliation(s)
- Shijie He
- Department of Applied Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, China
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yoav Green
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Mechanical Engineering, Ben-Gurion University of the Negev, Beersheva 8410501, Israel
| | - Nima Saeidi
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaojun Li
- Department of Applied Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Jeffrey J. Fredberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Baohua Ji
- Department of Applied Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China
- Corresponding authors. (B. Ji), (L.M. Pismen)
| | - Len M. Pismen
- Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
- Corresponding authors. (B. Ji), (L.M. Pismen)
| |
Collapse
|
53
|
Guzmán-Herrera A, Mao Y. Polarity during tissue repair, a multiscale problem. Curr Opin Cell Biol 2020; 62:31-36. [PMID: 31514044 PMCID: PMC7036748 DOI: 10.1016/j.ceb.2019.07.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
Tissue repair is essential for all organisms, as it protects the integrity and function of tissues and prevents infections and diseases. It takes place at multiple scales, from macroscopic to microscopic levels. Most mechanisms driving tissue repair rely on the correct polarisation of collective cell behaviours, such as migration and proliferation, and polarisation of cytoskeletal and junctional components. Furthermore, re-establishment and maintenance of cell polarity are fundamental for a tissue to be fully repaired and for withstanding mechanical stress during homeostasis and repair. Recent evidence highlights an important role for the interplay between cell polarity and tissue mechanics that are critical in tissue repair.
Collapse
Affiliation(s)
- Alejandra Guzmán-Herrera
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, London, UK
| | - Yanlan Mao
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, London, UK; College of Information and Control, Nanjing University of Information Science and Technology, Nanjing, Jiangsu 210044, People's Republic of China.
| |
Collapse
|
54
|
Taffoni C, Omi S, Huber C, Mailfert S, Fallet M, Rupprecht JF, Ewbank JJ, Pujol N. Microtubule plus-end dynamics link wound repair to the innate immune response. eLife 2020; 9:e45047. [PMID: 31995031 PMCID: PMC7043892 DOI: 10.7554/elife.45047] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/27/2020] [Indexed: 01/20/2023] Open
Abstract
The skin protects animals from infection and physical damage. In Caenorhabditis elegans, wounding the epidermis triggers an immune reaction and a repair response, but it is not clear how these are coordinated. Previous work implicated the microtubule cytoskeleton in the maintenance of epidermal integrity (Chuang et al., 2016). Here, by establishing a simple wounding system, we show that wounding provokes a reorganisation of plasma membrane subdomains. This is followed by recruitment of the microtubule plus end-binding protein EB1/EBP-2 around the wound and actin ring formation, dependent on ARP2/3 branched actin polymerisation. We show that microtubule dynamics are required for the recruitment and closure of the actin ring, and for the trafficking of the key signalling protein SLC6/SNF-12 toward the injury site. Without SNF-12 recruitment, there is an abrogation of the immune response. Our results suggest that microtubule dynamics coordinate the cytoskeletal changes required for wound repair and the concomitant activation of innate immunity.
Collapse
Affiliation(s)
- Clara Taffoni
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | - Shizue Omi
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | - Caroline Huber
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | - Sébastien Mailfert
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | - Mathieu Fallet
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | | | - Jonathan J Ewbank
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| | - Nathalie Pujol
- CIML, Centre d’Immunologie de Marseille-Luminy, Turing Centre for Living SystemsAix Marseille Univ, INSERM, CNRSMarseilleFrance
| |
Collapse
|
55
|
Kobb AB, Rothenberg KE, Fernandez-Gonzalez R. Actin and myosin dynamics are independent during Drosophila embryonic wound repair. Mol Biol Cell 2019; 30:2901-2912. [PMID: 31553671 PMCID: PMC6822589 DOI: 10.1091/mbc.e18-11-0703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Collective cell movements play a central role in embryonic development, tissue repair, and metastatic disease. Cell movements are often coordinated by supracellular networks formed by the cytoskeletal protein actin and the molecular motor nonmuscle myosin II. During wound closure in the embryonic epidermis, the cells around the wound migrate collectively into the damaged region. In Drosophila embryos, mechanical tension stabilizes myosin at the wound edge, facilitating the assembly of a supracellular myosin cable around the wound that coordinates cell migration. Here, we show that actin is also stabilized at the wound edge. However, loss of tension or myosin activity does not affect the dynamics of actin at the wound margin. Conversely, pharmacological stabilization of actin does not affect myosin levels or dynamics around the wound. Together, our data suggest that actin and myosin are independently regulated during embryonic wound closure, thus conferring robustness to the embryonic wound healing response.
Collapse
Affiliation(s)
- Anna B Kobb
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Katheryn E Rothenberg
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
56
|
Wang Z, Zhu X, Zhang R. Characterization and Analysis of Collective Cellular Behaviors in 3D Dextran Hydrogels with Homogenous and Clustered RGD Compositions. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3391. [PMID: 31627307 PMCID: PMC6829236 DOI: 10.3390/ma12203391] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/06/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
Abstract
The interactions between substrate materials and cells usually play an important role in the hydrogel-based 3D cell cultures. However, the hydrogels that are usually used could not be parametrically regulated, especially for quantitatively regulating the spatial distribution of the adhesion sites for cells in 3D. Here, we employed the semisynthetic hydrogel consisting of maleimide-dextran, Arg-Gly-Asp (RGD) peptides, and cell degradable crosslinkers to biochemically characterize the evolutionary behaviors of NIH-3T3 fibroblasts and C2C12 cells in 3D. Moreover, by comparing the cell-adhesive efficacy of 3D dextran hydrogels with four different RGD clustering rates, we explored the underlying regulation law of C2C12 connections and 3T3 aggregations. The results showed that mal-dextran hydrogel could promise cells stable viability and continuous proliferation, and induce more self-organized multicellular structures relative to 2D culture. More importantly, we found that RGD-clustered mal-dextran hydrogel has the advantage of enhancing C2C12 cell elongation and the breadthwise-aggregated connection, and promoting the 3T3 cell aggregating degree compared to that with homogenous RGD. Further, the advantages of RGD clustering hydrogel could be amplified by appropriately reducing RGD concentration. Such RGD-composition controllable mal-dextran hydrogel can function as a regulator of the collective cellular behaviors, which provides useful information for quantitatively designing the tailored hydrogel system and exploiting advanced biomaterials.
Collapse
Affiliation(s)
- Zheng Wang
- College of Mechanical & Electrical Engineering, Hohai University, Changzhou 213022, China.
| | - Xiaolu Zhu
- College of Mechanical & Electrical Engineering, Hohai University, Changzhou 213022, China.
- Jiangsu Key Laboratory of Special Robot Technology, Hohai University, Changzhou 213022, China.
- Changzhou Key Laboratory of Digital Manufacture Technology, Hohai University, Changzhou 213022, China.
| | - Ruiyuan Zhang
- College of Mechanical & Electrical Engineering, Hohai University, Changzhou 213022, China.
| |
Collapse
|
57
|
Tsai CR, Galko MJ. Casein kinase 1α decreases β-catenin levels at adherens junctions to facilitate wound closure in Drosophila larvae. Development 2019; 146:dev175133. [PMID: 31511254 PMCID: PMC6826034 DOI: 10.1242/dev.175133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 09/04/2019] [Indexed: 12/25/2022]
Abstract
Skin wound repair is essential to restore barrier function and prevent infection after tissue damage. Wound-edge epidermal cells migrate as a sheet to close the wound. However, it is still unclear how cell-cell junctions are regulated during wound closure (WC). To study this, we examined adherens junctions during WC in Drosophila larvae. β-Catenin is reduced at the lateral cell-cell junctions of wound-edge epidermal cells in the early healing stages. Destruction complex components, including Ck1α, GSK3β and β-TrCP, suppress β-catenin levels in the larval epidermis. Tissue-specific RNAi targeting these genes also caused severe WC defects. The Ck1αRNAi -induced WC defect is related to adherens junctions because loss of either β-catenin or E-cadherin significantly rescued this WC defect. In contrast, TCFRNAi does not rescue the Ck1αRNAi -induced WC defect, suggesting that Wnt signaling is not related to this defect. Direct overexpression of β-catenin recapitulates most of the features of Ck1α reduction during wounding. Finally, loss of Ck1α also blocked junctional E-cadherin reduction around the wound. Our results suggest that Ck1α and the destruction complex locally regulate cell adhesion to facilitate efficient wound repair.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael J Galko
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Genetics & Epigenetics Graduate Program, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
58
|
Viji Babu PK, Rianna C, Mirastschijski U, Radmacher M. Nano-mechanical mapping of interdependent cell and ECM mechanics by AFM force spectroscopy. Sci Rep 2019; 9:12317. [PMID: 31444369 PMCID: PMC6707266 DOI: 10.1038/s41598-019-48566-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM), as a dynamic component of the tissue, influences cell behavior and plays an important role in cell mechanics and tissue homeostasis. Reciprocally, this three-dimensional scaffold is dynamically, structurally and mechanically modified by cells. In the field of biophysics, the independent role of cell and ECM mechanics has been largely investigated; however, there is a lack of experimental data reporting the interdependent interplay between cell and ECM mechanics, measured simultaneously. Here, using Atomic Force Microscopy (AFM) we have characterized five different decellularized matrices diverse in their topography, ECM composition and stiffness and cultured them with normal and pathological fibroblasts (scar and Dupuytren's). We investigated the change in topography and elasticity of these matrices due to cell seeding, by using AFM peak force imaging and mechanical mapping, respectively. We found normal fibroblasts soften these matrices more than pathological fibroblasts, suggesting that pathological fibroblasts are profoundly influencing tissue stiffening in fibrosis. We detected different ECM composition of decellularized matrices used here influences fibroblast stiffness, thus highlighting that cell mechanics not only depends on ECM stiffness but also on their composition. We used confocal microscopy to assess fibroblasts invasion and found pathological fibroblasts were invading the matrices deeper than normal fibroblasts.
Collapse
Affiliation(s)
| | - Carmela Rianna
- Institute of Biophysics, University of Bremen, Bremen, Germany
| | - Ursula Mirastschijski
- Wound Repair Unit, Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | |
Collapse
|
59
|
Spracklen AJ, Thornton-Kolbe EM, Bonner AN, Florea A, Compton PJ, Fernandez-Gonzalez R, Peifer M. The Crk adapter protein is essential for Drosophila embryogenesis, where it regulates multiple actin-dependent morphogenic events. Mol Biol Cell 2019; 30:2399-2421. [PMID: 31318326 PMCID: PMC6741062 DOI: 10.1091/mbc.e19-05-0302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Small Src homology domain 2 (SH2) and 3 (SH3) adapter proteins regulate cell fate and behavior by mediating interactions between cell surface receptors and downstream signaling effectors in many signal transduction pathways. The CT10 regulator of kinase (Crk) family has tissue-specific roles in phagocytosis, cell migration, and neuronal development and mediates oncogenic signaling in pathways like that of Abelson kinase. However, redundancy among the two mammalian family members and the position of the Drosophila gene on the fourth chromosome precluded assessment of Crk's full role in embryogenesis. We circumvented these limitations with short hairpin RNA and CRISPR technology to assess Crk's function in Drosophila morphogenesis. We found that Crk is essential beginning in the first few hours of development, where it ensures accurate mitosis by regulating orchestrated dynamics of the actin cytoskeleton to keep mitotic spindles in syncytial embryos from colliding. In this role, it positively regulates cortical localization of the actin-related protein 2/3 complex (Arp2/3), its regulator suppressor of cAMP receptor (SCAR), and filamentous actin to actin caps and pseudocleavage furrows. Crk loss leads to the loss of nuclei and formation of multinucleate cells. We also found roles for Crk in embryonic wound healing and in axon patterning in the nervous system, where it localizes to the axons and midline glia. Thus, Crk regulates diverse events in embryogenesis that require orchestrated cytoskeletal dynamics.
Collapse
Affiliation(s)
- Andrew J Spracklen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Emma M Thornton-Kolbe
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Alison N Bonner
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Alexandru Florea
- Institute of Biomaterials and Biomedical Engineering, Ted Rogers Centre for Heart Research, and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Peter J Compton
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, Ted Rogers Centre for Heart Research, and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Mark Peifer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
60
|
Tetley RJ, Staddon MF, Heller D, Hoppe A, Banerjee S, Mao Y. Tissue Fluidity Promotes Epithelial Wound Healing. NATURE PHYSICS 2019; 15:1195-1203. [PMID: 31700525 PMCID: PMC6837871 DOI: 10.1038/s41567-019-0618-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The collective behaviour of cells in epithelial tissues is dependent on their mechanical properties. However, the contribution of tissue mechanics to wound healing in vivo remains poorly understood. Here we investigate the relationship between tissue mechanics and wound healing in live Drosophila wing imaginal discs and show that by tuning epithelial cell junctional tension, we can systematically alter the rate of wound healing. Coincident with the contraction of an actomyosin purse string, we observe cells flowing past each other at the wound edge by intercalating, reminiscent of molecules in a fluid, resulting in seamless wound closure. Using a cell-based physical model, we predict that a reduction in junctional tension fluidises the tissue through an increase in intercalation rate and corresponding reduction in bulk viscosity, in the manner of an unjamming transition. The resultant fluidisation of the tissue accelerates wound healing. Accordingly, when we experimentally reduce tissue tension in wing discs, intercalation rate increases and wounds repair in less time.
Collapse
Affiliation(s)
- Robert J. Tetley
- MRC Laboratory for Molecular Cell Biology, University College
London, Gower Street, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College
London, London, United Kingdom
| | - Michael F. Staddon
- Institute for the Physics of Living Systems, University College
London, London, United Kingdom
- Department of Physics & Astronomy, University College London,
London, United Kingdom
| | - Davide Heller
- Institute of Molecular Life Sciences, University of Zurich,
Winterthurerstrasse 190, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, Quartier Sorge, Batiment
Genopode, Lausanne, 1015, Switzerland
| | - Andreas Hoppe
- Faculty of Science, Engineering and Computing, Kingston University,
Kingston-upon-Thames, United Kingdom
| | - Shiladitya Banerjee
- Institute for the Physics of Living Systems, University College
London, London, United Kingdom
- Department of Physics & Astronomy, University College London,
London, United Kingdom
| | - Yanlan Mao
- MRC Laboratory for Molecular Cell Biology, University College
London, Gower Street, London WC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College
London, London, United Kingdom
- College of Information and Control, Nanjing University of
Information Science and Technology, Nanjing, Jiangsu 210044, China
- Correspondence:
| |
Collapse
|
61
|
Rothenberg KE, Fernandez-Gonzalez R. Forceful closure: cytoskeletal networks in embryonic wound repair. Mol Biol Cell 2019; 30:1353-1358. [PMID: 31145669 PMCID: PMC6724689 DOI: 10.1091/mbc.e18-04-0248] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/26/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022] Open
Abstract
Embryonic tissues heal wounds rapidly and without scarring, in a process conserved across species and driven by collective cell movements. The mechanisms of coordinated cell movement during embryonic wound closure also drive tissue development and cancer metastasis; therefore, embryonic wound repair has received considerable attention as a model of collective cell migration. During wound closure, a supracellular actomyosin cable at the wound edge coordinates cells, while actin-based protrusions contribute to cell crawling and seamless wound healing. Other cytoskeletal networks are reorganized during wound repair: microtubules extend into protrusions and along cell-cell boundaries as cells stretch into damaged regions, septins accumulate at the wound margin, and intermediate filaments become polarized in the cells adjacent to the wound. Thus, diverse cytoskeletal networks work in concert to maintain tissue structure, while also driving and organizing cell movements to promote rapid repair. Understanding the signals that coordinate the dynamics of different cytoskeletal networks, and how adhesions between cells or with the extracellular matrix integrate forces across cells, will be important to elucidate the mechanisms of efficient embryonic wound healing and may have far-reaching implications for developmental and cancer cell biology.
Collapse
Affiliation(s)
- Katheryn E. Rothenberg
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
62
|
Shellard A, Mayor R. Supracellular migration - beyond collective cell migration. J Cell Sci 2019; 132:132/8/jcs226142. [PMID: 30988138 DOI: 10.1242/jcs.226142] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Collective cell migration is a highly complex process in which groups of cells move together. A fundamental question is how cell ensembles can migrate efficiently. In some cases, the group is no more than a collection of individual cells. In others, the group behaves as a supracellular unit, whereby the cell group could be considered as a giant 'supracell', the concept of which was conceived over a century ago. The development of recent tools has provided considerable evidence that cell collectives are highly cooperative, and their migration can better be understood at the tissue level, rather than at the cell level. In this Review, we will define supracellular migration as a type of collective cell migration that operates at a scale higher than the individual cells. We will discuss key concepts of supracellular migration, review recent evidence of collectives exhibiting supracellular features and argue that many seemingly complex collective movements could be better explained by considering the participating cells as supracellular entities.
Collapse
Affiliation(s)
- Adam Shellard
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
63
|
Chen T, Callan-Jones A, Fedorov E, Ravasio A, Brugués A, Ong HT, Toyama Y, Low BC, Trepat X, Shemesh T, Voituriez R, Ladoux B. Large-scale curvature sensing by directional actin flow drives cellular migration mode switching. NATURE PHYSICS 2019; 15:393-402. [PMID: 30984281 PMCID: PMC6456019 DOI: 10.1038/s41567-018-0383-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 11/14/2018] [Indexed: 05/21/2023]
Abstract
Cell migration over heterogeneous substrates during wound healing or morphogenetic processes leads to shape changes driven by different organizations of the actin cytoskeleton and by functional changes including lamellipodial protrusions and contractile actin cables. Cells distinguish between cell-sized positive and negative curvatures in their physical environment by forming protrusions at positive ones and actin cables at negative ones; however, the cellular mechanisms remain unclear. Here, we report that concave edges promote polarized actin structures with actin flow directed towards the cell edge, in contrast to well-documented retrograde flow at convex edges. Anterograde flow and contractility induce a tension anisotropy gradient. A polarized actin network is formed, accompanied by a local polymerization-depolymerization gradient, together with leading-edge contractile actin cables in the front. These cables extend onto non-adherent regions while still maintaining contact with the substrate through focal adhesions. The contraction and dynamic reorganization of this actin structure allows forward movements enabling cell migration over non-adherent regions on the substrate. These versatile functional structures may help cells sense and navigate their environment by adapting to external geometric and mechanical cues.
Collapse
Affiliation(s)
- Tianchi Chen
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Andrew Callan-Jones
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université Paris Diderot, Paris, France
| | | | - Andrea Ravasio
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Agustí Brugués
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Hui Ting Ong
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore
- Temasek Life Sciences Laboratory, Singapore
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), and Universitat de Barcelona, Barcelona, Spain
| | - Tom Shemesh
- Technion – Israel Institute of Technology, Israel
| | - Raphaël Voituriez
- Laboratoire Jean Perrin and Laboratoire de Physique Theorique de la Matiere Condensee, CNRS/Sorbonne Université, Paris, France
- Corresponding authors: Benoît Ladoux; ; Raphaël Voituriez;
| | - Benoît Ladoux
- Mechanobiology Institute, National University of Singapore, Singapore
- Institut Jacques Monod (IJM), CNRS UMR 7592 & Université Paris Diderot, Paris, France
- Corresponding authors: Benoît Ladoux; ; Raphaël Voituriez;
| |
Collapse
|
64
|
Hunter MV, Willoughby PM, Bruce AE, Fernandez-Gonzalez R. Oxidative Stress Orchestrates Cell Polarity to Promote Embryonic Wound Healing. Dev Cell 2018; 47:377-387.e4. [DOI: 10.1016/j.devcel.2018.10.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/02/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023]
|
65
|
Agarwal P, Zaidel-Bar R. Principles of Actomyosin Regulation In Vivo. Trends Cell Biol 2018; 29:150-163. [PMID: 30385150 DOI: 10.1016/j.tcb.2018.09.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Abstract
The actomyosin cytoskeleton is responsible for most force-driven processes in cells and tissues. How it assembles into the necessary structures at the right time and place is an important question. Here, we focus on molecular mechanisms of actomyosin regulation recently elucidated in animal models, and highlight several common principles that emerge. The architecture of the actomyosin network - an important determinant of its function - results from actin polymerization, crosslinking and turnover, localized myosin activation, and contractility-driven self-organization. Spatiotemporal regulation is achieved by tissue-specific expression and subcellular localization of Rho GTPase regulators. Subcellular anchor points of actomyosin structures control the outcome of their contraction, and molecular feedback mechanisms dictate whether they are transient, cyclic, or persistent.
Collapse
Affiliation(s)
- Priti Agarwal
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
66
|
Carvalho L, Patricio P, Ponte S, Heisenberg CP, Almeida L, Nunes AS, Araújo NAM, Jacinto A. Occluding junctions as novel regulators of tissue mechanics during wound repair. J Cell Biol 2018; 217:4267-4283. [PMID: 30228162 PMCID: PMC6279375 DOI: 10.1083/jcb.201804048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/15/2018] [Accepted: 09/05/2018] [Indexed: 01/02/2023] Open
Abstract
Simple epithelial repair is mediated by the contraction of an actomyosin cable and cellular rearrangements at the wound edge. Carvalho et al. show that occluding junctions are required for epithelial repair by regulating these cellular rearrangements and tissue mechanical properties. In epithelial tissues, cells tightly connect to each other through cell–cell junctions, but they also present the remarkable capacity of reorganizing themselves without compromising tissue integrity. Upon injury, simple epithelia efficiently resolve small lesions through the action of actin cytoskeleton contractile structures at the wound edge and cellular rearrangements. However, the underlying mechanisms and how they cooperate are still poorly understood. In this study, we combine live imaging and theoretical modeling to reveal a novel and indispensable role for occluding junctions (OJs) in this process. We demonstrate that OJ loss of function leads to defects in wound-closure dynamics: instead of contracting, wounds dramatically increase their area. OJ mutants exhibit phenotypes in cell shape, cellular rearrangements, and mechanical properties as well as in actin cytoskeleton dynamics at the wound edge. We propose that OJs are essential for wound closure by impacting on epithelial mechanics at the tissue level, which in turn is crucial for correct regulation of the cellular events occurring at the wound edge.
Collapse
Affiliation(s)
- Lara Carvalho
- Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Pedro Patricio
- Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal.,Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal.,Centro de Física Teórica e Computacional, Universidade de Lisboa, Campo Grande, Lisbon, Portugal
| | - Susana Ponte
- Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | | | - Luis Almeida
- Centre National de la Recherche Scientifique/Sorbonne Université/Team Mamba, French Institute for Research in Computer Science and Automation Paris, Laboratoire Jacques-Louis Lions, BC187, Paris, France
| | - André S Nunes
- Centro de Física Teórica e Computacional, Universidade de Lisboa, Campo Grande, Lisbon, Portugal.,Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisbon, Portugal
| | - Nuno A M Araújo
- Centro de Física Teórica e Computacional, Universidade de Lisboa, Campo Grande, Lisbon, Portugal.,Departamento de Física, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisbon, Portugal
| | - Antonio Jacinto
- Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal .,The Discoveries Centre for Regenerative and Precision Medicine, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
67
|
Park J, Ahn HM, Kwon T, Seo S, Park S, Jin YW, Seong KM. Epithelial cell shape change of Drosophila as a biomonitoring model for the dose assessment of environmental radiation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 157:292-299. [PMID: 29627413 DOI: 10.1016/j.ecoenv.2018.03.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/14/2018] [Accepted: 03/30/2018] [Indexed: 06/08/2023]
Abstract
Inevitable exposure to ionizing radiation from natural and human-made sources has been increasing over time. After nuclear disasters, such as the Fukushima accident, the public concerns on health risk of radiation exposure because of radioactive contamination of the environment have increased. However, it is very difficult to assess the biological effects of exposure caused by environmental radiation. A reliable and rapid bioassay to monitor the physiological effects of radiation exposure is therefore needed. Here, we quantitatively analyzed the changes in cell shape in Drosophila epidermis after irradiation as a model for biomonitoring of radiation. Interestingly, the number of irregularly shaped epithelial cells was increased by irradiation in a dose-dependent manner. A dose-response curve constructed with the obtained data suggests that the measurement of the number of irregular shaped cell in the epidermis is useful for the assessment of radiation dose. In addition, a comparison of the variation in the different samples and the data scored by different observers showed that our evaluation for cellular morphology was highly reliable and accurate and would, therefore, have immense practical application. Overall, our study suggests that detection of morphological changes in the epithelial cells is one of the efficient ways to quantify the levels of exposure to radioactive radiation from the environment.
Collapse
Affiliation(s)
- Jina Park
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hyo Min Ahn
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - TaeWoo Kwon
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Songwon Seo
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Sunhoo Park
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea; Departments of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Young Woo Jin
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Ki Moon Seong
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| |
Collapse
|
68
|
Shindo A, Audrey A, Takagishi M, Takahashi M, Wallingford JB, Kinoshita M. Septin-dependent remodeling of cortical microtubule drives cell reshaping during epithelial wound healing. J Cell Sci 2018; 131:jcs212647. [PMID: 29777035 PMCID: PMC6031381 DOI: 10.1242/jcs.212647] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/10/2018] [Indexed: 12/31/2022] Open
Abstract
Wounds in embryos heal rapidly through contraction of the wound edges. Despite well-recognized significance of the actomyosin purse string for wound closure, roles for other cytoskeletal components are largely unknown. Here, we report that the septin cytoskeleton cooperates with actomyosin and microtubules to coordinate circumferential contraction of the wound margin and concentric elongation of wound-proximal cells in Xenopus laevis embryos. Microtubules reoriented radially, forming bundles along lateral cell cortices in elongating wound-proximal cells. Depletion of septin 7 (Sept7) slowed wound closure by attenuating the wound edge contraction and cell elongation. ROCK/Rho-kinase inhibitor-mediated suppression of actomyosin contractility enhanced the Sept7 phenotype, whereas the Sept7 depletion did not affect the accumulation of actomyosin at the wound edge. The cortical microtubule bundles were reduced in wound-proximal cells in Sept7 knockdown (Sept7-KD) embryos, but forced bundling of microtubules mediated by the microtubule-stabilizing protein Map7 did not rescue the Sept7-KD phenotype. Nocodazole-mediated microtubule depolymerization enhanced the Sept7-KD phenotype, suggesting that Sept7 is required for microtubule reorganization during cell elongation. Our findings indicate that septins are required for the rapid wound closure by facilitating cortical microtubule reorganization and the concentric elongation of surrounding cells.
Collapse
Affiliation(s)
- Asako Shindo
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Anastasia Audrey
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| | - Maki Takagishi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Makoto Kinoshita
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| |
Collapse
|
69
|
Yang Y, Levine H. Role of the supracellular actomyosin cable during epithelial wound healing. SOFT MATTER 2018; 14:4866-4873. [PMID: 29850664 DOI: 10.1039/c7sm02521a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The closure of wounds in epithelia is center to many physiological processes in both development and repair of multicellular organisms. Depending on the biochemical and mechanical environment as well as cell type, this collective cellular movement often involves coordinated cell crawling and the purse-string contraction of a supracellular actomyosin ring around the wound. However, it remains uncertain how these two mechanisms cooperatively contribute to the wound healing, and especially the role of the ring is not clear. To decipher this complex process, we develop a particle-based model that includes purse-string contraction, cell crawling and other properties incorporated with monolayers of Madin-Darby canine kidney (MDCK) cells. Our model captures the traction force patterns under several different conditions in experiments. In addition to traction force pointing away from the wound on the leading edge, we observed patterns of traction force pointing towards the wound. We show this inward pointing force pattern is induced by the purse-string contraction. Our model also explains the effects of the purse-string ring and which parameters affect the relative efficiency of these two mechanisms.
Collapse
Affiliation(s)
- Yanjun Yang
- Department of Applied Physics and Center for Theoretical Biological Physics, Rice University, Houston TX, 77251-1892, USA.
| | | |
Collapse
|
70
|
Poon AD, McGill SH, Bhupanapadu Sunkesula SR, Burgess ZS, Dunne PJ, Kang EE, Bittner GD. Ca2+/calmodulin-dependent protein kinase II and Dimethyl Sulfoxide affect the sealing frequencies of transected hippocampal neurons. J Neurosci Res 2018; 96:1208-1222. [PMID: 29577375 DOI: 10.1002/jnr.24232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 11/11/2022]
Abstract
Traumatic injury often results in axonal severance, initiating obligatory Wallerian degeneration of distal segments, whereas proximal segments often survive. Calcium ion (Ca2+ ) influx at severed proximal axonal ends activates pathways that can induce apoptosis. However, this same Ca2+ -influx also activates multiple parallel pathways that seal the plasmalemma by inducing accumulation and fusion of vesicles at the lesion site that reduce Ca2+ -influx and enhance survival. We examined whether various inhibitors of Ca2+ /calmodulin-dependent protein kinases (CaMKs), and/or dimethyl sulfoxide (DMSO), a common solvent for biologically active substances, affected the ability of a hippocampal-derived neuronal cell line (B104 cells) to seal membrane damage following axotomy. Axolemmal sealing frequencies were assessed at different transection distances from the axon hillock and at various times after Ca2+ -influx (PC times) by observing whether transected cells took-up fluorescent dyes. Inhibition of CaMKII by tatCN21 and KN-93, but not inhibition of CaMKI and CaMKIV by STO-609, affected axonal sealing frequencies. That is, CaMKII is a component of previously reported parallel pathways that induce membrane sealing, whereas CaMKI and CaMKIV are not involved. The effects of these CaMKII inhibitors on plasmalemmal sealing depended on their mechanism of inhibition, transection distance, and PC time. DMSO at low concentrations (90 µM-28 mM or 0.00064%-0.2% v/v) significantly increased membrane-sealing frequencies at most PC times and transection distances, possibly by permeabilizing the plasmalemma to Ca2+ . Inhibition of CaMKII, DMSO, PC time, and the transection distance significantly affect plasmalemmal sealing that is critical to somal survival in traumatic lesions.
Collapse
Affiliation(s)
- Andrew D Poon
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| | - Sarah H McGill
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| | | | - Zachary S Burgess
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| | - Patrick J Dunne
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| | - Edward E Kang
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| | - George D Bittner
- Department of Neuroscience, The University of Texas at Austin, 2415 Speedway Austin, TX, 78712, USA
| |
Collapse
|
71
|
Coburn L, Lopez H, Schouwenaar IM, Yap AS, Lobaskin V, Gomez GA. Role of contact inhibition of locomotion and junctional mechanics in epithelial collective responses to injury. Phys Biol 2018; 15:024001. [PMID: 29091048 DOI: 10.1088/1478-3975/aa976b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epithelial tissues form physically integrated barriers against the external environment protecting organs from infection and invasion. Within each tissue, epithelial cells respond to different challenges that can potentially compromise tissue integrity. In particular, cells collectively respond to injuries by reorganizing their cell-cell junctions and migrating directionally towards the sites of damage. Notwithstanding, the mechanisms that drive collective responses in epithelial aggregates remain poorly understood. In this work, we develop a minimal mechanistic model that is able to capture the essential features of epithelial collective responses to injuries. We show that a model that integrates the mechanics of cells at the cell-cell and cell-substrate interfaces as well as contact inhibition of locomotion (CIL) correctly predicts two key properties of epithelial response to injury as: (1) local relaxation of the tissue and (2) collective reorganization involving the extension of cryptic lamellipodia that extend, on average, up to 3 cell diameters from the site of injury and morphometric changes in the basal regions. Our model also suggests that active responses (like the actomyosin purse string and softening of cell-cell junctions) are needed to drive morphometric changes in the apical region. Therefore, our results highlight the importance of the crosstalk between junctional biomechanics, cell substrate adhesion, and CIL, as well as active responses, in guiding the collective rearrangements that are required to preserve the epithelial barrier in response to injury.
Collapse
Affiliation(s)
- Luke Coburn
- Institute of Complex Systems and Mathematical Biology, University of Aberdeen, United Kingdom. Authors to whom any correspondence should be addressed
| | | | | | | | | | | |
Collapse
|
72
|
Harn HIC, Ogawa R, Hsu CK, Hughes MW, Tang MJ, Chuong CM. The tension biology of wound healing. Exp Dermatol 2017; 28:464-471. [PMID: 29105155 DOI: 10.1111/exd.13460] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2017] [Indexed: 12/30/2022]
Abstract
Following skin wounding, the healing outcome can be: regeneration, repair with normal scar tissue, repair with hypertrophic scar tissue or the formation of keloids. The role of chemical factors in wound healing has been extensively explored, and while there is evidence suggesting the role of mechanical forces, its influence is much less well defined. Here, we provide a brief review on the recent progress of the role of mechanical force in skin wound healing by comparing laboratory mice, African spiny mice, fetal wound healing and adult scar keloid formation. A comparison across different species may provide insight into key regulators. Interestingly, some findings suggest tension can induce an immune response, and this provides a new link between mechanical and chemical forces. Clinically, manipulating skin tension has been demonstrated to be effective for scar prevention and treatment, but not for tissue regeneration. Utilising this knowledge, specialists may modulate regulatory factors and develop therapeutic strategies to reduce scar formation and promote regeneration.
Collapse
Affiliation(s)
- Hans I-Chen Harn
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Chao-Kai Hsu
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.,Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Michael W Hughes
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Jer Tang
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Ming Chuong
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
73
|
|
74
|
Shannon EK, Stevens A, Edrington W, Zhao Y, Jayasinghe AK, Page-McCaw A, Hutson MS. Multiple Mechanisms Drive Calcium Signal Dynamics around Laser-Induced Epithelial Wounds. Biophys J 2017; 113:1623-1635. [PMID: 28978452 PMCID: PMC5627067 DOI: 10.1016/j.bpj.2017.07.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Epithelial wound healing is an evolutionarily conserved process that requires coordination across a field of cells. Studies in many organisms have shown that cytosolic calcium levels rise within a field of cells around the wound and spread to neighboring cells, within seconds of wounding. Although calcium is a known potent second messenger and master regulator of wound-healing programs, it is unknown what initiates the rise of cytosolic calcium across the wound field. Here we use laser ablation, a commonly used technique for the precision removal of cells or subcellular components, as a tool to investigate mechanisms of calcium entry upon wounding. Despite its precise ablation capabilities, we find that this technique damages cells outside the primary wound via a laser-induced cavitation bubble, which forms and collapses within microseconds of ablation. This cavitation bubble damages the plasma membranes of cells it contacts, tens of microns away from the wound, allowing direct calcium entry from extracellular fluid into damaged cells. Approximately 45 s after this rapid influx of calcium, we observe a second influx of calcium that spreads to neighboring cells beyond the footprint of cavitation. The occurrence of this second, delayed calcium expansion event is predicted by wound size, indicating that a separate mechanism of calcium entry exists, corresponding to cell loss at the primary wound. Our research demonstrates that the damage profile of laser ablation is more similar to a crush injury than the precision removal of individual cells. The generation of membrane microtears upon ablation is consistent with studies in the field of optoporation, which investigate ablation-induced cellular permeability. We conclude that multiple types of damage, including microtears and cell loss, result in multiple mechanisms of calcium influx around epithelial wounds.
Collapse
Affiliation(s)
- Erica K Shannon
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Aaron Stevens
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Westin Edrington
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Yunhua Zhao
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Aroshan K Jayasinghe
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee.
| | - M Shane Hutson
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee; Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
75
|
Chen T, Zhao H, Gao L, Song L, Yang F, Du J. Hypotonicity promotes epithelial gap closure by lamellipodial protrusion. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 148:60-64. [PMID: 28962936 DOI: 10.1016/j.pbiomolbio.2017.09.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 01/24/2023]
Abstract
The closure of gaps within epithelia is an essential part of many physiological and pathological processes, such as embryonic development, organ remodeling and wound healing. Emerging evidence proved that the physical microenvironment plays important roles in cell behaviors. However, the effect of osmolarity of extracellular medium on gap closure is least understood. Using a gap closure model of epithelial cells, we found that hypotonic condition significantly facilitated the process of gap closure. Moreover, instead of actomyosin ring, enhanced migration leading by lamellipodia primarily contributed to the rapid gap closure in hypotonic condition. These findings provide insights for understanding the physiology of epithelial gap closure.
Collapse
Affiliation(s)
- T Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, People's Republic of China; Department of Orthopaedics, Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi 530027, People's Republic of China
| | - H Zhao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, People's Republic of China
| | - L Gao
- College of Life Science, Hebei Normal University, Heibei 050024, People's Republic of China
| | - L Song
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, People's Republic of China; College of Life Science, Hebei Normal University, Heibei 050024, People's Republic of China
| | - F Yang
- Department of Orthopaedics, Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi 530027, People's Republic of China
| | - J Du
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, People's Republic of China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, People's Republic of China.
| |
Collapse
|
76
|
Mochizuki T, Luo YJ, Tsai HF, Hagiwara A, Masai I. Cell division and cadherin-mediated adhesion regulate lens epithelial cell movement in zebrafish. Development 2017; 144:708-719. [PMID: 28196805 DOI: 10.1242/dev.138909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/04/2017] [Indexed: 11/20/2022]
Abstract
In vertebrates, lens epithelial cells cover the anterior half of the lens fiber core. During development, lens epithelial cells proliferate, move posteriorly and differentiate into lens fiber cells after passing through the equator. To elucidate the mechanisms underlying lens epithelial cell movement, we conducted time-lapse imaging of zebrafish lens epithelium. Lens epithelial cells do not intermingle but maintain their relative positions during development. Cell division induces epithelial rearrangement, which subsequently promotes cell movement towards the equator. These data suggest that cell division is the major driving force for cell movement. In zebrafish, E-cadherin is expressed in lens epithelium, whereas N-cadherin is required for lens fiber growth. E-cadherin reduced lens epithelial cell movement, whereas N-cadherin enhanced it. Laser ablation experiments revealed that lens epithelium is governed by pulling tension, which is modulated by these cadherins. Thus, cell division and cadherin-mediated adhesion regulate lens epithelial cell movement via modulation of epithelial tension.
Collapse
Affiliation(s)
- Toshiaki Mochizuki
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Yi-Jyun Luo
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Hsieh-Fu Tsai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Akane Hagiwara
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
77
|
Quantitative modelling of epithelial morphogenesis: integrating cell mechanics and molecular dynamics. Semin Cell Dev Biol 2017; 67:153-160. [DOI: 10.1016/j.semcdb.2016.07.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/28/2016] [Accepted: 07/27/2016] [Indexed: 12/22/2022]
|
78
|
Hunter MV, Fernandez-Gonzalez R. Coordinating cell movements in vivo: junctional and cytoskeletal dynamics lead the way. Curr Opin Cell Biol 2017. [PMID: 28622576 DOI: 10.1016/j.ceb.2017.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Collective cell movements drive embryonic development and tissue repair, and can cause disease. However, the mechanisms that coordinate the migration of groups of cells in vivo are unclear. Cells generate, transmit and sense mechanical forces to align their movements. Therefore, the machinery used by cells to generate force (cytoskeleton) and to transmit and sense mechanical signals (cell-cell adhesion) is critical for collective movement. Here, we review the components and organization of the cytoskeletal and cell-cell adhesive machineries, and how they are organized to promote collective cell movements in living animals. We discuss the signals that orchestrate molecular rearrangements necessary for coordinated cell motility, and we provide specific examples of movements both in the plane of the tissue (wound healing) and perpendicular to that plane (apical constriction).
Collapse
Affiliation(s)
- Miranda V Hunter
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
79
|
Kobb AB, Zulueta-Coarasa T, Fernandez-Gonzalez R. Tension regulates myosin dynamics during Drosophila embryonic wound repair. J Cell Sci 2017; 130:689-696. [DOI: 10.1242/jcs.196139] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT
Embryos repair epithelial wounds rapidly in a process driven by collective cell movements. Upon wounding, actin and the molecular motor non-muscle myosin II are redistributed in the cells adjacent to the wound, forming a supracellular purse string around the lesion. Purse string contraction coordinates cell movements and drives rapid wound closure. By using fluorescence recovery after photobleaching in Drosophila embryos, we found that myosin turns over as the purse string contracts. Myosin turnover at the purse string was slower than in other actomyosin networks that had a lower level of contractility. Mathematical modelling suggested that myosin assembly and disassembly rates were both reduced by tension at the wound edge. We used laser ablation to show that tension at the purse string increased as wound closure progressed, and that the increase in tension was associated with reduced myosin turnover. Reducing purse string tension by laser-mediated severing resulted in increased turnover and loss of myosin. Finally, myosin motor activity was necessary for its stabilization around the wound and for rapid wound closure. Our results indicate that mechanical forces regulate myosin dynamics during embryonic wound repair.
Collapse
Affiliation(s)
- Anna B. Kobb
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada M5G 1M1
| | - Teresa Zulueta-Coarasa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada M5G 1M1
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada M5G 1M1
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada M5S 3G5
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| |
Collapse
|
80
|
Zulueta-Coarasa T, Fernandez-Gonzalez R. Tension (re)builds: Biophysical mechanisms of embryonic wound repair. Mech Dev 2016; 144:43-52. [PMID: 27989746 DOI: 10.1016/j.mod.2016.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 12/24/2022]
Abstract
Embryonic tissues display an outstanding ability to rapidly repair wounds. Epithelia, in particular, serve as protective layers that line internal organs and form the skin. Thus, maintenance of epithelial integrity is of utmost importance for animal survival, particularly at embryonic stages, when an immune system has not yet fully developed. Rapid embryonic repair of epithelial tissues is conserved across species, and involves the collective migration of the cells around the wound. The migratory cell behaviours associated with wound repair require the generation and transmission of mechanical forces, not only for the cells to move, but also to coordinate their movements. Here, we review the forces involved in embryonic wound repair. We discuss how different force-generating structures are assembled at the molecular level, and the mechanisms that maintain the balance between force-generating structures as wounds close. Finally, we describe the mechanisms that cells use to coordinate the generation of mechanical forces around the wound. Collective cell movements and their misregulation have been associated with defective tissue repair, developmental abnormalities and cancer metastasis. Thus, we propose that understanding the role of mechanical forces during embryonic wound closure will be crucial to develop therapeutic interventions that promote or prevent collective cell movements under pathological conditions.
Collapse
Affiliation(s)
- Teresa Zulueta-Coarasa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
81
|
Mamiya A, Kitano H, Kokubun S, Hidai C. Activation peptide of coagulation factor IX regulates endothelial permeability. Transl Res 2016; 177:70-84.e5. [PMID: 27392935 DOI: 10.1016/j.trsl.2016.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/11/2016] [Accepted: 06/15/2016] [Indexed: 01/07/2023]
Abstract
Endothelial hyperpermeability is involved in several critical illnesses, and its regulatory mechanisms have been intensively investigated. It was recently reported that the activation peptide of coagulation factor IX enhances cell matrix and intercellular adhesion. The aim of this study was to investigate the role of activation peptide of coagulation factor IX in intercellular adhesion of endothelial cells and evaluate its effects on endothelial permeability. In the presence of activation peptide, cells spread with lamellipodium-like broad protrusions multidirectionally, increasing the area of adhesion to matrix by 16% within 30 minutes. In intercellular adhesion, treatment with activation peptide induced overlapping of adjacent cell edges and remodeling of intercellular adhesion sites, with colocalization of the adherens junction proteins VE-cadherin and β-catenin and a marker protein of the lateral border recycling compartment, PECAM. Activation peptide decreased gaps between cells by 66% in cultured endothelial cells and suppressed increased endothelial cell monolayer permeability induced by interleukin-1β in a dose-dependent manner. Treatment with activation peptide decreased eNOS protein expression and altered its subcellular distribution, decreasing intracellular cGMP. An analogue of cGMP suppressed the effects of activation peptide on cell spreading. In addition, the effect of activation peptide on hyperpermeability was investigated in mice injected with lipopolysaccharide. Intravenous injection of lipopolysaccharide increased lung weight by 28%, and treatment with activation peptide significantly suppressed the increase in lung weight to 5%. Our results indicate that activation peptide of factor IX regulates endothelial intercellular adhesion and thus could be used in the treatment of vascular hyperpermeability.
Collapse
Affiliation(s)
- Atsushi Mamiya
- Department of Otorhinolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Hisataka Kitano
- Department of Otorhinolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Shinichiro Kokubun
- Department of Biomedical Science, Nihon University School of Medicine, Tokyo, Japan
| | - Chiaki Hidai
- Department of Biomedical Science, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
82
|
Koch L, Feicht S, Sun R, Sen A, Krahn MP. Domain-specific functions of Stardust in Drosophila embryonic development. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160776. [PMID: 28018665 PMCID: PMC5180163 DOI: 10.1098/rsos.160776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/19/2016] [Indexed: 06/06/2023]
Abstract
In Drosophila, the adaptor protein Stardust is essential for the stabilization of the polarity determinant Crumbs in various epithelial tissues, including the embryonic epidermis, the follicular epithelium and photoreceptor cells of the compound eye. In turn, Stardust recruits another adaptor protein, PATJ, to the subapical region to support adherens junction formation and morphogenetic events. Moreover, Stardust binds to Lin-7, which is dispensable in epithelial cells but functions in postsynaptic vesicle fusion. Finally, Stardust has been reported to bind directly to PAR-6, thereby linking the Crumbs-Stardust-PATJ complex to the PAR-6/aPKC complex. PAR-6 and aPKC are also capable of directly binding Bazooka (the Drosophila homologue of PAR-3) to form the PAR/aPKC complex, which is essential for apical-basal polarity and cell-cell contact formation in most epithelia. However, little is known about the physiological relevance of these interactions in the embryonic epidermis of Drosophila in vivo. Thus, we performed a structure-function analysis of the annotated domains with GFP-tagged Stardust and evaluated the localization and function of the mutant proteins in epithelial cells of the embryonic epidermis. The data presented here confirm a crucial role of the PDZ domain in binding Crumbs and recruiting the protein to the subapical region. However, the isolated PDZ domain is not capable of being recruited to the cortex, and the SH3 domain is essential to support the binding to Crumbs. Notably, the conserved N-terminal regions (ECR1 and ECR2) are not crucial for epithelial polarity. Finally, the GUK domain plays an important role for the protein's function, which is not directly linked to Crumbs stabilization, and the L27N domain is essential for epithelial polarization independently of recruiting PATJ.
Collapse
Affiliation(s)
| | | | | | | | - Michael P. Krahn
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany
| |
Collapse
|
83
|
Charafeddine RA, Nosanchuk JD, Sharp DJ. Targeting Microtubules for Wound Repair. Adv Wound Care (New Rochelle) 2016; 5:444-454. [PMID: 27785378 DOI: 10.1089/wound.2015.0658] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/08/2015] [Indexed: 12/16/2022] Open
Abstract
Significance: Fast and seamless healing is essential for both deep and chronic wounds to restore the skin and protect the body from harmful pathogens. Thus, finding new targets that can both expedite and enhance the repair process without altering the upstream signaling milieu and causing serious side effects can improve the way we treat wounds. Since cell migration is key during the different stages of wound healing, it presents an ideal process and intracellular structural machineries to target. Recent Advances and Critical Issues: The microtubule (MT) cytoskeleton is rising as an important structural and functional regulator of wound healing. MTs have been reported to play different roles in the migration of the various cell types involved in wound healing. Specific microtubule regulatory proteins (MRPs) can be targeted to alter a section or subtype of the MT cytoskeleton and boost or hinder cell motility. However, inhibiting intracellular components can be challenging in vivo, especially using unstable molecules, such as small interfering RNA. Nanoparticles can be used to protect these unstable molecules and topically deliver them to the wound. Utilizing this approach, we recently showed that fidgetin-like 2, an uncharacterized MRP, can be targeted to enhance cell migration and wound healing. Future Directions: To harness the full potential of the current MRP therapeutic targets, studies should test them with different delivery platforms, dosages, and skin models. Screening for new MT effectors that boost cell migration in vivo would also help find new targets for skin repair.
Collapse
Affiliation(s)
- Rabab A. Charafeddine
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| | - Joshua D. Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - David J. Sharp
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
84
|
Coburn L, Lopez H, Caldwell BJ, Moussa E, Yap C, Priya R, Noppe A, Roberts AP, Lobaskin V, Yap AS, Neufeld Z, Gomez GA. Contact inhibition of locomotion and mechanical cross-talk between cell-cell and cell-substrate adhesion determine the pattern of junctional tension in epithelial cell aggregates. Mol Biol Cell 2016; 27:3436-3448. [PMID: 27605701 PMCID: PMC5221537 DOI: 10.1091/mbc.e16-04-0226] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
A computational approach is used to analyze the biomechanics of epithelial cells based on their capacity to adhere to one another and to the substrate and exhibit contact inhibition of locomotion. This approach reproduces emergent properties of epithelial cell aggregates and makes predictions for experimental validation. We used a computational approach to analyze the biomechanics of epithelial cell aggregates—islands, stripes, or entire monolayers—that combines both vertex and contact-inhibition-of-locomotion models to include cell–cell and cell–substrate adhesion. Examination of the distribution of cell protrusions (adhesion to the substrate) in the model predicted high-order profiles of cell organization that agree with those previously seen experimentally. Cells acquired an asymmetric distribution of basal protrusions, traction forces, and apical aspect ratios that decreased when moving from the edge to the island center. Our in silico analysis also showed that tension on cell–cell junctions and apical stress is not homogeneous across the island. Instead, these parameters are higher at the island center and scale up with island size, which we confirmed experimentally using laser ablation assays and immunofluorescence. Without formally being a three-dimensional model, our approach has the minimal elements necessary to reproduce the distribution of cellular forces and mechanical cross-talk, as well as the distribution of principal stress in cells within epithelial cell aggregates. By making experimentally testable predictions, our approach can aid in mechanical analysis of epithelial tissues, especially when local changes in cell–cell and/or cell–substrate adhesion drive collective cell behavior.
Collapse
Affiliation(s)
- Luke Coburn
- School of Physics and Complex and Adaptive Systems Laboratory, University College Dublin, Belfield, Dublin 4, Ireland .,Institute of Complex Systems and Mathematical Biology, University of Aberdeen, Aberdeen AB24 3FX, United Kingdom
| | - Hender Lopez
- School of Physics and Complex and Adaptive Systems Laboratory, University College Dublin, Belfield, Dublin 4, Ireland.,Center for BioNano Interactions, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Benjamin J Caldwell
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Elliott Moussa
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Chloe Yap
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Rashmi Priya
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Adrian Noppe
- School of Mathematics and Physics, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Anthony P Roberts
- School of Mathematics and Physics, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Vladimir Lobaskin
- School of Physics and Complex and Adaptive Systems Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | - Alpha S Yap
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Zoltan Neufeld
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia.,School of Mathematics and Physics, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Guillermo A Gomez
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, University of Queensland, St. Lucia, Brisbane 4072, Australia
| |
Collapse
|
85
|
Matsubayashi Y, Millard TH. Analysis of the Molecular Mechanisms of Reepithelialization in Drosophila Embryos. Adv Wound Care (New Rochelle) 2016; 5:243-250. [PMID: 27274434 PMCID: PMC4876545 DOI: 10.1089/wound.2014.0549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Significance: The epidermis provides the main barrier function of skin, and therefore its repair following wounding is an essential component of wound healing. Repair of the epidermis, also known as reepithelialization, occurs by collective migration of epithelial cells from around the wound edge across the wound until the advancing edges meet and fuse. Therapeutic manipulation of this process could potentially be used to accelerate wound healing. Recent Advances: It is difficult to analyze the cellular and molecular mechanisms of reepithelialization in human tissue, so a variety of model organisms have been used to improve our understanding of the process. One model system that has been especially useful is the embryo of the fruit fly Drosophila, which provides a simple, accessible model of the epidermis and can be manipulated genetically, allowing detailed analysis of reepithelialization at the molecular level. This review will highlight the key insights that have been gained from studying reepithelialization in Drosophila embryos. Critical Issues: Slow reepithelialization increases the risk of wounds becoming infected and ulcerous; therefore, the development of therapies to accelerate or enhance the process would be a great clinical advance. Improving our understanding of the molecular mechanisms that underlie reepithelialization will help in the development of such therapies. Future Directions: Research in Drosophila embryos has identified a variety of genes and proteins involved in triggering and driving reepithelialization, many of which are conserved in humans. These novel reepithelialization proteins are potential therapeutic targets and therefore findings obtained in Drosophila may ultimately lead to significant clinical advances.
Collapse
Affiliation(s)
- Yutaka Matsubayashi
- Faculty of Life Sciences, The Healing Foundation Centre, University of Manchester, Manchester, United Kingdom
| | - Tom H. Millard
- Faculty of Life Sciences, The Healing Foundation Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
86
|
Begnaud S, Chen T, Delacour D, Mège RM, Ladoux B. Mechanics of epithelial tissues during gap closure. Curr Opin Cell Biol 2016; 42:52-62. [PMID: 27131272 DOI: 10.1016/j.ceb.2016.04.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 12/15/2022]
Abstract
The closure of gaps is crucial to maintaining epithelium integrity during developmental and repair processes such as dorsal closure and wound healing. Depending on biochemical as well as physical properties of the microenvironment, gap closure occurs through assembly of multicellular actin-based contractile cables and/or protrusive activity of cells lining the gap. This review discusses the relative contributions of 'purse-string' and cell crawling mechanisms regulated by cell-substrate and cell-cell interactions, cellular mechanics and physical constraints from the environment.
Collapse
Affiliation(s)
- Simon Begnaud
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France
| | - Tianchi Chen
- Mechanobiology Institute (MBI), National University of Singapore, Singapore
| | - Delphine Delacour
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France
| | - René-Marc Mège
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France.
| | - Benoît Ladoux
- Institut Jacques Monod (IJM), CNRS UMR 7592 & University Paris Diderot, Paris, France; Mechanobiology Institute (MBI), National University of Singapore, Singapore.
| |
Collapse
|
87
|
Cellular forces and matrix assembly coordinate fibrous tissue repair. Nat Commun 2016; 7:11036. [PMID: 26980715 PMCID: PMC4799373 DOI: 10.1038/ncomms11036] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 02/12/2016] [Indexed: 01/17/2023] Open
Abstract
Planar in vitro models have been invaluable tools to identify the mechanical basis of wound closure. Although these models may recapitulate closure dynamics of epithelial cell sheets, they fail to capture how a wounded fibrous tissue rebuilds its 3D architecture. Here we develop a 3D biomimetic model for soft tissue repair and demonstrate that fibroblasts ensconced in a collagen matrix rapidly close microsurgically induced defects within 24 h. Traction force microscopy and time-lapse imaging reveal that closure of gaps begins with contractility-mediated whole-tissue deformations. Subsequently, tangentially migrating fibroblasts along the wound edge tow and assemble a progressively thickening fibronectin template inside the gap that provide the substrate for cells to complete closure. Unlike previously reported mechanisms based on lamellipodial protrusions and purse-string contraction, our data reveal a mode of stromal closure in which coordination of tissue-scale deformations, matrix assembly and cell migration act together to restore 3D tissue architecture. Planar in vitro models for wound closure stress the role of lamellipodial protrusions and purse-string contraction. Here the authors develop a 3D biomimetic model for tissue repair and show a mode of stromal closure that relies on whole tissue deformations, cell migration and matrix deposition.
Collapse
|
88
|
Wu X, Yang L, Zheng Z, Li Z, Shi J, Li Y, Han S, Gao J, Tang C, Su L, Hu D. Src promotes cutaneous wound healing by regulating MMP-2 through the ERK pathway. Int J Mol Med 2016; 37:639-48. [PMID: 26821191 PMCID: PMC4771097 DOI: 10.3892/ijmm.2016.2472] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 01/22/2016] [Indexed: 12/21/2022] Open
Abstract
Wound healing is a highly orchestrated, multistep process, and delayed wound healing is a significant symptomatic clinical problem. Keratinocyte migration and re-epithelialization play the most important roles in wound healing, as they determine the rate of wound healing. In our previous study, we found that Src, one of the oldest proto-oncogenes encoding a membrane-associated, non-receptor protein tyrosine kinase, promotes keratinocyte migration. We therefore hypothesized that Src promotes wound healing through enhanced keratinocyte migration. In order to test this hypothesis, vectors for overexpressing Src and small interfering RNAs (siRNAs) for silencing of Src were used in the present study. We found that the overexpression of Src accelerated keratinocyte migration in vitro and promoted wound healing in vivo without exerting a marked effect on cell proliferation. The extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathways play important roles in Src-accelerated keratinocyte migration. Further experiments demonstrated that Src induced the protein expression of matrix metallopro-teinase-2 (MMP-2) and decreased the protein expression of E-cadherin. We suggest that ERK signaling is involved in the Src-mediated regulation of MMP-2 expression. The present study provided evidence that Src promotes keratinocyte migration and cutaneous wound healing, in which the regulation of MMP-2 through the ERK pathway plays an important role, and thus we also demonstrated a potential therapeutic role for Src in cutaneous wound healing.
Collapse
Affiliation(s)
- Xue Wu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Longlong Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhenzhen Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shichao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jianxin Gao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Chaowu Tang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Linlin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
89
|
Abstract
Epithelial repair in the Drosophila embryo is achieved through 2 dynamic cytoskeletal machineries: a contractile actomyosin cable and actin-based cellular protrusions. Rho family small GTPases (Rho, Rac, and Cdc42) are cytoskeletal regulators that control both of these wound repair mechanisms. Cdc42 is necessary for cellular protrusions and, when absent, wounds are slow to repair and never completely close. Rac proteins accumulate at specific regions in the wound leading edge cells and Rac-deficient embryos exhibit slower repair kinetics. Mutants for both Rho1 and its effector Rok impair the ability of wounds to close by disrupting the leading-edge actin cable. Our studies highlight the importance of these proteins in wound repair and identify a downstream effector of Rho1 signaling in this process.
Collapse
Affiliation(s)
- Jeffrey M Verboon
- a Division of Basic Sciences; Fred Hutchinson Cancer Research Center ; Seattle , WA USA
| | | |
Collapse
|
90
|
Broaders KE, Cerchiari AE, Gartner ZJ. Coupling between apical tension and basal adhesion allow epithelia to collectively sense and respond to substrate topography over long distances. Integr Biol (Camb) 2015; 7:1611-21. [PMID: 26507156 PMCID: PMC4666816 DOI: 10.1039/c5ib00240k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Epithelial sheets fold into complex topographies that contribute to their function in vivo. Cells can sense and respond to substrate topography in their immediate vicinity by modulating their interfacial mechanics, but the extent to which these mechanical properties contribute to their ability to sense substrate topography across length scales larger than a single cell has not been explored in detail. To study the relationship between the interfacial mechanics of single cells and their collective behavior as tissues, we grew cell-sheets on substrates engraved with surface features spanning macroscopic length-scales. We found that many epithelial cell-types sense and respond to substrate topography, even when it is locally nearly planar. Cells clear or detach from regions of local negative curvature, but not from regions with positive or no curvature. We investigated this phenomenon using a finite element model where substrate topography is coupled to epithelial response through a balance of tissue contractility and adhesive forces. The model correctly predicts the focal sites of cell-clearing and epithelial detachment. Furthermore, the model predicts that local tissue response to substrate curvature is a function of the surrounding topography of the substrate across long distances. Analysis of cell-cell and cell-substrate contact angles suggests a relationship between these single-cell interfacial properties, epithelial interfacial properties, and collective epithelial response to substrate topography. Finally, we show that contact angles change upon activation of oncogenes or inhibition of cell-contractility, and that these changes correlate with collective epithelial response. Our results demonstrate that in mechanically integrated epithelial sheets, cell contractility can be transmitted through multiple cells and focused by substrate topography to affect a behavioral response at distant sites.
Collapse
Affiliation(s)
- Kyle E. Broaders
- Department of Chemistry, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Alec E. Cerchiari
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
91
|
Flores-Benitez D, Knust E. Crumbs is an essential regulator of cytoskeletal dynamics and cell-cell adhesion during dorsal closure in Drosophila. eLife 2015; 4. [PMID: 26544546 PMCID: PMC4718732 DOI: 10.7554/elife.07398] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved Crumbs protein is required for epithelial polarity and morphogenesis. Here we identify a novel role of Crumbs as a negative regulator of actomyosin dynamics during dorsal closure in the Drosophila embryo. Embryos carrying a mutation in the FERM (protein 4.1/ezrin/radixin/moesin) domain-binding motif of Crumbs die due to an overactive actomyosin network associated with disrupted adherens junctions. This phenotype is restricted to the amnioserosa and does not affect other embryonic epithelia. This function of Crumbs requires DMoesin, the Rho1-GTPase, class-I p21-activated kinases and the Arp2/3 complex. Data presented here point to a critical role of Crumbs in regulating actomyosin dynamics, cell junctions and morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.001 A layer of epithelial cells covers the body surface of animals. Epithelial cells have a property known as polarity; this means that they have two different poles, one of which is in contact with the environment. Midway through embryonic development, the Drosophila embryo is covered by two kinds of epithelial sheets; the epidermis on the front, the belly and the sides of the embryo, and the amnioserosa on the back. In the second half of embryonic development, the amnioserosa is brought into the embryo in a process called dorsal closure, while the epidermis expands around the back of the embryo to encompass it. One of the major activities driving dorsal closure is the contraction of amnioserosa cells. This contraction depends on the highly dynamic activity of the protein network that helps give cells their shape, known as the actomyosin cytoskeleton. One major question in the field is how changes in the actomyosin cytoskeleton are controlled as tissues take shape (a process known as “morphogenesis”) and how the integrity of epithelial tissues is maintained during these processes. A key regulator of epidermal and amnioserosa polarity is an evolutionarily conserved protein called Crumbs. The epithelial tissues of mutant embryos that do not produce Crumbs lose polarity and integrity, and the embryos fail to develop properly. Flores-Benitez and Knust have now studied the role of Crumbs in the morphogenesis of the amnioserosa during dorsal closure. This revealed that fly embryos that produce a mutant Crumbs protein that cannot interact with a protein called Moesin (which links the cell membrane and the actomyosin cytoskeleton) are unable to complete dorsal closure. Detailed analyses showed that this failure of dorsal closure is due to the over-activity of the actomyosin cytoskeleton in the amnioserosa. This results in increased and uncoordinated contractions of the cells, and is accompanied by defects in cell-cell adhesion that ultimately cause the amnioserosa to lose integrity. Flores-Benitez and Knust’s genetic analyses further showed that several different signalling systems participate in this process. Flores-Benitez and Knust’s results reveal an unexpected role of Crumbs in coordinating polarity, actomyosin activity and cell-cell adhesion. Further work is now needed to understand the molecular mechanisms and interactions that enable Crumbs to coordinate these processes; in particular, to unravel how Crumbs influences the periodic contractions that drive changes in cell shape. It will also be important to investigate whether Crumbs is involved in similar mechanisms that operate in other developmental events in which actomyosin oscillations have been linked to tissue morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.07398.002
Collapse
Affiliation(s)
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
92
|
Plasma membrane and cytoskeleton dynamics during single-cell wound healing. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [DOI: 10.1016/j.bbamcr.2015.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
93
|
Hunter MV, Lee DM, Harris TJC, Fernandez-Gonzalez R. Polarized E-cadherin endocytosis directs actomyosin remodeling during embryonic wound repair. J Cell Biol 2015; 210:801-16. [PMID: 26304727 PMCID: PMC4555830 DOI: 10.1083/jcb.201501076] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 07/14/2015] [Indexed: 12/26/2022] Open
Abstract
Clathrin, dynamin, and ARF6 accumulate around wounds in Drosophila embryos in a calcium- and actomyosin-dependent manner and drive polarized E-cadherin endocytosis, which is necessary for actomyosin remodeling during wound repair. Embryonic epithelia have a remarkable ability to rapidly repair wounds. A supracellular actomyosin cable around the wound coordinates cellular movements and promotes wound closure. Actomyosin cable formation is accompanied by junctional rearrangements at the wound margin. We used in vivo time-lapse quantitative microscopy to show that clathrin, dynamin, and the ADP-ribosylation factor 6, three components of the endocytic machinery, accumulate around wounds in Drosophila melanogaster embryos in a process that requires calcium signaling and actomyosin contractility. Blocking endocytosis with pharmacological or genetic approaches disrupted wound repair. The defect in wound closure was accompanied by impaired removal of E-cadherin from the wound edge and defective actomyosin cable assembly. E-cadherin overexpression also resulted in reduced actin accumulation around wounds and slower wound closure. Reducing E-cadherin levels in embryos in which endocytosis was blocked rescued actin localization to the wound margin. Our results demonstrate a central role for endocytosis in wound healing and indicate that polarized E-cadherin endocytosis is necessary for actomyosin remodeling during embryonic wound repair.
Collapse
Affiliation(s)
- Miranda V Hunter
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Donghoon M Lee
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Tony J C Harris
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
94
|
Matsubayashi Y, Coulson-Gilmer C, Millard TH. Endocytosis-dependent coordination of multiple actin regulators is required for wound healing. J Cell Biol 2015. [PMID: 26216900 PMCID: PMC4523608 DOI: 10.1083/jcb.201411037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ability to heal wounds efficiently is essential for life. After wounding of an epithelium, the cells bordering the wound form dynamic actin protrusions and/or a contractile actomyosin cable, and these actin structures drive wound closure. Despite their importance in wound healing, the molecular mechanisms that regulate the assembly of these actin structures at wound edges are not well understood. In this paper, using Drosophila melanogaster embryos, we demonstrate that Diaphanous, SCAR, and WASp play distinct but overlapping roles in regulating actin assembly during wound healing. Moreover, we show that endocytosis is essential for wound edge actin assembly and wound closure. We identify adherens junctions (AJs) as a key target of endocytosis during wound healing and propose that endocytic remodeling of AJs is required to form "signaling centers" along the wound edge that control actin assembly. We conclude that coordination of actin assembly, AJ remodeling, and membrane traffic is required for the construction of a motile leading edge during wound healing.
Collapse
Affiliation(s)
- Yutaka Matsubayashi
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, England, UK
| | - Camilla Coulson-Gilmer
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, England, UK
| | - Tom H Millard
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, England, UK
| |
Collapse
|
95
|
Abstract
Tissue fusion eliminates physical voids in a tissue to form a continuous structure and is central to many processes in development and repair. Fusion events in vivo, particularly in embryonic development, often involve the purse-string contraction of a pluricellular actomyosin cable at the free edge. However, in vitro, adhesion of the cells to their substrate favors a closure mechanism mediated by lamellipodial protrusions, which has prevented a systematic study of the purse-string mechanism. Here, we show that monolayers can cover well-controlled mesoscopic nonadherent areas much larger than a cell size by purse-string closure and that active epithelial fluctuations are required for this process. We have formulated a simple stochastic model that includes purse-string contractility, tissue fluctuations, and effective friction to qualitatively and quantitatively account for the dynamics of closure. Our data suggest that, in vivo, tissue fusion adapts to the local environment by coordinating lamellipodial protrusions and purse-string contractions.
Collapse
|
96
|
Ravasio A, Cheddadi I, Chen T, Pereira T, Ong HT, Bertocchi C, Brugues A, Jacinto A, Kabla AJ, Toyama Y, Trepat X, Gov N, Neves de Almeida L, Ladoux B. Gap geometry dictates epithelial closure efficiency. Nat Commun 2015; 6:7683. [PMID: 26158873 PMCID: PMC4510701 DOI: 10.1038/ncomms8683] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/02/2015] [Indexed: 12/12/2022] Open
Abstract
Closure of wounds and gaps in tissues is fundamental for the correct development and physiology of multicellular organisms and, when misregulated, may lead to inflammation and tumorigenesis. To re-establish tissue integrity, epithelial cells exhibit coordinated motion into the void by active crawling on the substrate and by constricting a supracellular actomyosin cable. Coexistence of these two mechanisms strongly depends on the environment. However, the nature of their coupling remains elusive because of the complexity of the overall process. Here we demonstrate that epithelial gap geometry in both in vitro and in vivo regulates these collective mechanisms. In addition, the mechanical coupling between actomyosin cable contraction and cell crawling acts as a large-scale regulator to control the dynamics of gap closure. Finally, our computational modelling clarifies the respective roles of the two mechanisms during this process, providing a robust and universal mechanism to explain how epithelial tissues restore their integrity.
Collapse
Affiliation(s)
- Andrea Ravasio
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Ibrahim Cheddadi
- Sorbonne Universités, UPMC University Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, F-75252 Paris, France
| | - Tianchi Chen
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Telmo Pereira
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, Rua Camara Pestana, 6, Lisbon, 1150-082 Lisbon, Portugal
| | - Hui Ting Ong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Cristina Bertocchi
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Agusti Brugues
- ICREA at Institute for Bioengineering of Catalonia and Universitat de Barcelona, 08028 Barcelona, Spain
| | - Antonio Jacinto
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, Rua Camara Pestana, 6, Lisbon, 1150-082 Lisbon, Portugal
| | - Alexandre J Kabla
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, UK
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.,Department of Biological Sciences National University of Singapore, Singapore 117543, Singapore.,Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | - Xavier Trepat
- ICREA at Institute for Bioengineering of Catalonia and Universitat de Barcelona, 08028 Barcelona, Spain
| | - Nir Gov
- Weizmann Institute of Science, Rehovot 76100, Israel
| | - Luís Neves de Almeida
- Sorbonne Universités, UPMC University Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, F-75252 Paris, France.,INRIA-Paris-Rocquencourt, MAMBA Team, 78153 Le Chesnay, Domaine de Voluceau BP105, France
| | - Benoit Ladoux
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore.,Institut Jacques Monod (IJM), CNRS UMR 7592 and Université Paris Diderot, 75013 Paris, France
| |
Collapse
|
97
|
Hashimoto H, Robin FB, Sherrard KM, Munro EM. Sequential contraction and exchange of apical junctions drives zippering and neural tube closure in a simple chordate. Dev Cell 2015; 32:241-55. [PMID: 25625209 DOI: 10.1016/j.devcel.2014.12.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 11/07/2014] [Accepted: 12/21/2014] [Indexed: 10/24/2022]
Abstract
Unidirectional zippering is a key step in neural tube closure that remains poorly understood. Here, we combine experimental and computational approaches to identify the mechanism for zippering in a basal chordate, Ciona intestinalis. We show that myosin II is activated sequentially from posterior to anterior along the neural/epidermal (Ne/Epi) boundary just ahead of the advancing zipper. This promotes rapid shortening of Ne/Epi junctions, driving the zipper forward and drawing the neural folds together. Cell contact rearrangements (Ne/Epi + Ne/Epi → Ne/Ne + Epi/Epi) just behind the zipper lower tissue resistance to zipper progression by allowing transiently stretched cells to detach and relax toward isodiametric shapes. Computer simulations show that measured differences in junction tension, timing of primary contractions, and delay before cell detachment are sufficient to explain the speed and direction of zipper progression and highlight key advantages of a sequential contraction mechanism for robust efficient zippering.
Collapse
Affiliation(s)
- Hidehiko Hashimoto
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Francois B Robin
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Kristin M Sherrard
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Edwin M Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
98
|
Vlisidou I, Wood W. Drosophila blood cells and their role in immune responses. FEBS J 2015; 282:1368-82. [PMID: 25688716 DOI: 10.1111/febs.13235] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/02/2015] [Accepted: 02/12/2015] [Indexed: 12/17/2022]
Abstract
Drosophila melanogaster has been extensively used to study the humoral arm of innate immunity because of the developmental and functional parallels with mammalian innate immunity. However, the fly cellular response to infection is far less understood. Investigative work on Drosophila haemocytes, the immunosurveillance cells of the insect, has revealed that they fulfil roles similar to mammalian monocytes and macrophages. They respond to wound signals and orchestrate the coagulation response. In addition, they phagocytose and encapsulate invading pathogens, and clear up apoptotic bodies controlling inflammation. This review briefly describes the Drosophila haematopoietic system and discusses what is currently known about the contribution of haemocytes to the immune response upon infection and wounding, during all stages of development.
Collapse
Affiliation(s)
- Isabella Vlisidou
- School of Cellular and Molecular Medicine, University of Bristol, UK
| | | |
Collapse
|
99
|
Verboon JM, Rahe TK, Rodriguez-Mesa E, Parkhurst SM. Wash functions downstream of Rho1 GTPase in a subset of Drosophila immune cell developmental migrations. Mol Biol Cell 2015; 26:1665-74. [PMID: 25739458 PMCID: PMC4436778 DOI: 10.1091/mbc.e14-08-1266] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 02/25/2015] [Indexed: 01/12/2023] Open
Abstract
Drosophila immune cells undergo four stereotypical developmental migrations to populate the embryo. Wash is a downstream effector of Rho1 and establishes Rho1>Wash>Arp2/3 as the regulatory pathway controlling the cytoskeleton during one of these developmental hemocyte migrations in a WASH regulatory complex–independent manner. Drosophila immune cells, the hemocytes, undergo four stereotypical developmental migrations to populate the embryo, where they provide immune reconnoitering, as well as a number of non–immune-related functions necessary for proper embryogenesis. Here, we describe a role for Rho1 in one of these developmental migrations in which posteriorly located hemocytes migrate toward the head. This migration requires the interaction of Rho1 with its downstream effector Wash, a Wiskott–Aldrich syndrome family protein. Both Wash knockdown and a Rho1 transgene harboring a mutation that prevents Wash binding exhibit the same developmental migratory defect as Rho1 knockdown. Wash activates the Arp2/3 complex, whose activity is needed for this migration, whereas members of the WASH regulatory complex (SWIP, Strumpellin, and CCDC53) are not. Our results suggest a WASH complex–independent signaling pathway to regulate the cytoskeleton during a subset of hemocyte developmental migrations.
Collapse
Affiliation(s)
- Jeffrey M Verboon
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Travis K Rahe
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Evelyn Rodriguez-Mesa
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Susan M Parkhurst
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| |
Collapse
|
100
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|